Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.420
Filter
1.
World J Microbiol Biotechnol ; 40(9): 285, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073503

ABSTRACT

The accelerated spread of antimicrobial-resistant bacteria has caused a serious health problem and rendered antimicrobial treatments ineffective. Innovative approaches are crucial to overcome the health threat posed by resistant pathogens and prevent the emergence of untreatable infections. Triggering stress responses in bacteria can diminish susceptibility to various antimicrobials by inducing resistance mechanisms. Therefore, a thorough understanding of stress response control, especially in relation to antimicrobial resistance, offers valuable perspectives for innovative and efficient therapeutic approaches to combat antimicrobial resistance. The aim of this study was to evaluate the stress responses of 8 different bacteria by analyzing reporter metabolites, around which significant alterations were observed, using a pathway-driven computational approach. For this purpose, the transcriptomic data that the bacterial pathogens were grown under 11 different stress conditions mimicking the human host environments were integrated with the genome-scale metabolic models of 8 pathogenic species (Enterococcus faecalis OG1R, Escherichia coli EPEC O127:H6 E2348/69, Escherichia coli ETEC H10407, Escherichia coli UPEC 536, Klebsiella pneumoniae MGH 78578, Pseudomonas aeruginosa PAO1, Staphylococcus aureus MRSA252, and Staphylococcus aureus MSSA476). The resulting reporter metabolites were enriched in multiple metabolic pathways, with cofactor biosynthesis being the most important. The results of this study will serve as a guide for the development of antimicrobial agents as they provide a first insight into potential drug targets.


Subject(s)
Anti-Bacterial Agents , Bacteria , Stress, Physiological , Bacteria/drug effects , Bacteria/genetics , Bacteria/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Metabolic Networks and Pathways , Humans , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Transcriptome , Escherichia coli/drug effects , Escherichia coli/genetics , Microbial Sensitivity Tests , Gene Expression Profiling
2.
Nat Commun ; 15(1): 5625, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987237

ABSTRACT

Competence for natural transformation is a central driver of genetic diversity in bacteria. In the human pathogen Streptococcus pneumoniae, competence exhibits a populational character mediated by the stress-induced ComABCDE quorum-sensing (QS) system. Here, we explore how this cell-to-cell communication mechanism proceeds and the functional properties acquired by competent cells grown under lethal stress. We show that populational competence development depends on self-induced cells stochastically emerging in response to stresses, including antibiotics. Competence then propagates through the population from a low threshold density of self-induced cells, defining a biphasic Self-Induction and Propagation (SI&P) QS mechanism. We also reveal that a competent population displays either increased sensitivity or improved tolerance to lethal doses of antibiotics, dependent in the latter case on the competence-induced ComM division inhibitor. Remarkably, these surviving competent cells also display an altered transformation potential. Thus, the unveiled SI&P QS mechanism shapes pneumococcal competence as a health sensor of the clonal population, promoting a bet-hedging strategy that both responds to and drives cells towards heterogeneity.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Quorum Sensing , Streptococcus pneumoniae , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/physiology , Anti-Bacterial Agents/pharmacology , Quorum Sensing/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Gene Expression Regulation, Bacterial/drug effects , Transformation, Bacterial
3.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38970380

ABSTRACT

Alternative strategies for controlling Staphylococcus aureus and other pathogens have been continuously investigated, with nisin, a bacteriocin widely used in the food industry as a biopreservative, gaining increasing attention. In addition to its antimicrobial properties, bacteriocins have significant effects on genome functionality even at inhibitory concentrations. This study investigated the impact of subinhibitory concentrations of nisin on S. aureus. Culturing in the presence of 0.625 µmol l-1 nisin, led to the increased relative expression of hla, saeR, and sarA, genes associated with virulence while expression of the sea gene, encoding staphylococcal enterotoxin A (SEA), decreased. In an in vivo experiment, Galleria mellonella larvae inoculated with S. aureus cultured in the presence of nisin exhibited 97% mortality at 72 h post-infection, compared to over 40% of larvae mortality in larvae infected with S. aureus. A comprehensive understanding of the effect of nisin on the transcriptional response of virulence genes and the impact of these changes on the virulence of S. aureus can contribute to assessing the application of this bacteriocin in food and medical contexts.


Subject(s)
Anti-Bacterial Agents , Larva , Moths , Nisin , Staphylococcus aureus , Nisin/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Animals , Virulence/genetics , Larva/microbiology , Larva/drug effects , Anti-Bacterial Agents/pharmacology , Moths/microbiology , Staphylococcal Infections/microbiology , Gene Expression Regulation, Bacterial/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence Factors/genetics , Microbial Sensitivity Tests
4.
Nat Commun ; 15(1): 5683, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971825

ABSTRACT

Bacteria within mature biofilms are highly resistant to antibiotics than planktonic cells. Oxygen limitation contributes to antibiotic resistance in mature biofilms. Nitric oxide (NO) induces biofilm dispersal; however, low NO levels stimulate biofilm formation, an underexplored process. Here, we introduce a mechanism of anaerobic biofilm formation by investigating the antibiofilm activity of tyrosol, a component in wine. Tyrosol inhibits E. coli and Pseudomonas aeruginosa biofilm formation by enhancing NO production. YbfA is identified as a target of tyrosol and its downstream targets are sequentially determined. YbfA activates YfeR, which then suppresses the anaerobic regulator FNR. This suppression leads to decreased NO production, elevated bis-(3'-5')-cyclic dimeric GMP levels, and finally stimulates anaerobic biofilm formation in the mature stage. Blocking YbfA with tyrosol treatment renders biofilm cells as susceptible to antibiotics as planktonic cells. Thus, this study presents YbfA as a promising antibiofilm target to address antibiotic resistance posed by biofilm-forming bacteria, with tyrosol acting as an inhibitor.


Subject(s)
Anti-Bacterial Agents , Biofilms , Escherichia coli , Nitric Oxide , Phenylethyl Alcohol , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Nitric Oxide/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/antagonists & inhibitors , Anaerobiosis/drug effects , Microbial Sensitivity Tests , Gene Expression Regulation, Bacterial/drug effects , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors
5.
Appl Microbiol Biotechnol ; 108(1): 418, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012538

ABSTRACT

Pseudomonas aeruginosa (P. aeruginosa) biofilm formation is a crucial cause of enhanced antibiotic resistance. Quorum sensing (QS) is involved in regulating biofilm formation; QS inhibitors block the QS signaling pathway as a new strategy to address bacterial resistance. This study investigated the potential and mechanism of L-HSL (N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide) as a QS inhibitor for P. aeruginosa. The results showed that L-HSL effectively inhibited the biofilm formation and dispersed the pre-formed biofilm of P. aeruginosa. The production of extracellular polysaccharides and the motility ability of P. aeruginosa were suppressed by L-HSL. C. elegans infection experiment showed that L-HSL was non-toxic and provided protection to C. elegans against P. aeruginosa infection. Transcriptomic analysis revealed that L-HSL downregulated genes related to QS pathways and biofilm formation. L-HSL exhibits a promising potential as a therapeutic drug for P. aeruginosa infection. KEY POINTS: • Chemical synthesis of N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide, named L-HSL. • L-HSL does not generate survival pressure on the growth of P. aeruginosa and can inhibit the QS system. • KEGG enrichment analysis found that after L-HSL treatment, QS-related genes were downregulated.


Subject(s)
4-Butyrolactone , Biofilms , Caenorhabditis elegans , Pseudomonas aeruginosa , Quorum Sensing , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/genetics , Biofilms/drug effects , Biofilms/growth & development , Quorum Sensing/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Animals , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , 4-Butyrolactone/metabolism , Anti-Bacterial Agents/pharmacology , Gene Expression Profiling , Homoserine/analogs & derivatives , Homoserine/metabolism , Homoserine/pharmacology , Gene Expression Regulation, Bacterial/drug effects
6.
Sci Rep ; 14(1): 16483, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39013998

ABSTRACT

The drug efflux pump is a crucial mechanism implicated in resistance to multiple antimicrobials. Thymoquinone (TQ) has evidently demonstrated multiple activities, antibacterial being the most effective. Knowledge about TQ activity against multidrug-resistant Staphylococcus aureus is very scarce. Therefore, the present study was conducted to investigate TQ resistance modulation in ciprofloxacin (CIP) and doxycycline (DO) multidrug-resistant S. aureus. Forty-seven samples were collected from different sources, and S. aureus was isolated and identified. Then, S. aureus resistance profiles to antimicrobials, N. sativa essential oil, and TQ; the correlation between TQ-MIC readings and disc diffusion; cartwheel and ethidium bromide (EtBr) accumulation assays; and norA gene expression were all described within silico molecular docking for TQ interactions with norA efflux pump protein. TQ-MICs ranged from 5-320 µg/ml. TQ down-regulated norA gene expression, resulting in a drop in efflux pump activity of 77.5-90.6% in the examined strains, comparable to that observed with verapamil. Exposure of S. aureus strains to CIP and DO raises the initial basal efflux pumping expression to 34.2 and 22.9 times, respectively. This induced efflux pumping overexpression was substantially reduced by 97.7% when TQ was combined with CIP or DO. There was a significant reduction of MICs of CIP and DO MICs by 2-15 and 2-4 folds, respectively, after treatment with 0.5XMIC-TQ in resistance modulation assays. These results refer to TQ ligand inhibitory interactions with NorA protein in molecular docking. Interpretations of inhibition zone diameters (IZDs) of disc diffusion and TQ-MICs exhibit independence of MICs from IZDs, as indicated by invalid linear regression analysis. TQ significantly reduced efflux pumping S. aureus induced by CIP and DO, but further investigations are needed to improve TQ-pharmacokinetics to restore CIP and DO activity and suppress fluoroquinolone and doxycycline-resistant S. aureus selection in clinical and animal settings.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Benzoquinones , Ciprofloxacin , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests , Molecular Docking Simulation , Multidrug Resistance-Associated Proteins , Staphylococcus aureus , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Benzoquinones/pharmacology , Benzoquinones/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Doxycycline/pharmacology , Gene Expression Regulation, Bacterial/drug effects
7.
Gene ; 927: 148754, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38972555

ABSTRACT

Pseudomonas aeruginosa PA2196 is a TetR family transcriptional repressor. In this study, the deletion of the PA2196 gene caused increased expression of the downstream gene curA (PA2197), which encodes for a NADPH-dependent curcumin/dihydrocurcumin reductase. The PA2196 gene was then identified as curR, and a DNA footprinting assay showed that CurR directly bound to the curA promoter at an imperfect 15-bp inverted repeat, 5'-TAGTTGA-C-TGGTCTA-3'. A curA promoter-lacZ fusion assay and site-directed mutagenesis further demonstrated that the identified CurR binding site plays a crucial role in curA repression by CurR. curA transcription was inducible by sodium hypochlorite (NaOCl) and N-ethylmaleimide (NEM) but not by hydrogen peroxide, organic hydroperoxide, or curcumin. The oxidation and alkylation of CurR by NaOCl and NEM, respectively, resulted in the inactivation of its DNA-binding activity, which induced curA expression. Under the tested conditions, the deletion of either curR or curA did not affect the survival of P. aeruginosa under NaOCl stress in the absence or presence of curcumin.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Pseudomonas aeruginosa , Repressor Proteins , Sodium Hypochlorite , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/drug effects , Sodium Hypochlorite/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Repressor Proteins/genetics , Repressor Proteins/metabolism , Promoter Regions, Genetic , Curcumin/pharmacology , Binding Sites , Oxidoreductases/genetics , Oxidoreductases/metabolism
8.
Med Microbiol Immunol ; 213(1): 16, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033094

ABSTRACT

Melioidosis is a severe infectious disease caused by Burkholderia pseudomallei, an intracellular pathogen with a high mortality rate and significant antibiotic resistance. The high mortality rate and resistance to antibiotics have drawn considerable attention from researchers studying melioidosis. This study evaluated the effects of various concentrations (75, 50, and 25 µg/mL) of promethazine hydrochloride (PTZ), a potent antihistamine, on biofilm formation and lipase activity after 24 h of exposure to B. thailandensis E264. A concentration-dependent decrease in both biofilm biomass and lipase activity was observed. RT-PCR analysis revealed that PTZ treatment not only made the biofilm structure loose but also reduced the expression of btaR1, btaR2, btaR3, and scmR. Single gene knockouts of quorum sensing (QS) receptor proteins (∆btaR1, ∆btaR2, and ∆btaR3) were successfully constructed. Deletion of btaR1 affected biofilm formation in B. thailandensis, while deletion of btaR2 and btaR3 led to reduced lipase activity. Molecular docking and biological performance results demonstrated that PTZ inhibits biofilm formation and lipase activity by suppressing the expression of QS-regulated genes. This study found that repositioning PTZ reduced biofilm formation in B. thailandensis E264, suggesting a potential new approach for combating melioidosis.


Subject(s)
Biofilms , Burkholderia , Drug Repositioning , Promethazine , Biofilms/drug effects , Biofilms/growth & development , Burkholderia/drug effects , Burkholderia/physiology , Burkholderia/genetics , Promethazine/pharmacology , Molecular Docking Simulation , Anti-Bacterial Agents/pharmacology , Lipase/metabolism , Lipase/genetics , Gene Expression Regulation, Bacterial/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Quorum Sensing/drug effects
9.
Arch Microbiol ; 206(7): 289, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847838

ABSTRACT

Staphylococcus epidermidis is an opportunistic pathogen commonly implicated in medical device-related infections. Its propensity to form biofilms not only leads to chronic infections but also exacerbates the issue of antibiotic resistance, necessitating high-dose antimicrobial treatments. In this study, we explored the use of diclofenac sodium, a non-steroidal anti-inflammatory drug, as an anti-biofilm agent against S. epidermidis. In this study, crystal violet staining and confocal laser scanning microscope analysis showed that diclofenac sodium, at subinhibitory concentration (0.4 mM), significantly inhibited biofilm formation in both methicillin-susceptible and methicillin-resistant S. epidermidis isolates. MTT assays demonstrated that 0.4 mM diclofenac sodium reduced the metabolic activity of biofilms by 25.21-49.01% compared to untreated controls. Additionally, the treatment of diclofenac sodium resulted in a significant decrease (56.01-65.67%) in initial bacterial adhesion, a crucial early phase of biofilm development. Notably, diclofenac sodium decreased the production of polysaccharide intercellular adhesin (PIA), a key component of the S. epidermidis biofilm matrix, in a dose-dependent manner. Real-time quantitative PCR analysis revealed that diclofenac sodium treatment downregulated biofilm-associated genes icaA, fnbA, and sigB and upregulated negative regulatory genes icaR and luxS, providing potential mechanistic insights. These findings indicate that diclofenac sodium inhibits S. epidermidis biofilm formation by affecting initial bacterial adhesion and the PIA synthesis. This underscores the potential of diclofenac sodium as a supplementary antimicrobial agent in combating staphylococcal biofilm-associated infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Diclofenac , Staphylococcus epidermidis , Biofilms/drug effects , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/physiology , Diclofenac/pharmacology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Adhesion/drug effects , Humans , Polysaccharides, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Gene Expression Regulation, Bacterial/drug effects
10.
Arch Microbiol ; 206(7): 301, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874781

ABSTRACT

Biofilm formation by methicillin-resistant Staphylococcus aureus (MRSA) on indwelling medical devices complicates the treatment of infection. Tetrabromobisphenol A (TBBPA), a synthetic, lipophilic, halogenated aromatic compound widely used as an additive in plastics and electronic products, has raised environmental concerns due to its potential for bioaccumulation. This study investigated the impact of sub-inhibitory concentrations of TBBPA on MRSA biofilm formation. Crystal violet staining and confocal laser scanning microscopy analysis demonstrated that 1/8 MIC (0.5 µg/mL) of TBBPA significantly stimulated MRSA biofilm formation (P < 0.0001). MTT assays indicated that the metabolic activity within the biofilms increased by 15.60-40.85% compared to untreated controls. Dot blot immunoassay, autolysis assay, and extracellular DNA (eDNA) quantification further revealed TBBPA enhanced the production of polysaccharide intercellular adhesin (PIA) and eDNA, which are key biofilm components. Additionally, TBBPA was found to enhance the production of staphyloxanthin, facilitating MRSA survival under oxidative conditions and in human whole blood. RT-qPCR analysis showed that TBBPA significantly upregulated genes associated with biofilm formation (icaA, atlA, sarA), staphyloxanthin biosynthesis (crtM and sigB), and oxidative stress responses (sodA and katA). These findings suggest that TBBPA promotes MRSA biofilm development and enhances bacterial resistance to adverse conditions, thereby potentially exacerbating risks to human health.


Subject(s)
Biofilms , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Polybrominated Biphenyls , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Polybrominated Biphenyls/pharmacology , Humans , Xanthophylls/metabolism , Xanthophylls/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects
11.
Mol Biol Rep ; 51(1): 761, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874884

ABSTRACT

BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) poses a great health threat to humans. Looking for compounds that could reduce the resistance of S. aureus towards methicillin is an effective way to alleviate the antimicrobial resistance crisis. METHODS AND RESULTS: Minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), Time-killing growth curve, staphyloxanthin and penicillin-binding protein 2a (PBP2a) were detected. A quantitative polymerase chain reaction was used to measure the effect of BBH on the gene transcription profiles of MRSA. The MIC of MRSA-ST59-t437 towards oxacillin was 8 µg/ml, and MBC was 128 µg/ml. After adding a sub-inhibitory concentration of BBH, the MIC and MBC of MRSA-ST59-t478 towards oxacillin went down to 0.125 and 32 µg/ml respectively. The amount of PBP2a and staphyloxanthin were reduced after treatment with BBH. Moreover, the transcription levels of sarA, mecA and fni genes were downregulated. CONCLUSIONS: It is for the first time reported that BBH could inhibit staphyloxanthin synthesis by inhibiting fni gene. Moreover, fni might be the target gene of sarA, and there might be another regulatory pathway to inhibit staphyloxanthin biosynthesis. BBH could effectively reduce the methicillin resistance of MRSA-ST59-t437 by downregulating fni, sarA and mecA genes.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Berberine , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Xanthophylls , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Xanthophylls/pharmacology , Berberine/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Oxacillin/pharmacology
12.
Nat Commun ; 15(1): 5498, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38944647

ABSTRACT

IncX3 plasmids carrying the New Delhi metallo-ß-lactamase-encoding gene, blaNDM-5, are rapidly spreading globally in both humans and animals. Given that carbapenems are listed on the WHO AWaRe watch group and are prohibited for use in animals, the drivers for the successful dissemination of Carbapenem-Resistant Enterobacterales (CRE) carrying blaNDM-5-IncX3 plasmids still remain unknown. We observe that E. coli carrying blaNDM-5-IncX3 can persist in chicken intestines either under the administration of amoxicillin, one of the largest veterinary ß-lactams used in livestock, or without any antibiotic pressure. We therefore characterise the blaNDM-5-IncX3 plasmid and identify a transcription regulator, VirBR, that binds to the promoter of the regulator gene actX enhancing the transcription of Type IV secretion systems (T4SS); thereby, promoting conjugation of IncX3 plasmids, increasing pili adhesion capacity and enhancing the colonisation of blaNDM-5-IncX3 transconjugants in animal digestive tracts. Our mechanistic and in-vivo studies identify VirBR as a major factor in the successful spread of blaNDM-5-IncX3 across one-health AMR sectors. Furthermore, VirBR enhances the plasmid conjugation and T4SS expression by the presence of copper and zinc ions, thereby having profound ramifications on the use of universal animal feeds.


Subject(s)
Anti-Bacterial Agents , Chickens , Conjugation, Genetic , Escherichia coli , Plasmids , beta-Lactamases , Animals , Plasmids/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism , Chickens/microbiology , Humans , Escherichia coli/genetics , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amoxicillin/pharmacology , Promoter Regions, Genetic/genetics , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Escherichia coli Infections/transmission , Gene Expression Regulation, Bacterial/drug effects , Intestines/microbiology
13.
Microb Pathog ; 193: 106762, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936638

ABSTRACT

Enterococcus faecalis is a troublesome nosocomial pathogen that acquired resistance to most available antimicrobial agents. Antivirulence agents represent an unconventional treatment approach. Here, transcription factor decoy (TFD)-loaded cationic liposomes (TLL) were developed as an inhibitor of the Fsr quorum-sensing system and its associated virulence traits, in E. faecalis. The consensus sequence of the FsrA binding site was found conserved among 651 E. faecalis annotated genomes. The TFD was synthesized as an 82 bp DNA duplex, containing the conserved binding sequence, and loaded onto cationic liposomes. The optimum loading capacity, mean particle size, and zeta potential of the TLL were characterized. The developed TLL lacked any effect on E. faecalis growth and significantly inhibited the in vitro production of the proteolytic enzymes controlled by the Fsr system; gelatinase and serine protease, in a concentration-dependent manner. This inhibition was accompanied by a significant reduction in the transcription levels of FsrA-regulated genes (fsrB, gelE, and sprE). The developed TLL were safe as evidenced by the nonhemolytic effect on human RBCs and the negligible cytotoxicity on human skin fibroblast cells. Moreover, in the larvae infection model, TLL displayed a significant abolish in the mortality rates of Galleria mellonella larvae infected with E. faecalis. In conclusion, the developed TLL offer a new safe strategy for combating E. faecalis infection through the inhibition of quorum-sensing-mediated virulence; providing a platform for the development of similar agents to combat many other pathogens.


Subject(s)
Bacterial Proteins , Enterococcus faecalis , Gram-Positive Bacterial Infections , Quorum Sensing , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Animals , Virulence/drug effects , Humans , Quorum Sensing/drug effects , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/drug therapy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Liposomes , Larva/microbiology , Transcription Factors/metabolism , Transcription Factors/genetics , Virulence Factors/genetics , Gelatinases/metabolism , Gelatinases/antagonists & inhibitors , Moths/microbiology , Erythrocytes/drug effects , Disease Models, Animal , Serine Proteases/metabolism , Serine Proteases/genetics , Gene Expression Regulation, Bacterial/drug effects , Nanoparticles/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
14.
mSystems ; 9(7): e0016524, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38837409

ABSTRACT

The communication between plants and their microbiota is highly dynamic and involves a complex network of signal molecules. Among them, the auxin indole-3-acetic acid (IAA) is a critical phytohormone that not only regulates plant growth and development, but is emerging as an important inter- and intra-kingdom signal that modulates many bacterial processes that are important during interaction with their plant hosts. However, the corresponding signaling cascades remain largely unknown. Here, we advance our understanding of the largely unknown mechanisms by which IAA carries out its regulatory functions in plant-associated bacteria. We showed that IAA caused important changes in the global transcriptome of the rhizobacterium Serratia plymuthica and multidisciplinary approaches revealed that IAA sensing interferes with the signaling mediated by other pivotal plant-derived signals such as amino acids and 4-hydroxybenzoic acid. Exposure to IAA caused large alterations in the transcript levels of genes involved in amino acid metabolism, resulting in significant metabolic alterations. IAA treatment also increased resistance to toxic aromatic compounds through the induction of the AaeXAB pump, which also confers resistance to IAA. Furthermore, IAA promoted motility and severely inhibited biofilm formation; phenotypes that were associated with decreased c-di-GMP levels and capsule production. IAA increased capsule gene expression and enhanced bacterial sensitivity to a capsule-dependent phage. Additionally, IAA induced the expression of several genes involved in antibiotic resistance and led to changes in the susceptibility and responses to antibiotics with different mechanisms of action. Collectively, our study illustrates the complexity of IAA-mediated signaling in plant-associated bacteria. IMPORTANCE: Signal sensing plays an important role in bacterial adaptation to ecological niches and hosts. This communication appears to be particularly important in plant-associated bacteria since they possess a large number of signal transduction systems that respond to a wide diversity of chemical, physical, and biological stimuli. IAA is emerging as a key inter- and intra-kingdom signal molecule that regulates a variety of bacterial processes. However, despite the extensive knowledge of the IAA-mediated regulatory mechanisms in plants, IAA signaling in bacteria remains largely unknown. Here, we provide insight into the diversity of mechanisms by which IAA regulates primary and secondary metabolism, biofilm formation, motility, antibiotic susceptibility, and phage sensitivity in a biocontrol rhizobacterium. This work has important implications for our understanding of bacterial ecology in plant environments and for the biotechnological and clinical applications of IAA, as well as related molecules.


Subject(s)
Cyclic GMP , Indoleacetic Acids , Serratia , Indoleacetic Acids/metabolism , Serratia/metabolism , Serratia/genetics , Serratia/drug effects , Serratia/pathogenicity , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial/drug effects , Bacteriophages , Signal Transduction/drug effects , Biofilms/drug effects , Biofilms/growth & development , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Plant Growth Regulators/metabolism , Transcriptome/drug effects
15.
Acta Microbiol Immunol Hung ; 71(2): 127-133, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38869956

ABSTRACT

Pseudomonas aeruginosa has been in the center of attention for several years as an opportunistic human pathogen implicated in many severe acute and chronic infections particularly in immunocompromised patients. Its high persistence and resistance against many antimicrobial agents are mostly attributed to biofilm formation. Biofilms are microbial communities mainly consisting of extracellular polymeric substances that encapsulate bacteria together and protect them from extracellular stresses. This cell aggregation is a stress response that P. aeruginosa employes as a survival strategy during growth with the toxic detergents. This process has shown to involve several operons such as psl, pel, and alg. Here we used P. aeruginosa strain PAO1 in control group, 40 P. aeruginosa strains from sink and 40 strains from surface of public places. Biofilm formation and gene expression were measured before and after exposure to sub minimum inhibitory concentration (sub-MIC) of biocides chlorhexidine diacetate and benzalkonium chloride. The qRT-PCR and biofilm formation results demonstrated an increase in biofilm formation ability and gene expression of pslA/B and pelA/B in two groups collected from sink and surface in contrast to the control group. A remarkable increase was observed in the biofilm formation and expression of pslA in the bacterial strain collected from the sink after exposure to biocides chlorhexidine diacetate. Both Pel and Psl appeared to have redundant functions as structural scaffolds in biofilms. Sub-MIC levels of detergents can improve biofilm formation ability of P. aeruginosa and therefore trigger resistance.


Subject(s)
Bacterial Proteins , Biofilms , Detergents , Gene Expression Regulation, Bacterial , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/drug effects , Detergents/pharmacology , Humans
16.
mSphere ; 9(6): e0031724, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38837389

ABSTRACT

The emerging prevalence of drug-resistant Staphylococcus aureus isolates underscores the urgent need for alternative therapeutic strategies due to the declining effectiveness of traditional antibiotics in clinical settings. MgrA, a key virulence regulator in S. aureus, orchestrates the expression of numerous virulence factors. Here, we report the discovery of isorhapontigenin, a methoxylated analog of resveratrol, as a potential anti-virulence agent against S. aureus. Isorhapontigenin effectively inhibits the hemolytic activity of S. aureus in a non-bactericidal manner. Additionally, it significantly reduces the cytotoxicity of S. aureus and impairs its ability to survive in macrophages. Mechanistically, isorhapontigenin modulates the expression of virulence factors, dose-dependently downregulating hla and upregulating the MgrA-regulated gene spa. Electrophoretic mobility shift assays demonstrated that isorhapontigenin inhibits the binding of MgrA to the hla promoter in a dose-dependent manner. Thermal shift assays confirmed the direct interaction between isorhapontigenin and the MgrA protein. The in vivo experiments demonstrated that isorhapontigenin significantly reduced the area of skin abscesses and improved survival in a pneumonia model while decreasing bacterial burden and inflammation in the lungs. In conclusion, isorhapontigenin holds potential as a candidate drug for further development as an anti-virulence agent for treating S. aureus infections. IMPORTANCE: The emergence of antibiotic-resistant Staphylococcus aureus strains presents a formidable challenge to public health, necessitating novel approaches in combating these pathogens. Traditional antibiotics are becoming increasingly ineffective, leading to a pressing need for innovative therapeutic strategies. In this study, targeting virulence factors that play a crucial role in the pathogenesis of bacterial infections offers a promising alternative to circumvent resistance mechanisms. The discovery of isorhapontigenin as an inhibitor of S. aureus virulence represents a significant advance in anti-virulence therapy.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Gene Expression Regulation, Bacterial , Staphylococcal Infections , Staphylococcus aureus , Virulence Factors , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/genetics , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Virulence/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Mice , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Gene Expression Regulation, Bacterial/drug effects , Stilbenes/pharmacology , Humans , Macrophages/microbiology , Macrophages/drug effects , Female , Mice, Inbred BALB C , RAW 264.7 Cells
17.
Int J Biol Macromol ; 273(Pt 1): 133029, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852716

ABSTRACT

This study determined the inhibitory mechanism as well as anti-biofilm activity of chlorogenic acid-grafted-chitosan (CS-g-CA) against Pseudomonas fluorescens (P. fluorescens) in terms of biofilm content, oxidative stress, quorum sensing and cyclic diguanosine monophosphate (c-di-GMP) concentration, and detected the changes in the expression levels of related genes by quantitative real-time PCR (qRT-PCR). Results indicated that treatment with sub-concentrations of CS-g-CA for P. fluorescens led to reduce the biofilm size of large colonies, decrease the content of biofilm and extracellular polymers, weaken the motility and adhesion of P. fluorescens. Moreover, CS-g-CA resulted in higher ROS levels, diminished catalase activity (CAT), and increased superoxide dismutase (SOD) in P. fluorescens. CS-g-CA reduced the production of quorum-sensing signaling molecules (AHLs) and the concentration of c-di-GMP in bacteria. Genes for flagellar synthesis (flgA), the resistance to stress (rpoS and hfq), and pde (phosphodiesterases that degrade c-di-GMP) were significantly down-regulated as determined by RT-PCR. Overall, CS-g-CA leads to the accumulation of ROS in bacteria via P. fluorescens environmental resistance genes and decreases the activity of enzymes in the bacterial antioxidant system, and interferes with the production and reception of quorum-sensing signaling molecules and the synthesis of c-di-GMP in P. fluorescens, which regulates the generation of biofilms.


Subject(s)
Biofilms , Chitosan , Chlorogenic Acid , Cyclic GMP , Oxidative Stress , Pseudomonas fluorescens , Quorum Sensing , Pseudomonas fluorescens/drug effects , Pseudomonas fluorescens/metabolism , Chitosan/chemistry , Chitosan/pharmacology , Biofilms/drug effects , Quorum Sensing/drug effects , Oxidative Stress/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Chlorogenic Acid/pharmacology , Chlorogenic Acid/chemistry , Gene Expression Regulation, Bacterial/drug effects , Reactive Oxygen Species/metabolism
18.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38928116

ABSTRACT

Achromobacter insolitus and Achromobacter aegrifaciens, bacterial degraders of the herbicide glyphosate, were found to induce phosphonatase (phosphonoacetaldehyde hydrolase, EC 3.11.1.1) when grown on minimal media with glyphosate as the sole source of phosphorus. The phosphonatases of the strains were purified to an electrophoretically homogeneous state and characterized. The enzymes differed in their kinetic characteristics and some other parameters from the previously described phosphonatases. The phosphonatase of A. insolitus was first revealed to separate into two stable forms, which had similar kinetic characteristics but interacted differently with affinity and ion-exchange resins. The genomes of the investigated bacteria were sequenced. The phosphonatase genes were identified, and their context was determined: the bacteria were shown to have gene clusters, which, besides the phosphonatase operon, included genes for LysR-type transcription activator (substrate sensor) and putative iron-containing oxygenase PhnHD homologous to monooxygenases PhnY and TmpB of marine organophosphonate degraders. Genes of 2-aminoethylphosphonate aminotransferase (PhnW, EC 2.6.1.37) were absent in the achromobacterial phosphonatase operons; instead, we revealed the presence of genes encoding the putative flavin oxidase HpnW. In silico simulation showed 1-hydroxy-2-aminoethylphosphonate to be the most likely substrate of the new monooxygenase, and a number of glycine derivatives structurally similar to glyphosate to be substrates of flavin oxidase.


Subject(s)
Achromobacter , Glycine , Glyphosate , Operon , Soil Microbiology , Glycine/analogs & derivatives , Achromobacter/genetics , Operon/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Herbicides , Multigene Family , Kinetics , Gene Expression Regulation, Bacterial/drug effects
19.
Microb Pathog ; 193: 106739, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857709

ABSTRACT

Enterococcus faecalis, an opportunistic pathogen responsible for nosocomial infections, exhibits increased pathogenicity via biofilm formation. Theaflavin-3,3'-digallate (TF3), a theaflavin extracted from black tea, exhibits potent antibacterial effects. In the present study, we investigated the inhibitory effect of TF3 on E. faecalis. Our results indicated that TF3 significantly inhibited E. faecalis ATCC 29212 biofilm formation. This observation was further confirmed via crystal violet staining, confocal laser scanning microscopy, and field emission-scanning electron microscopy. To disclose the underlying mechanisms, RNA-seq was applied. TF3 treatment significantly altered the transcriptomic profile of E. faecalis, as evidenced by identification of 248 differentially expressed genes (DEGs). Through functional annotation of these DEGs, several quorum-sensing pathways were found to be suppressed in TF3-treated cultures. Further, gene expression verification via real-time PCR confirmed the downregulation of gelE, sprE, and secY by TF3. These findings highlighted the ability of TF3 to impede E. faecalis biofilm formation, suggesting a novel preventive strategy against E. faecalis infections.


Subject(s)
Anti-Bacterial Agents , Biflavonoids , Biofilms , Catechin , Enterococcus faecalis , Gene Expression Regulation, Bacterial , Quorum Sensing , Biofilms/drug effects , Biofilms/growth & development , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Quorum Sensing/drug effects , Biflavonoids/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Catechin/pharmacology , Catechin/analogs & derivatives , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcriptome/drug effects , Gene Expression Profiling
20.
Proc Natl Acad Sci U S A ; 121(27): e2407832121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38935560

ABSTRACT

In 1967, in this journal, Evelyn Witkin proposed the existence of a coordinated DNA damage response in Escherichia coli, which later came to be called the "SOS response." We revisited this response using the replication inhibitor azidothymidine (AZT) and RNA-Seq analysis and identified several features. We confirm the induction of classic Save our ship (SOS) loci and identify several genes, including many of the pyrimidine pathway, that have not been previously demonstrated to be DNA damage-inducible. Despite a strong dependence on LexA, these genes lack LexA boxes and their regulation by LexA is likely to be indirect via unknown factors. We show that the transcription factor "stringent starvation protein" SspA is as important as LexA in the regulation of AZT-induced genes and that the genes activated by SspA change dramatically after AZT exposure. Our experiments identify additional LexA-independent DNA damage inducible genes, including 22 small RNA genes, some of which appear to activated by SspA. Motility and chemotaxis genes are strongly down-regulated by AZT, possibly as a result of one of more of the small RNAs or other transcription factors such as AppY and GadE, whose expression is elevated by AZT. Genes controlling the iron siderophore, enterobactin, and iron homeostasis are also strongly induced, independent of LexA. We confirm that IraD antiadaptor protein is induced independent of LexA and that a second antiadaptor, IraM is likewise strongly AZT-inducible, independent of LexA, suggesting that RpoS stabilization via these antiadaptor proteins is an integral part of replication stress tolerance.


Subject(s)
DNA Damage , DNA Replication , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/drug effects , Gene Expression Regulation, Bacterial/drug effects , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , DNA Replication/drug effects , SOS Response, Genetics/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Serine Endopeptidases
SELECTION OF CITATIONS
SEARCH DETAIL