Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Infect Immun ; 91(4): e0049622, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36912636

ABSTRACT

Among the unfavorable conditions bacteria encounter within the host is restricted access to essential trace metals such as iron. To overcome iron deficiency, bacteria deploy multiple strategies to scavenge iron from host tissues, with abundant examples of iron acquisition systems being implicated in bacterial pathogenesis. Yet the mechanisms utilized by the major nosocomial pathogen Enterococcus faecalis to maintain intracellular iron balance are poorly understood. In this study, we conducted a systematic investigation to identify and characterize the iron acquisition mechanisms of E. faecalis and to determine their contribution to virulence. Bioinformatic analysis and literature surveys revealed that E. faecalis possesses three conserved iron uptake systems. Through transcriptomics, we discovered two novel ABC-type transporters that mediate iron uptake. While inactivation of a single transporter had minimal impact on the ability of E. faecalis to maintain iron homeostasis, inactivation of all five systems (Δ5Fe strain) disrupted intracellular iron homeostasis and considerably impaired cell growth under iron deficiency. Virulence of the Δ5Fe strain was generally impaired in different animal models but showed niche-specific variations in mouse models, leading us to suspect that heme can serve as an iron source to E. faecalis during mammalian infections. Indeed, heme supplementation restored growth of Δ5Fe under iron depletion and virulence in an invertebrate infection model. This study revealed that the collective contribution of five iron transporters promotes E. faecalis virulence and that the ability to acquire and utilize heme as an iron source is critical to the systemic dissemination of E. faecalis.


Subject(s)
ATP-Binding Cassette Transporters , Bacterial Proteins , Biological Transport , Enterococcus faecalis , Iron , Enterococcus faecalis/metabolism , Enterococcus faecalis/pathogenicity , Virulence , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Iron/metabolism , Gene Expression Regulation, Bacterial , Bacterial Proteins/metabolism , Heme/metabolism , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/microbiology , Humans
2.
mBio ; 14(1): e0307322, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36629455

ABSTRACT

The bacterial cell membrane is an interface for cell envelope synthesis, protein secretion, virulence factor assembly, and a target for host cationic antimicrobial peptides (CAMPs). To resist CAMP killing, several Gram-positive pathogens encode the multiple peptide resistance factor (MprF) enzyme that covalently attaches cationic amino acids to anionic phospholipids in the cell membrane. While E. faecalis encodes two mprF paralogs, MprF2 plays a dominant role in conferring resistance to killing by the CAMP human ß-defensin 2 (hBD-2) in E. faecalis strain OG1RF. The goal of the current study is to understand the broader lipidomic and functional roles of E. faecalis mprF. We analyzed the lipid profiles of parental wild-type and mprF mutant strains and show that while ΔmprF2 and ΔmprF1 ΔmprF2 mutants completely lacked cationic lysyl-phosphatidylglycerol (L-PG), the ΔmprF1 mutant synthesized ~70% of L-PG compared to the parent. Unexpectedly, we also observed a significant reduction of PG in ΔmprF2 and ΔmprF1 ΔmprF2. In the mprF mutants, particularly ΔmprF1 ΔmprF2, the decrease in L-PG and phosphatidylglycerol (PG) is compensated by an increase in a phosphorus-containing lipid, glycerophospho-diglucosyl-diacylglycerol (GPDGDAG), and D-ala-GPDGDAG. These changes were accompanied by a downregulation of de novo fatty acid biosynthesis and an accumulation of long-chain acyl-acyl carrier proteins (long-chain acyl-ACPs), suggesting that the suppression of fatty acid biosynthesis was mediated by the transcriptional repressor FabT. Growth in chemically defined media lacking fatty acids revealed severe growth defects in the ΔmprF1 ΔmprF2 mutant strain, but not the single mutants, which was partially rescued through supplementation with palmitic and stearic acids. Changes in lipid homeostasis correlated with lower membrane fluidity, impaired protein secretion, and increased biofilm formation in both ΔmprF2 and ΔmprF1 ΔmprF2, compared to the wild type and ΔmprF1. Collectively, our findings reveal a previously unappreciated role for mprF in global lipid regulation and cellular physiology, which could facilitate the development of novel therapeutics targeting MprF. IMPORTANCE The cell membrane plays a pivotal role in protecting bacteria against external threats, such as antibiotics. Cationic phospholipids such as lysyl-phosphatidyglycerol (L-PG) resist the action of cationic antimicrobial peptides through electrostatic repulsion. Here we demonstrate that L-PG depletion has several unexpected consequences in Enterococcus faecalis, including a reduction of phosphatidylglycerol (PG), enrichment of a phosphorus-containing lipid, reduced fatty acid synthesis accompanied by an accumulation of long-chain acyl-acyl carrier proteins (long chain acyl-ACPs), lower membrane fluidity, and impaired secretion. These changes are not deleterious to the organism as long as exogenous fatty acids are available for uptake from the culture medium. Our findings suggest an adaptive mechanism involving compensatory changes across the entire lipidome upon removal of a single phospholipid modification. Such adaptations must be considered when devising antimicrobial strategies that target membrane lipids.


Subject(s)
Anti-Bacterial Agents , Anti-Infective Agents , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Enterococcus faecalis/metabolism , Drug Resistance, Bacterial , Phospholipids/metabolism , Anti-Infective Agents/metabolism , Fatty Acids/metabolism , Phosphatidylglycerols/metabolism , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/metabolism , Cations/metabolism , Carrier Proteins/metabolism , Bacterial Proteins/metabolism
3.
J Photochem Photobiol B ; 234: 112547, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36030693

ABSTRACT

Knowledge of photo-oxidative stress responses in bacteria that survive antimicrobial photodynamic therapy (aPDT) is scarce. Whereas aPDT is attracting growing clinical interest, subsequent stress responses are crucial to evaluate as they may lead to the up-regulation of pathogenic traits. Here, we aimed to assess transcriptional responses to sublethal aPDT-stress and identify potential connections with virulence-related genes. Six Enterococcus faecalis strains were investigated; ATCC 29212, three dental root-canal isolates labelled UmID1, UmID2 and UmID3 and two vancomycin-resistant isolates labelled A1 and A2. TMPyP was employed as a photosensitiser. A viability dose-response curve to increasing concentrations of TMPyP was determined by culture plating. Differential expression of genes involved in oxidative stress responses (dps and hypR), general stress responses (dnaK, sigma-factorV and relA), virulence-related genes (ace, fsrC and gelE) and vancomycin-resistance (vanA) was assessed by reverse-transcription qPCR. TMPyP-mediated aPDT inactivated all strains with comparable efficiencies. TMPyP at 0.015 µM was selected to induce sublethal photo-oxidative stress. Despite heterogeneities in gene expression between strains, transcriptional profiles revealed up-regulations of transcripts dps, hypR as well as dnaK and sigma factorV after exposure to TMPyP alone and to light-irradiated TMPyP. Specifically, the alternative sigma factorV reached up to 39 ± 113-fold (median ± IQR) (p = 0.0369) in strain A2. Up-regulation of the quorum sensing operon, fsr, and its downstream virulence-related gelatinase gelE were also observed in strains ATCC-29212, A1, A2 and UmID3. Finally, photo-oxidative stress induced vanA-type vancomycin-resistance gene in both carrier isolates, reaching up to 3.3 ± 17-fold in strain A2 (p = 0.015). These findings indicate that, while aPDT successfully inactivates vancomycin-resistant and naïve strains of E. faecalis, subpopulations of surviving cells respond by co-ordinately up-regulating a network of genes involved in stress survival and virulence. This includes the induction of vancomycin-resistance genes in carrier isolates. These data may provide the mechanistic basis to circumvent bacterial responses and improve future clinical protocols.


Subject(s)
Enterococcus faecalis , Oxidative Stress , Photochemotherapy , Vancomycin , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterococcus faecalis/metabolism , Enterococcus faecalis/pathogenicity , Microbial Sensitivity Tests , Oxidative Stress/physiology , Sigma Factor/metabolism , Vancomycin/pharmacology , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
4.
Microbiol Spectr ; 10(1): e0099121, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019708

ABSTRACT

With the increasing reports of community-acquired and nosocomial infection caused by multidrug-resistant Gram-positive pathogens, there is an urgent need to develop new antimicrobial agents with novel antibacterial mechanisms. Here, we investigated the antibacterial activity of the natural product ginkgolic acid (GA) (15:1), derived from Ginkgo biloba, and its potential mode of action against the Gram-positive bacteria Enterococcus faecalis and Staphylococcus aureus. The MIC values of GA (15:1) against clinical E. faecalis and S. aureus isolates from China were ≤4 and ≤8 µg/mL, respectively, from our test results. Moreover, GA (15:1) displayed high efficiency in biofilm formation inhibition and bactericidal activity against E. faecalis and S. aureus. During its inhibition of the planktonic bacteria, the antibacterial activity of GA (15:1) was significantly improved under the condition of abolishing iron homeostasis. When iron homeostasis was abolished, inhibition of planktonic bacteria by GA (15:1) was significantly improved. This phenomenon can be interpreted as showing that iron homeostasis disruption facilitated the disruption of the functions of ribosome and protein synthesis by GA (15:1), resulting in inhibition of bacterial growth and cell death. Genetic mutation of ferric uptake regulator (Fur) led to GA (15:1) tolerance in in vitro-induced resistant derivatives, while overexpression of Fur led to increased GA (15:1) susceptibility. Additionally, GA (15:1) significantly decreased the bacterial loads of S. aureus strain USA300 in the lung tissues of mice in a pneumonic murine model. Conclusively, this study revealed an antimicrobial mechanism of GA (15:1) involving cross talk with iron homeostasis against Gram-positive pathogens. In the future, the natural product GA (15:1) might be applied to combat infections caused by Gram-positive pathogens. IMPORTANCE The increasing emergence of infectious diseases associated with multidrug-resistant Gram-positive pathogens has raised the urgent need to develop novel antibiotics. GA (15:1) is a natural product derived from Ginkgo biloba and possesses a wide range of bioactivities, including antimicrobial activity. However, its antibacterial mechanisms remain unclear. Our current study found that the function of ferric uptake regulator (Fur) was highly correlated with the antimicrobial activity of GA (15:1) against E. faecalis and that the antibacterial activity of GA (15:1) could be strengthened by the disruption of iron homeostasis. This study provided important insight into the mode of action of GA (15:1) against Gram-positive bacteria and suggested that GA (15:1) holds the potential to be an antimicrobial treatment option for infection caused by multidrug-resistant Gram-positive pathogens.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Enterococcus faecalis/drug effects , Gram-Positive Bacterial Infections/drug therapy , Iron/metabolism , Plant Extracts/administration & dosage , Salicylates/administration & dosage , Staphylococcus aureus/drug effects , Animals , Enterococcus faecalis/metabolism , Female , Ginkgo biloba , Gram-Positive Bacterial Infections/microbiology , Homeostasis/drug effects , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Staphylococcus aureus/metabolism
5.
Nature ; 595(7867): 409-414, 2021 07.
Article in English | MEDLINE | ID: mdl-34194038

ABSTRACT

Social interactions among animals mediate essential behaviours, including mating, nurturing, and defence1,2. The gut microbiota contribute to social activity in mice3,4, but the gut-brain connections that regulate this complex behaviour and its underlying neural basis are unclear5,6. Here we show that the microbiome modulates neuronal activity in specific brain regions of male mice to regulate canonical stress responses and social behaviours. Social deviation in germ-free and antibiotic-treated mice is associated with elevated levels of the stress hormone corticosterone, which is primarily produced by activation of the hypothalamus-pituitary-adrenal (HPA) axis. Adrenalectomy, antagonism of glucocorticoid receptors, or pharmacological inhibition of corticosterone synthesis effectively corrects social deficits following microbiome depletion. Genetic ablation of glucocorticoid receptors in specific brain regions or chemogenetic inactivation of neurons in the paraventricular nucleus of the hypothalamus that produce corticotrophin-releasing hormone (CRH) reverse social impairments in antibiotic-treated mice. Conversely, specific activation of CRH-expressing neurons in the paraventricular nucleus induces social deficits in mice with a normal microbiome. Via microbiome profiling and in vivo selection, we identify a bacterial species, Enterococcus faecalis, that promotes social activity and reduces corticosterone levels in mice following social stress. These studies suggest that specific gut bacteria can restrain the activation of the HPA axis, and show that the microbiome can affect social behaviours through discrete neuronal circuits that mediate stress responses in the brain.


Subject(s)
Brain/cytology , Brain/physiology , Gastrointestinal Microbiome/physiology , Neurons/metabolism , Social Behavior , Stress, Psychological , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Enterococcus faecalis/metabolism , Germ-Free Life , Glucocorticoids/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/metabolism , Signal Transduction
6.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946710

ABSTRACT

Considering the advent of antibiotic resistance, the study of bacterial metabolic behavior stimulated by novel antimicrobial agents becomes a relevant tool to elucidate involved adaptive pathways. Profiling of volatile metabolites was performed to monitor alterations of bacterial metabolism induced by biosynthesized silver nanoparticles (bio-AgNPs). Escherichia coli, Enterococcus faecalis, Klebsiella pneumoniae and Proteus mirabilis were isolated from pressure ulcers, and their cultures were prepared in the presence/absence of bio-AgNPs at 12.5, 25 and 50 µg mL-1. Headspace solid phase microextraction associated to gas chromatography-mass spectrometry was the employed analytical platform. At the lower concentration level, the agent promoted positive modulation of products of fermentation routes and bioactive volatiles, indicating an attempt of bacteria to adapt to an ongoing suppression of cellular respiration. Augmented response of aldehydes and other possible products of lipid oxidative cleavage was noticed for increasing levels of bio-AgNPs. The greatest concentration of agent caused a reduction of 44 to 80% in the variety of compounds found in the control samples. Pathway analysis indicated overall inhibition of amino acids and fatty acids routes. The present assessment may provide a deeper understanding of molecular mechanisms of bio-AgNPs and how the metabolic response of bacteria is untangled.


Subject(s)
Bacteria/drug effects , Bacteria/metabolism , Metal Nanoparticles/therapeutic use , Pressure Ulcer/drug therapy , Pressure Ulcer/microbiology , Silver/therapeutic use , Volatile Organic Compounds/metabolism , Anti-Bacterial Agents/therapeutic use , Bacteria/isolation & purification , Drug Resistance, Bacterial , Enterococcus faecalis/drug effects , Enterococcus faecalis/isolation & purification , Enterococcus faecalis/metabolism , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Humans , In Vitro Techniques , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/metabolism , Metabolic Networks and Pathways/drug effects , Metabolome/drug effects , Metabolomics , Microbial Sensitivity Tests , Proteus mirabilis/drug effects , Proteus mirabilis/isolation & purification , Proteus mirabilis/metabolism , Volatile Organic Compounds/chemistry , Volatile Organic Compounds/classification
7.
Gut ; 69(7): 1239-1247, 2020 07.
Article in English | MEDLINE | ID: mdl-31744910

ABSTRACT

OBJECTIVE: Dietary fibre has beneficial effects on energy metabolism, and the majority of studies have focused on short-chain fatty acids produced by gut microbiota. Ginseng has been reported to aid in body weight management, however, its mechanism of action is not yet clear. In this study, we focused on the potential modulating effect of ginseng on gut microbiota, aiming to identify specific strains and their metabolites, especially long-chain fatty acids (LCFA), which mediate the anti-obesity effects of ginseng. DESIGN: Db/db mice were gavaged with ginseng extract (GE) and the effects of GE on gut microbiota were evaluated using 16S rDNA-based high throughput sequencing. To confirm the candidate fatty acids, untargeted metabolomics analyses of the serum and medium samples were performed. RESULTS: We demonstrated that GE can induce Enterococcus faecalis, which can produce an unsaturated LCFA, myristoleic acid (MA). Our results indicate that E. faecalis and its metabolite MA can reduce adiposity by brown adipose tissue (BAT) activation and beige fat formation. In addition, the gene of E. faecalis encoding Acyl-CoA thioesterases (ACOTs) exhibited the biosynthetic potential to synthesise MA, as knockdown (KD) of the ACOT gene by CRISPR-dCas9 significantly reduced MA production. Furthermore, exogenous treatment with KD E. faecalis could not reproduce the beneficial effects of wild type E. faecalis, which work by augmenting the circulating MA levels. CONCLUSIONS: Our results demonstrated that the gut microbiota-LCFA-BAT axis plays an important role in host metabolism, which may provide a strategic advantage for the next generation of anti-obesity drug development.


Subject(s)
Adipose Tissue, Brown/metabolism , Enterococcus faecalis/metabolism , Fatty Acids, Monounsaturated/metabolism , Obesity/metabolism , Animals , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Male , Mice , Mice, Inbred C57BL , Panax , Plant Extracts/pharmacology , RNA, Ribosomal, 16S/genetics
8.
Metabolomics ; 15(4): 53, 2019 03 27.
Article in English | MEDLINE | ID: mdl-30919213

ABSTRACT

INTRODUCTION: Lipidomics can reveal global alterations in a broad class of molecules whose functions are innately linked to physiology. Monitoring changes in the phospholipid composition of biological membranes in response to stressors can aid the development of targeted therapies. However, exact quantitation of cardiolipins is not a straightforward task due to low ionization efficiencies and poor chromatographic separation of these compounds. OBJECTIVE: The aim of this study was to develop a quantitative method for the detection of cardiolipins and other phospholipids using both a targeted and untargeted analyses with a Q-Exactive. METHODS: HILIC chromatography and high-resolution mass spectrometry with parallel reaction monitoring was used to measure changes in lipid concentration. Internal standards and fragmentation techniques allowed for the reliable quantitation of lipid species including: lysyl-phosphatidylglycerol, phosphatidylglycerol, and cardiolipin. RESULTS: The untargeted analysis was capable to detecting 6 different phospholipid classes as well as free fatty acids. The targeted analysis quantified up to 23 cardiolipins, 10 phosphatidylglycerols and 10 lysyl-phosphatidylglycerols with detection limits as low as 50 nM. Biological validation with Enterococcus faecalis demonstrates sensitivity in monitoring the incorporation of exogenously supplied free fats into membrane phospholipids. When supplemented with oleic acid, the amount of free oleic acid in the membrane was 100 times greater and the concentration of polyunsaturated cardiolipin increased to over 3.5 µM compared to controls. CONCLUSIONS: This lipidomics method is capable of targeted quantitation for challenging biologically relevant cardiolipins as well as broad, untargeted lipid profiling.


Subject(s)
Lipidomics/methods , Metabolomics/methods , Tandem Mass Spectrometry/methods , Cardiolipins/analysis , Chromatography, High Pressure Liquid/methods , Enterococcus faecalis/metabolism , Fatty Acids, Nonesterified/analysis , Lysine/analysis , Phosphatidylglycerols/analysis , Phospholipids/analysis
9.
J Dairy Sci ; 101(12): 10626-10635, 2018 12.
Article in English | MEDLINE | ID: mdl-30316597

ABSTRACT

Selenium is an essential micronutrient for living beings, as it helps to maintain the normal physiological functions of the organism. The numerous discoveries involving the importance of this element to the health of human beings have fostered interest in research to develop enriched and functional foods. The present study evaluated the potential for bacterial strains of Enterococcus faecalis (CH121 and CH124), Lactobacillus parabuchneri (ML4), Lactobacillus paracasei (ML13, ML33, CH135, and CH139), and Lactobacillus plantarum (CH131) to bioaccumulate Se in their biomass by adding different concentrations of sodium selenite (30 to 200 mg/L) to the culture medium. Quantification of Se with UV and visible molecular absorption spectroscopy showed that the investigated bacteria were able to bioaccumulate this micromineral into their biomass. Two of the L. paracasei strains (ML13 and CH135) bioaccumulated the highest Se concentrations (38.1 ± 1.7 mg/g and 40.7 ± 1.1 mg/g, respectively) after culture in the presence of 150 mg/L of Se. This bioaccumulation potential has applications in the development of dairy products and may be an alternative Se source in the diets of humans and other animals.


Subject(s)
Enterococcus faecalis/metabolism , Lactobacillus/metabolism , Selenium/metabolism , Animals , Cattle , Culture Media/analysis , Culture Media/metabolism , Dairy Products/microbiology , Enterococcus faecalis/genetics , Enterococcus faecalis/growth & development , Humans , Lactic Acid/metabolism , Lactobacillus/growth & development , Sodium Selenite/analysis , Sodium Selenite/metabolism
10.
Sci Rep ; 8(1): 4212, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523862

ABSTRACT

In the past few years, the World Health Organization has been warning that the post-antibiotic era is an increasingly real threat. The rising and disseminated resistance to antibiotics made mandatory the search for new drugs and/or alternative therapies that are able to eliminate resistant microorganisms and impair the development of new forms of resistance. In this context, antimicrobial photodynamic therapy (aPDT) and helical cationic antimicrobial peptides (AMP) are highlighted for the treatment of localized infections. This study aimed to combine the AMP aurein 1.2 to aPDT using Enterococcus faecalis as a model strain. Our results demonstrate that the combination of aPDT with aurein 1.2 proved to be a feasible alternative capable of completely eliminating E. faecalis employing low concentrations of both PS and AMP, in comparison with the individual therapies. Aurein 1.2 is capable of enhancing the aPDT activity whenever mediated by methylene blue or chlorin-e6, but not by curcumin, revealing a PS-dependent mechanism. The combined treatment was also effective against different strains; noteworthy, it completely eliminated a vancomycin-resistant strain of Enterococcus faecium. Our results suggest that this combined protocol must be exploited for clinical applications in localized infections as an alternative to antibiotics.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Antimicrobial Cationic Peptides/metabolism , Biological Transport , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/radiation effects , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/radiation effects , Drug Synergism , Enterococcus faecalis/cytology , Enterococcus faecalis/drug effects , Enterococcus faecalis/metabolism , Enterococcus faecalis/radiation effects , Humans , Reactive Oxygen Species/metabolism
11.
Sci Rep ; 7(1): 3747, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623336

ABSTRACT

The opportunistic fish pathogen, Enterococcus faecalis has been reported to cause mass mortality in several fish species in different countries. The objectives of this study were to (i) identify E. faecalis from the diseased fishes through molecular techniques; (ii) assess the antibiotic susceptibility profile of E. faecalis isolates; and (iii) control disease in tilapia fish by treatment with medicinal plant extracts. A total of 48 isolates were phenotypically identified as Enterococcus species from tilapia, stinging catfish and walking catfish cultivated in several fish farms in Gazipur. Ten randomly selected isolates were identified as E. faecalis by 16S rRNA gene sequencing. Artificial infection revealed that most of the isolates caused moderate to high mortality in fishes with characteristic disease symptoms. These isolates exhibited resistance to multiple antibiotics in vitro. Bioassay revealed that organic extracts of Tamarindus indica and Emblica officinalis leaves, Allium sativum bulb, and Syzygium aromaticum bud inhibited the growth of E. faecalis. Methanol extracts of A. sativum and methanol and acetone extracts of S. aromaticum significantly reduced the mortality of fish artificially infected with E. faecalis as both preventive and therapeutic agents. This is the first report on molecular identification, and herbal control of fish pathogenic E. faecalis in Bangladesh.


Subject(s)
Catfishes/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Enterococcus faecalis , Fish Diseases , Gram-Positive Bacterial Infections , Plant Extracts/pharmacology , Tilapia/microbiology , Animals , Drug Resistance, Multiple, Bacterial/drug effects , Enterococcus faecalis/genetics , Enterococcus faecalis/isolation & purification , Enterococcus faecalis/metabolism , Fish Diseases/drug therapy , Fish Diseases/genetics , Fish Diseases/microbiology , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/veterinary , Plant Extracts/chemistry
12.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28483855

ABSTRACT

In Enterococcus faecalis, the regulatory nucleotides pppGpp and ppGpp, collectively, (p)ppGpp, are required for growth in blood, survival within macrophages, and virulence. However, a clear understanding of how (p)ppGpp promotes virulence in E. faecalis and other bacterial pathogens is still lacking. In the host, the essential transition metals iron (Fe) and manganese (Mn) are not readily available to invading pathogens because of a host-driven process called nutritional immunity. Considering its central role in adaptation to nutritional stresses, we hypothesized that (p)ppGpp mediates E. faecalis virulence through regulation of metal homeostasis. Indeed, supplementation of serum with either Fe or Mn restored growth and survival of the Δrel ΔrelQ [(p)ppGpp0] strain to wild-type levels. Using a chemically defined medium, we found that (p)ppGpp accumulates in response to either Fe depletion or Mn depletion and that the (p)ppGpp0 strain has a strong growth requirement for Mn that is alleviated by Fe supplementation. Although inactivation of the nutrient-sensing regulator codY restored some phenotypes of the (p)ppGpp0 strain, transcriptional analysis showed that the (p)ppGpp/CodY network does not promote transcription of known metal transporters. Interestingly, physiologic and enzymatic investigations suggest that the (p)ppGpp0 strain requires higher levels of Mn in order to cope with high levels of endogenously produced reactive oxygen species (ROS). Because (p)ppGpp mediates antibiotic persistence and virulence in several bacteria, our findings have broad implications and provide new leads for the development of novel therapeutic and preventive strategies against E. faecalis and beyond.


Subject(s)
Enterococcus faecalis/growth & development , Enterococcus faecalis/metabolism , Gene Expression Regulation, Bacterial , Guanosine Pentaphosphate/metabolism , Homeostasis , Iron/metabolism , Manganese/metabolism , Microbial Viability , Virulence
13.
J Trace Elem Med Biol ; 39: 135-139, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27908405

ABSTRACT

Microorganisms are capable of synthesizing metal nanoparticles, and specifically Enterococcus faecalis bacteria were tested for its ability to synthesize selenium nanoparticles (Se-NPs) from sodium selenite. The biosynthesized Se-NPs were spherical in shape with the size range of 29-195nm. Also, the TEM microscopy showed the accumulation of nano-structures as extracellular deposits. The ability of the bacteria to tolerate high levels of toxic selenite was studied by changing with different concentrations of sodium selenite (0.19mM-2.97mM). Also, the effect of Se-NPs was studied on the growth profile of number of pathogenic Gram-positive and -negative bacteria. High concentrations of sodium selenite in the medium led to the production of small amounts of selenium nanostructures by bacteria. In addition, Se-NPs can be used as an anti-staphylococcal element to effectively prevent and treat S. aureus infections.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Enterococcus faecalis/metabolism , Nanoparticles/metabolism , Selenium/metabolism , Selenium/pharmacology , Staphylococcus aureus/drug effects , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Particle Size , Structure-Activity Relationship , Surface Properties
14.
Food Funct ; 6(6): 1875-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25943486

ABSTRACT

The methanolic extract of the wild edible mushroom Cantharellus cibarius Fr. (chanterelle) was analyzed for in vitro antioxidative, cytotoxic, antihypertensive and antibacterial activities. Various primary and secondary metabolites were found. Phenols were the major antioxidant components found in the extract (49.8 mg g(-1)), followed by flavonoids, whose content was approximately 86% of the total phenol content. Antioxidant activity, measured by four different methods, was high for inhibition of lipid peroxidation (EC50 = 1.21 mg mL(-1)) and chelating ability (EC50 = 0.64 mg mL(-1)). The antioxidant activity of the C. cibarius methanol extract was achieved through chelating iron compared to hydrogen atom and/or electron transfer. The extract showed good selectivity in cytotoxicity on human cervix adenocarcinoma HeLa, breast carcinoma MDA-MB-453 and human myelogenous leukemia K562, compared to normal control human fetal lung fibroblasts MRC-5 and human lung bronchial epithelial cells BEAS-2B. The extract had inhibitory activity against angiotensin converting I enzyme (ACE) (IC50 = 0.063 mg mL(-1)). The extract revealed selective antimicrobial activity against Gram-positive bacteria with the highest potential against E. faecalis. The medicinal and health benefits, observed in wild C. cibarius mushroom, seem an additional reason for its traditional use as a popular delicacy food.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Basidiomycota/chemistry , Biological Products/chemistry , Dietary Supplements , Phytochemicals/chemistry , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/isolation & purification , Angiotensin-Converting Enzyme Inhibitors/metabolism , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Antioxidants/adverse effects , Antioxidants/isolation & purification , Antioxidants/metabolism , Basidiomycota/growth & development , Basidiomycota/metabolism , Biological Products/adverse effects , Biological Products/isolation & purification , Biological Products/metabolism , Cell Line , Cell Line, Tumor , Cell Survival , Dietary Supplements/adverse effects , Dietary Supplements/analysis , Enterococcus faecalis/growth & development , Enterococcus faecalis/metabolism , Flavonoids/adverse effects , Flavonoids/analysis , Flavonoids/chemistry , Flavonoids/metabolism , Forests , Fruiting Bodies, Fungal/chemistry , Fruiting Bodies, Fungal/growth & development , Fruiting Bodies, Fungal/metabolism , Humans , Iron Chelating Agents/adverse effects , Iron Chelating Agents/chemistry , Iron Chelating Agents/isolation & purification , Iron Chelating Agents/metabolism , Lipid Peroxidation , Methanol/chemistry , Montenegro , Phenols/adverse effects , Phenols/analysis , Phenols/chemistry , Phenols/metabolism , Phytochemicals/adverse effects , Phytochemicals/analysis , Phytochemicals/biosynthesis , Solvents/chemistry
15.
Metallomics ; 7(7): 1137-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25906431

ABSTRACT

By integrating the microarray expression data and a global E. faecalis transcriptional network we identified a sub-network activated by zinc and copper. Our analyses indicated that the transcriptional response of the bacterium to copper and zinc exposure involved the activation of two modules, module I that contains genes implicated in zinc homeostasis, including the Zur transcriptional repressor, and module II containing a set of genes associated with general stress response and basal metabolism. Bacterial exposure to zinc and copper led to the repression of the zinc uptake systems of module I. Upon deletion of Zur, exposure to different zinc and copper conditions induced complementary homeostatic mechanisms (ATPase efflux proteins) to control the intracellular concentrations of zinc. The transcriptional activation of zinc homeostasis genes by zinc and copper reveals a functional interplay between these two metals, in which exposure to copper also impacts on the zinc homeostasis. Finally, we present a new zinc homeostasis model in E. faecalis, positioning this bacterium as one of the most complete systems biology model in metals described to date.


Subject(s)
Bacterial Proteins/genetics , Copper/metabolism , Enterococcus faecalis/genetics , Gene Expression Regulation, Bacterial , Repressor Proteins/genetics , Zinc/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Enterococcus faecalis/chemistry , Enterococcus faecalis/metabolism , Gram-Positive Bacterial Infections/microbiology , Homeostasis , Humans , Models, Molecular , Molecular Sequence Data , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Sequence Alignment
16.
PLoS Pathog ; 10(6): e1004210, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967632

ABSTRACT

The ability of innate immune cells to sense and respond to impending danger varies by anatomical location. The liver is considered tolerogenic but is still capable of mounting a successful immune response to clear various infections. To understand whether hepatic immune cells tune their response to different infectious challenges, we probed mononuclear cells purified from human healthy and diseased livers with distinct pathogen-associated molecules. We discovered that only the TLR8 agonist ssRNA40 selectively activated liver-resident innate immune cells to produce substantial quantities of IFN-γ. We identified CD161(Bright) mucosal-associated invariant T (MAIT) and CD56(Bright) NK cells as the responding liver-resident innate immune cells. Their activation was not directly induced by the TLR8 agonist but was dependent on IL-12 and IL-18 production by ssRNA40-activated intrahepatic monocytes. Importantly, the ssRNA40-induced cytokine-dependent activation of MAIT cells mirrored responses induced by bacteria, i.e., generating a selective production of high levels of IFN-γ, without the concomitant production of TNF-α or IL-17A. The intrahepatic IFN-γ production could be detected not only in healthy livers, but also in HBV- or HCV-infected livers. In conclusion, the human liver harbors a network of immune cells able to modulate their immunological responses to different pathogen-associated molecules. Their ability to generate a strong production of IFN-γ upon stimulation with TLR8 agonist opens new therapeutic opportunities for the treatment of diverse liver pathologies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Innate/drug effects , Leukocytes, Mononuclear/drug effects , Liver/drug effects , Oligoribonucleotides/pharmacology , Toll-Like Receptor 8/agonists , Up-Regulation/drug effects , Cells, Cultured , Coculture Techniques , Enterococcus faecalis/immunology , Enterococcus faecalis/metabolism , Enterococcus faecalis/pathogenicity , Escherichia coli/immunology , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Hepacivirus/immunology , Hepacivirus/pathogenicity , Hepatitis B/immunology , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B virus/immunology , Hepatitis B virus/pathogenicity , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatitis C/virology , Humans , Interferon-gamma Release Tests , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Liver/immunology , Liver/microbiology , Liver/pathology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Riboflavin/biosynthesis , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptor 8/metabolism
17.
J Bacteriol ; 196(1): 170-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24163335

ABSTRACT

Enoyl-acyl carrier protein (ACP) reductase catalyzes the last step of the bacterial fatty acid elongation cycle. Enterococcus faecalis is unusual in that it encodes two unrelated enoyl-ACP reductases, FabI and FabK. We recently reported that deletion of the gene encoding FabI results in an unsaturated fatty acid (UFA) auxotroph despite the presence of fabK, a gene encoding a second fully functional enoyl-ACP reductase. By process of elimination, our prior report argued that poor expression was the reason that fabK failed to functionally replace FabI. We now report that FabK is indeed poorly expressed and that the expression defect is at the level of translation rather than transcription. We isolated four spontaneous mutants that allowed growth of the E. faecalis ΔfabI strain on fatty acid-free medium. Each mutational lesion (single base substitution or deletion) extended the fabK ribosome binding site. Inactivation of fabK blocked growth, indicating that the mutations acted only on fabK rather than a downstream gene. The mutations activated fabK translation to levels that supported fatty acid synthesis and hence cell growth. Furthermore, site-directed and random mutagenesis experiments showed that point mutations that resulted in increased complementarity to the 3' end of the 16S rRNA increased FabK translation to levels sufficient to support growth, whereas mutations that decreased complementarity blocked fabK translation.


Subject(s)
Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Gene Expression , Protein Biosynthesis , Culture Media/chemistry , DNA Mutational Analysis , DNA, Complementary , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enterococcus faecalis/growth & development , Enterococcus faecalis/metabolism , Fatty Acids/metabolism , Gene Deletion , Mutant Proteins/genetics , Mutant Proteins/metabolism , RNA, Ribosomal, 16S/genetics , Ribosomes/metabolism
18.
Environ Sci Pollut Res Int ; 20(7): 5056-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23334551

ABSTRACT

Reactive dyes account for one of the major sources of dye wastes in textile effluent. In this study, decolorization of the monoazo dye, Acid Orange 7 (AO7) by the Enterococcus faecalis strain ZL that isolated from a palm oil mill effluent treatment plant has been investigated. Decolorization efficiency of azo dye is greatly affected by the types of nutrients and the size of inoculum used. In this work, one-factor-at-a-time (method and response surface methodology (RSM) was applied to optimize these operational factors and also to study the combined interaction between them. Analysis of AO7 decolorization was done using Fourier transform infrared (FTIR) spectroscopy, desorption study, UV-Vis spectral analysis, field emission scanning electron microscopy (FESEM), and high performance liquid chromatography (HPLC). The optimum condition via RSM for the color removal of AO7 was found to be as follows: yeast extract, 0.1% w/v, glycerol concentration of 0.1% v/v, and inoculum density of 2.5% v/v at initial dye concentration of 100 mg/L at 37 °C. Decolorization efficiency of 98% was achieved in only 5 h. The kinetic of AO7 decolorization was found to be first order with respect to dye concentration with a k value of 0.87/h. FTIR, desorption study, UV-Vis spectral analysis, FESEM, and HPLC findings indicated that the decolorization of AO7 was mainly due to the biosorption as well as biodegradation of the bacterial cells. In addition, HPLC analyses also showed the formation of sulfanilic acid as a possible degradation product of AO7 under facultative anaerobic condition. This study explored the ability of E. faecalis strain ZL in decolorizing AO7 by biosorption as well as biodegradation process.


Subject(s)
Azo Compounds/metabolism , Benzenesulfonates/metabolism , Coloring Agents/metabolism , Enterococcus faecalis/metabolism , Azo Compounds/analysis , Benzenesulfonates/analysis , Biodegradation, Environmental , Chromatography, High Pressure Liquid , Coloring Agents/analysis , Environmental Monitoring , Industrial Waste/analysis , Kinetics , Microscopy, Electron, Scanning , Palm Oil , Plant Oils/analysis , Plant Oils/chemistry , Reproducibility of Results , Spectroscopy, Fourier Transform Infrared , Sulfanilic Acids/analysis , Sulfanilic Acids/metabolism , Textiles , Water Purification/methods
19.
World J Microbiol Biotechnol ; 29(1): 137-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22961443

ABSTRACT

Bacterial resistance to antibiotics has become a serious problem of public health. Along with the controlled permeability by the cell-wall, active efflux systems can provide resistance by extruding antibiotics. Carnosic acid is capable to potentiate the antimicrobial activity of several antibiotics. However, the underlying molecular mechanism governing this effect remains unclear. The present study aims to investigate the effect of carnosic acid on the transport of ethidium bromide, on the permeability or the membrane potential in Enterococcus faecalis and Staphylococcus aureus. By using fluorimetric assays it was demonstrated that in E. faecalis, carnosic acid is a modulator of the uptake and efflux of ethidium bromide which does not induce cell membrane permeabilization phenomena. Such effect was sensitive to the inhibition caused by both the proton-motive force carbonyl cyanide m-chlorophenylhydrazone and the calcium antagonist verapamil, but not to vanadate, an ATPase inhibitor. In this work it was demonstrated, for the first time, that the activity of carnosic acid on the uptake/efflux of ethidium bromide is correlated with its capacity to change the membrane potential gradient in S. aureus and E. faecalis. In conclusion, carnosic acid is a natural compound, structurally unrelated to known antibiotics, which can function as an efflux pump modulator by dissipation of the membrane potential. Therefore, carnosic acid would be a good candidate to be employed as a novel therapeutic agent to be used in combination therapies against drug-resistant enterococci and S. aureus infections.


Subject(s)
Abietanes/pharmacology , Enterococcus faecalis/drug effects , Plant Extracts/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Resistance, Multiple, Bacterial , Enterococcus faecalis/metabolism , Ethidium/metabolism , Ethidium/pharmacokinetics , Membrane Potentials/drug effects , Permeability/drug effects , Proton-Motive Force/drug effects , Staphylococcus aureus/metabolism
20.
Mol Microbiol ; 78(6): 1393-402, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21143313

ABSTRACT

The S(MK) (SAM-III) box is an S-adenosylmethionine (SAM)-responsive riboswitch found in the 5' untranslated region of metK genes, encoding SAM synthetase, in many members of the Lactobacillales. SAM binding causes a structural rearrangement in the RNA that sequesters the Shine-Dalgarno (SD) sequence by pairing with a complementary anti-SD (ASD) sequence; sequestration of the SD sequence inhibits binding of the 30S ribosomal subunit and prevents translation initiation. We observed a slight increase in the half-life of the metK transcript in vivo when Enterococcus faecalis cells were depleted for SAM, but no significant change in overall transcript abundance, consistent with the model that this riboswitch regulates at the level of translation initiation. The half-life of the SAM-S(MK) box RNA complex in vitro is shorter than that of the metK transcript in vivo, raising the possibility of reversible binding of SAM. We used a fluorescence assay to directly visualize reversible switching between the SAM-free and SAM-bound conformations. We propose that the S(MK) box riboswitch can make multiple SAM-dependent regulatory decisions during the lifetime of the transcript in vivo, acting as a reversible switch that allows the cell to respond rapidly to fluctuations in SAM pools by modulating expression of the SAM synthetase gene.


Subject(s)
Bacterial Proteins/genetics , Enterococcus faecalis/enzymology , Gene Expression Regulation, Enzymologic , Methionine Adenosyltransferase/genetics , Response Elements , Riboswitch , 5' Untranslated Regions , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Enterococcus faecalis/chemistry , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Gene Expression Regulation, Bacterial , Methionine Adenosyltransferase/chemistry , Methionine Adenosyltransferase/metabolism , Nucleic Acid Conformation , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , S-Adenosylmethionine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL