Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.438
Filter
1.
Curr Microbiol ; 81(10): 344, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235595

ABSTRACT

The problem of bacterial resistance caused by antibiotic abuse is seriously detrimental to global human health and ecosystem security. The two-dimensional nanomaterial (2D) such as black phosphorus (BP) is recently expected to become a new bacterial inhibitor and has been widely used in the antibacterial field due to its specific physicochemical properties. Nevertheless, the effects of 2D-BP on the propagation of antibiotic resistance genes (ARGs) in environments and the relevant mechanisms are not clear. Herein, we observed that the sub-inhibitory concentrations of 2D-BP dramatically increased the conjugative transfer of ARGs mediated by the RP4 plasmid up to 2.6-fold at the 125 mg/L exposure level compared with the untreated bacterial cells. Nevertheless, 2D-BP with the inhibitory concentration caused a dramatic decrease in the conjugative frequency. The phenotypic changes revealed that the increase of the conjugative transfer caused by 2D-BP exposure were attributed to the excessive reactive oxygen species and oxidative stress, and increased bacterial cell membrane permeability. The genotypic evidence demonstrated that 2D-BP affecting the horizontal gene transfer of ARGs was probably through the upregulation of mating pair formation genes (trbBp and traF) and DNA transfer and replication genes (trfAp and traJ), as well as the downregulation of global regulatory gene expression (korA, korB, and trbA). In summary, the changes in the functional and regulatory genes in the conjugative transfer contributed to the stimulation of conjugative transfer. This research aims to broaden our comprehension of how nanomaterials influence the dissemination of ARGs by elucidating their effects and mechanisms.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Gene Transfer, Horizontal , Phosphorus , Plasmids , Plasmids/genetics , Phosphorus/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Conjugation, Genetic , Escherichia coli/genetics , Escherichia coli/drug effects , Nanostructures , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Bacteria/genetics , Bacteria/drug effects
2.
PLoS One ; 19(8): e0306597, 2024.
Article in English | MEDLINE | ID: mdl-39106246

ABSTRACT

Gossypol, a yellow polyphenolic compound found in the Gossypium genus, is toxic to animals that ingest cotton-derived feed materials. However, ruminants display a notable tolerance to gossypol, attributed to the pivotal role of ruminal microorganisms in its degradation. The mechanisms of how rumen microorganisms degrade and tolerate gossypol remain unclear. Therefore, in this study, Enterobacter sp. GD5 was isolated from rumen fluid, and the effects of gossypol on its metabolism and gene expression were investigated using liquid chromatography-mass spectrometry (LC-MS) and RNA analyses. The LC-MS results revealed that gossypol significantly altered the metabolic profiles of 15 metabolites (eight upregulated and seven downregulated). The Kyoto Encyclopedia of Genes and Genomes analysis results showed that significantly different metabolites were associated with glutathione metabolism in both positive and negative ion modes, where gossypol significantly affected the biosynthesis of amino acids in the negative ion mode. Transcriptomic analysis indicated that gossypol significantly affected 132 genes (104 upregulated and 28 downregulated), with significant changes observed in the expression of catalase peroxidase, glutaredoxin-1, glutathione reductase, thioredoxin 2, thioredoxin reductase, and alkyl hydroperoxide reductase subunit F, which are related to antioxidative stress. Furthermore, Gene Ontology analysis revealed significant changes in homeostatic processes following gossypol supplementation. Overall, these results indicate that gossypol induces oxidative stress, resulting in the increased expression of antioxidative stress-related genes in Enterobacter sp. GD5, which may partially explain its tolerance to gossypol.


Subject(s)
Enterobacter , Gossypol , Metabolomics , Gossypol/pharmacology , Gossypol/metabolism , Enterobacter/metabolism , Enterobacter/genetics , Enterobacter/drug effects , Animals , Transcriptome/drug effects , Gene Expression Regulation, Bacterial/drug effects , Metabolome/drug effects , Gene Expression Profiling , Rumen/microbiology , Rumen/metabolism , Rumen/drug effects
3.
Sci Rep ; 14(1): 19145, 2024 08 19.
Article in English | MEDLINE | ID: mdl-39160175

ABSTRACT

Carbapenem-resistant Acinetobacter baumannii (CRAB) is a major human pathogen and a research priority for developing new antimicrobial agents. CRAB is a causative agent of a variety of infections in different body sites. One of the manifestations is catheter-associated urinary tract infection, which exposes the bacteria to the host's urine, creating a particular environment. Exposure of two CRAB clinical isolates, AB5075 and AMA40, to human urine (HU) resulted in the differential expression levels of 264 and 455 genes, respectively, of which 112 were common to both strains. Genes within this group play roles in metabolic pathways such as phenylacetic acid (PAA) catabolism, the Hut system, the tricarboxylic acid (TCA) cycle, and other processes like quorum sensing and biofilm formation. These results indicate that the presence of HU induces numerous adaptive changes in gene expression of the infecting bacteria. These changes presumably help bacteria establish and thrive in the hostile conditions in the urinary tract. These analyses advance our understanding of CRAB's metabolic adaptations to human fluids, as well as expand knowledge on bacterial responses to distinct human fluids containing different concentrations of human serum albumin (HSA).


Subject(s)
Acinetobacter baumannii , Carbapenems , Urine , Acinetobacter baumannii/genetics , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/metabolism , Humans , Carbapenems/pharmacology , Urine/microbiology , Gene Expression Regulation, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Acinetobacter Infections/microbiology , Acinetobacter Infections/urine , Adaptation, Physiological/genetics , Urinary Tract Infections/microbiology , Biofilms/growth & development , Biofilms/drug effects , Drug Resistance, Bacterial/genetics
4.
BMC Microbiol ; 24(1): 306, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152378

ABSTRACT

BACKGROUND: Deoxynivalenol (DON) is a type B trichothecene mycotoxin that is commonly found in cereals and grains worldwide. The presence of this fungal secondary-metabolite raises public-health concerns at both the agriculture and food industry level. Recently, we have shown that DON has a negative impact on gut integrity, a feature also noticed for Campylobacter (C.) jejuni. We further demonstrated that DON increased the load of C. jejuni in the gut and inner organs. In contrast, feeding the less toxic DON metabolite deepoxy-deoxynivalenol (DOM-1) to broilers reduced the Campylobacter load in vivo. Consequently, it can be hypothesized that DON and DOM-1 have a direct effect on the growth profile of C. jejuni. The aim of the present study was to further resolve the nature of this interaction in vitro by co-incubation and RNA-sequencing. RESULTS: The co-incubation of C. jejuni with DON resulted in significantly higher bacterial growth rates from 30 h of incubation onwards. On the contrary, the co-incubation of C. jejuni with DOM-1 reduced the CFU counts, indicating that this DON metabolite might contribute to reduce the burden of C. jejuni in birds, altogether confirming in vivo data. Furthermore, the transcriptomic profile of C. jejuni following incubation with either DON or DOM-1 differed. Co-incubation of C. jejuni with DON significantly increased the expression of multiple genes which are critical for Campylobacter growth, particularly members of the Flagella gene family, frr (ribosome-recycling factor), PBP2 futA-like (Fe3+ periplasmic binding family) and PotA (ATP-binding subunit). Flagella are responsible for motility, biofilm formation and host colonization, which may explain the high Campylobacter load in the gut of DON-fed broiler chickens. On the contrary, DOM-1 downregulated the Flagella gene family and upregulated ribosomal proteins. CONCLUSION: The results highlight the adaptive mechanisms involved in the transcriptional response of C. jejuni to DON and its metabolite DOM-1, based on the following effects: (a) ribosomal proteins; (b) flagellar proteins; (c) engagement of different metabolic pathways. The results provide insight into the response of an important intestinal microbial pathogen against DON and lead to a better understanding of the luminal or environmental acclimation mechanisms in chickens.


Subject(s)
Campylobacter jejuni , Chickens , Transcriptome , Trichothecenes , Trichothecenes/metabolism , Campylobacter jejuni/drug effects , Campylobacter jejuni/genetics , Campylobacter jejuni/growth & development , Campylobacter jejuni/metabolism , Animals , Transcriptome/drug effects , Chickens/microbiology , Gene Expression Regulation, Bacterial/drug effects , Campylobacter Infections/microbiology , Campylobacter Infections/veterinary , Animal Feed/microbiology
5.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 15-21, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097900

ABSTRACT

A biofilm is a complex microbial structure that promotes the progression of persistent infections, particularly in nosocomial settings via indwelling medical devices. Conventional antibiotics are often ineffective treatments for biofilms; hence, it is crucial to investigate or design non-antibiotic antibiofilm compounds that can successfully reduce and eradicate biofilm-related infections. This study was an attempt to repurpose chronic disease medications of the antihypertensive and antilipidemic drug classes, including candesartan cilexetil (CC) and ursodeoxycholic acid (UDCA), respectively, to be used as antibiofilm agents against the two infectious pathogens Staphylococcus aureus and Enterococcus faecalis. Crystal violet (CV) staining assay was used to evaluate the antibiofilm activity of the drugs. Real-time polymerase chain reaction (RT-PCR) was performed to determine the transcription levels of the biofilm-related genes (icaA and icaR in S. aureus and fsrC and gelE in E. faecalis) following treatment with different concentrations of CC and UDCA. we found that a concentration of greater than 1.5 µg/ml of CC significantly (p < 0.005) inhibited the biofilm formation of both bacterial isolates, and a concentration of greater than 50 µg/ml of UDCA significantly (p < 0.005) inhibited the biofilm formation of both bacterial isolates. Interestingly, the mRNA expression levels of biofilm-related genes were decreased in the two bacterial isolates at concentrations that were lower than the human pharmaceutical daily doses.


Subject(s)
Biofilms , Enterococcus faecalis , Staphylococcus aureus , Ursodeoxycholic Acid , Biofilms/drug effects , Biofilms/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology , Humans , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Enterococcus faecalis/physiology , Ursodeoxycholic Acid/pharmacology , Anti-Bacterial Agents/pharmacology , Chronic Disease , Microbial Sensitivity Tests , Gene Expression Regulation, Bacterial/drug effects , Benzimidazoles/pharmacology , Tetrazoles/pharmacology , Biphenyl Compounds/pharmacology
6.
BMC Microbiol ; 24(1): 312, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182027

ABSTRACT

BACKGROUND: More than a century has passed since it was discovered that many bacteria produce indole, but research into the actual biological roles of this molecule is just now beginning. The influence of indole on bacterial virulence was extensively investigated in indole-producing bacteria like Escherichia coli. To gain a deeper comprehension of its functional role, this study investigated how indole at concentrations of 0.5-1.0 mM found in the supernatant of Escherichia coli stationary phase culture was able to alter the virulence of non-indole-producing bacteria, such as Pseudomonas aeruginosa, Proteus mirabilis, and Klebsiella pneumoniae, which are naturally exposed to indole in mixed infections with Escherichia coli. RESULTS: Biofilm formation, antimicrobial susceptibility, and efflux pump activity were the three phenotypic tests that were assessed. Indole was found to influence antibiotic susceptibly of Pseudomonas aeruginosa, Proteus mirabilis and Klebsiella pneumoniae to ciprofloxacin, imipenem, ceftriaxone, ceftazidime, and amikacin through significant reduction in MIC with fold change ranged from 4 to 16. Biofilm production was partially abrogated in both 32/45 Pseudomonas aeruginosa and all eight Proteus mirabilis, while induced biofilm production was observed in 30/40 Klebsiella pneumoniae. Moreover, acrAB and oqxAB, which encode four genes responsible for resistance-nodulation-division multidrug efflux pumps in five isolates of Klebsiella pneumoniae were investigated genotypically using quantitative real-time (qRT)-PCR. This revealed that all four genes exhibited reduced expression indicated by 2^-ΔΔCT < 1 in indole-treated isolates compared to control group. CONCLUSION: The outcomes of qRT-PCR investigation of efflux pump expression have established a novel clear correlation of the molecular mechanism that lies beneath the influence of indole on bacterial antibiotic tolerance. This research provides novel perspectives on the various mechanisms and diverse biological functions of indole signaling and how it impacts the pathogenicity of non-indole-producing bacteria.


Subject(s)
Anti-Bacterial Agents , Biofilms , Escherichia coli , Indoles , Klebsiella pneumoniae , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Biofilms/growth & development , Biofilms/drug effects , Indoles/metabolism , Indoles/pharmacology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Gene Expression Regulation, Bacterial/drug effects , Down-Regulation , Proteus mirabilis/genetics , Proteus mirabilis/drug effects , Proteus mirabilis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Virulence/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
7.
Gene ; 930: 148860, 2024 Dec 20.
Article in English | MEDLINE | ID: mdl-39151675

ABSTRACT

Since ectoine is a high-value product, overviewing strategies for identifying novel microbial sources becomes relevant. In the current study, by following a genome mining approach, the ectoine biosynthetic cluster in a tropical marine strain of Nocardiopsis dassonvillei (NCIM 5124) was located and compared with related organisms. Transcriptome analysis of Control and Test samples (with 0 and 5% NaCl, respectively) was carried out to understand salt induced stress response at the molecular level. There were 4950 differentially expressed genes with 25 transcripts being significantly upregulated in Test samples. NaCl induced upregulation of the ectoine biosynthesis cluster and some other genes (stress response, chaperone/Clp protease, cytoplasm, ribonucleoprotein and protein biosynthesis). The production of ectoine as a stress response molecule was experimentally validated via LCMS analysis. The investigation sheds light on the responses exhibited by this actinomycete in coping up with salt stress and provides a foundation for understanding salt induced molecular interactions.


Subject(s)
Amino Acids, Diamino , Transcriptome , Amino Acids, Diamino/metabolism , Amino Acids, Diamino/biosynthesis , Actinobacteria/genetics , Actinobacteria/metabolism , Gene Expression Profiling/methods , Genomics/methods , Genome, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Multigene Family , Salt Stress/genetics , Sodium Chloride/pharmacology
8.
PLoS One ; 19(8): e0309307, 2024.
Article in English | MEDLINE | ID: mdl-39196973

ABSTRACT

BACKGROUND: Colistin resistance in Acinetobacter baumannii is an emerging problem that limits antimicrobial therapy options. MATERIALS & METHODS: We isolated two pairs of colistin susceptible and colistin-resistant A. baumannii (K1007/K1006 and K408/K409) from two patients diagnosed with carbapenem-resistant A. baumannii infection. Colistin susceptible isolates were exposed to in vitro colistin induction for 50 generations. The selected cell populations were subjected to DNA and RNA sequencing and phenotypic assays. RESULTS: In the in vitro induction assay, K408 gained colistin resistance on the corresponding day of clinical resistance (K408-G25) and got resensitized to colistin in the consecutive generation (K408-G26). A significant upregulation of ompW, ata, adeFGH genes on K408-G25 was followed by a downregulation upon resensitization to colistin (G26). Despite the upregulation of the ompW gene in transcriptomic analysis, the ompW protein disappeared on K408-G25 and recovered in the resensitized generation (G26). In parallel, disrupted cell membrane integrity recovered in K408-G26. In the K408-G25, downregulation of pbpG and upregulation of pbp1a/pbp3 genes decreased serum-resistance which was reversed in the resensitized generation (G26). The K1007 did not gain colistin resistance amongst 50-generations, however, the generation corresponding to clinical resistance day (K1007-G9) had a similar trend with K408-G25. The clinical colistin-resistant K409 and K1006 had SNPs on pmrA and pmrB genes. CONCLUSION: In this study, we observed that A. baumannii regulates adhesion, efflux pumps and serum-resistance associated genes as an early response to colistin stress. Besides, the ompW protein disappears in the cell membrane of colistin resistant cells which recovers after resensitization to colistin. The lack of ompW protein in colistin-resistant cells should be taken into consideration for escape mutants in development of antivirulence vaccination or treatment options.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Colistin , Colistin/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Humans , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Drug Resistance, Bacterial/genetics , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Adhesion/drug effects , Gene Expression Regulation, Bacterial/drug effects , Carbapenems/pharmacology
9.
Front Cell Infect Microbiol ; 14: 1424038, 2024.
Article in English | MEDLINE | ID: mdl-39165918

ABSTRACT

Introduction: Pseudomonas aeruginosa is a ubiquitous pathogen that causes various infectious diseases through the regulation of quorum sensing (QS). The strategy of interfering with the QS systems of P. aeruginosa, coupled with a reduction in the dosage of conventional antibiotics, presents a potential solution to treating infection and mitigating antibiotic resistance. In this study, seven cinnamoyl hydroxamates were synthesized to evaluate their inhibitory effects on QS of P. aeruginosa. Among these cinnamic acid derivatives, we found cinnamoyl hydroxamic acid (CHA) and 3-methoxy-cinnamoyl hydroxamic acid (MCHA) were the two most effective candidates. Furtherly, the effect of CHA and MCHA on the production of virulence factors and biofilm of P. aeruginosa were evaluated. Ultimately, our study may offer promising potential for treating P. aeruginosa infections and reducing its virulence. Methods: The disc diffusion test were conducted to evaluate inhibitory effects on QS of seven cinnamoyl hydroxamates. The influence of CHA and MCHA on the production of virulence and flagellar motility of P. aeruginosa was furtherly explored. Scanning electron microscopy (SEM) experiment were conducted to evaluate the suppression of CHA and MCHA on the formed biofilm of P. aeruginosa. RT-qPCR was used to detect rhlI, lasA, lasB, rhlA, rhlB, and oprL genes in P. aeruginosa. In silico docking study was performed to explore the molecular mechanism of CHA and MCHA. The synergistic effects of CHA with gentamicin were detected on biofilm cell dispersal. Result: After treatment of CHA or MCHA, the production of multiple virulence factors, including pyocyanin, proteases, rhamnolipid, and siderophore, and swimming and swarming motilities in P. aeruginosa were inhibited significantly. And our results showed CHA and MCHA could eliminate the formed biofilm of P. aeruginosa. RT-qPCR revealed that CHA and MCHA inhibited the expression of QS related genes in P. aeruginosa. Molecular docking indicated that CHA and MCHA primarily inhibited the RhlI/R system in P. aeruginosa by competing with the cognate signaling molecule C4-HSL.Additionally, CHA exhibited potent synergistic effects with gentamicin on biofilm cell dispersal. Discussion: P. aeruginosa is one of the most clinically and epidemiologically important bacteria and a primary cause of catheter-related urinary tract infections and ventilator-associated pneumonia. This study aims to explore whether cinnamoyl hydroxamates have inhibitory effects on QS. And our results indicate that CHA and MCHA, as two novel QSIs, offer promising potential for treating P. aeruginosa infections and reducing its virulence.


Subject(s)
Anti-Bacterial Agents , Biofilms , Cinnamates , Hydroxamic Acids , Molecular Docking Simulation , Pseudomonas aeruginosa , Quorum Sensing , Virulence Factors , Quorum Sensing/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Biofilms/drug effects , Virulence Factors/metabolism , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Cinnamates/pharmacology , Cinnamates/chemistry , Hydroxamic Acids/pharmacology , Hydroxamic Acids/chemistry , Microbial Sensitivity Tests , Virulence/drug effects , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/drug effects
10.
PLoS Biol ; 22(8): e3002741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39146240

ABSTRACT

Clostridioides difficile is an important human pathogen, for which there are very limited treatment options, primarily the glycopeptide antibiotic vancomycin. In recent years, vancomycin resistance has emerged as a serious problem in several gram-positive pathogens, but high-level resistance has yet to be reported for C. difficile, although it is not known if this is due to constraints upon resistance evolution in this species. Here, we show that resistance to vancomycin can evolve rapidly under ramping selection but is accompanied by fitness costs and pleiotropic trade-offs, including sporulation defects that would be expected to severely impact transmission. We identified 2 distinct pathways to resistance, both of which are predicted to result in changes to the muropeptide terminal D-Ala-D-Ala that is the primary target of vancomycin. One of these pathways involves a previously uncharacterised D,D-carboxypeptidase, expression of which is controlled by a dedicated two-component signal transduction system. Our findings suggest that while C. difficile is capable of evolving high-level vancomycin resistance, this outcome may be limited clinically due to pleiotropic effects on key pathogenicity traits. Moreover, our data identify potential mutational routes to resistance that should be considered in genomic surveillance.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Vancomycin Resistance , Vancomycin , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Genetic Fitness , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Signal Transduction , Mutation , Gene Expression Regulation, Bacterial/drug effects , Spores, Bacterial/drug effects , Spores, Bacterial/genetics
11.
BMC Microbiol ; 24(1): 290, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095741

ABSTRACT

INTRODUCTION: Hormesis describes an inverse dose-response relationship, whereby a high dose of a toxic compound is inhibitory, and a low dose is stimulatory. This study explores the hormetic response of low concentrations of zinc oxide nanoparticles (ZnO NPs) toward Pseudomonas aeruginosa. METHOD: Samples of P. aeruginosa, i.e. the reference strain, ATCC 27,853, together with six strains recovered from patients with cystic fibrosis, were exposed to ten decreasing ZnO NPs doses (0.78-400 µg/mL). The ZnO NPs were manufactured from Peganum harmala using a chemical green synthesis approach, and their properties were verified utilizing X-ray diffraction and scanning electron microscopy. A microtiter plate technique was employed to investigate the impact of ZnO NPs on the growth, biofilm formation and metabolic activity of P. aeruginosa. Real-time polymerase chain reactions were performed to determine the effect of ZnO NPs on the expression of seven biofilm-encoding genes. RESULT: The ZnO NPs demonstrated concentration-dependent bactericidal and antibiofilm efficiency at concentrations of 100-400 µg/mL. However, growth was significantly stimulated at ZnO NPs concentration of 25 µg/mL (ATCC 27853, Pa 3 and Pa 4) and at 12.5 µg/mL and 6.25 µg/mL (ATCC 27853, Pa 2, Pa 4 and Pa 5). No significant positive growth was detected at dilutions < 6.25 µg/mL. similarly, biofilm formation was stimulated at concentration of 12.5 µg/mL (ATCC 27853 and Pa 1) and at 6.25 µg/mL (Pa 4). At concentration of 12.5 µg/mL, ZnO NPs upregulated the expression of LasB ( ATCC 27853, Pa 1 and Pa 4) and LasR and LasI (ATCC 27853 and Pa 1) as well as RhII expression (ATCC 27853, Pa 2 and Pa 4). CONCLUSION: When exposed to low ZnO NPs concentrations, P. aeruginosa behaves in a hormetic manner, undergoing positive growth and biofilm formation. These results highlight the importance of understanding the response of P. aeruginosa following exposure to low ZnO NPs concentrations.


Subject(s)
Anti-Bacterial Agents , Biofilms , Hormesis , Pseudomonas aeruginosa , Zinc Oxide , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/growth & development , Zinc Oxide/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Anti-Bacterial Agents/pharmacology , Hormesis/drug effects , Humans , Metal Nanoparticles/chemistry , Nanoparticles/chemistry , Cystic Fibrosis/microbiology , Gene Expression Regulation, Bacterial/drug effects , X-Ray Diffraction , Pseudomonas Infections/microbiology , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Dose-Response Relationship, Drug
12.
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
13.
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
14.
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
15.
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
16.
J Photochem Photobiol B ; 258: 112967, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996773

ABSTRACT

Antimicrobial blue light (aBL) is utilized as a new approach to inhibit the growth of Staphylococcus aureus (S. aureus). Mediated by the endogenous chromophore, aBL possesses the similar photokilling property with aPDI (antimicrobial photodynamic inactivation), however, their mechanistic discrepancies in triggering the death of staphylococcal cells are not yet understood. Here, we describe the use of a 460-nm-LED to curb the viability of S. aureus. According to the results, the bacterial survival was sharply decreased when blue light was applied, reaching a maximum of 4.11 ± 0.04 log10 units. Moreover, the membrane integrity was damaged by aBL, causing the leakage of intracellular DNA. Transcriptomic analysis indicates the divergent gene expression upon either aBL or aPDI, with pathways such as transport, DNA repair, expression regulation and porphyrin massively affected by aBL. Among the commonly regulated genes, LrgA was underpinned on account of its involvement with biofilm formation and protein transport. By comparing the wildtype with the LrgA-overexpressing (LrgA+) strain, the survival rate, membrane penetration, surface structure and biofilm formation were, to a varying degree, improved for LrgA+, which may suggest that LrgA plays essential roles in modulating the responsiveness of S. aureus. Besides, LrgA may function through regulating the expression of autolysis-related systems. Finally, LrgA overexpression did not attenuate but aggravate the impairment induced by aPDI, showcasing a distinct responsive strategy from aBL. Taken together, this study unveils a unique molecular alteration for the aBL-mediated inactivation, providing the basis of utilizing blue light to reduce the harm brought by S. aureus.


Subject(s)
Bacterial Proteins , Biofilms , Blue Light , Staphylococcus aureus , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/drug effects , Biofilms/radiation effects , Gene Expression Regulation, Bacterial/radiation effects , Gene Expression Regulation, Bacterial/drug effects , Microbial Viability/radiation effects , Microbial Viability/drug effects , Porphyrins/chemistry , Porphyrins/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/radiation effects , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology , Transcription, Genetic/radiation effects , Transcription, Genetic/drug effects
17.
Sci Rep ; 14(1): 16483, 2024 07 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
18.
Antimicrob Agents Chemother ; 68(8): e0063624, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39028191

ABSTRACT

In this study, we showed that phenazine-1 carboxylic acid (PCA) of Pseudomonas aeruginosa induced the expression of Tet38 efflux pump triggering Staphylococcus aureus resistance to tetracycline and phenazines. Exposure of S. aureus RN6390 to supernatants of P. aeruginosa PA14 and its pyocyanin (PYO)-deficient mutants showed that P. aeruginosa non-PYO phenazines could induce the expression of Tet38 efflux pump. Direct exposure of RN6390 to PCA compound at 0.25× MIC led to a five-fold increase in tet38 transcripts. Expression of Tet38 protein was identified through confocal microscopy using RN6390(pRN-tet38p-yfp) that expressed YFP under control of the tet38 promoter by PCA at 0.25× MIC. The MICs of PCA of a Tet38-overexpressor and a Δtet38 mutant showed a three-fold increase and a two-fold decrease, respectively, compared with that of wild-type. Pre-exposure of RN6390 to PCA (0.25× MIC) for 1 hour prior to addition of tetracycline (1× or 10× MIC) improved bacteria viability of 1.5-fold and 2.6-fold, respectively, but addition of NaCl 7% together with tetracycline at 10× MIC reduced the number of viable PCA-exposed RN6390 of a 2.0-log10 CFU/mL. The transcript levels of tetR21, a repressor of tet38, decreased and increased two-fold in the presence of PCA and NaCl, respectively, suggesting that the effects of PCA and NaCl on tet38 production occurred through TetR21 expression. These data suggest that PCA-induced Tet38 protects S. aureus against tetracycline during coinfection with P. aeruginosa; however, induced tet38-mediated S. aureus resistance to tetracycline is reversed by NaCl 7%, a nebulized treatment used to enhance sputum mobilization in CF patients.


Subject(s)
Anti-Bacterial Agents , Microbial Sensitivity Tests , Phenazines , Pseudomonas aeruginosa , Staphylococcus aureus , Phenazines/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Tetracycline/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Gene Expression Regulation, Bacterial/drug effects , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism
19.
Antimicrob Agents Chemother ; 68(8): e0026124, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39037241

ABSTRACT

Efflux of antibiotics is an important survival strategy in bacteria. Mycobacterium tuberculosis has approximately sixty efflux pumps, but little is known about the role of each pump or the substrates they efflux. The putative efflux pump, EfpA, is a member of the major facilitator superfamily and has been shown to be essential by saturation transposon mutagenesis studies. It has been implicated in the efflux of isoniazid (INH), which is a first-line drug used to treat tuberculosis (TB). This is supported by evidence from transcriptional profiling showing that efpA is induced in response to INH exposure. However, its roles in the physiology and adaptation of M. tuberculosis to antibiotics have yet to be determined. In this study, we describe the repression of efpA in M. tuberculosis, using CRISPR interference (CRISPRi) to knockdown the expression of this essential gene and the direct effect of this on the ability of M. tuberculosis to survive exposure to INH over a 45-day time course. We determined that wild-type levels of efpA were required for recovery of M. tuberculosis following INH exposure and that, after 45 days of INH exposure, only a few viable colonies were recoverable from efpA-repressed M. tuberculosis. We conclude that EfpA is required for recovery of M. tuberculosis following INH exposure, which could reduce the efficacy of INH in vivo, and that EfpA may have a role in the development of resistance during drug therapy.


Subject(s)
Antitubercular Agents , Bacterial Proteins , Isoniazid , Mycobacterium tuberculosis , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbial Sensitivity Tests , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects
20.
Int J Biol Macromol ; 277(Pt 1): 133934, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39025183

ABSTRACT

This study has explored the involvement of Intrinsically Disordered Proteins (IDPs) in cyanobacterial stress response. IDPs possess distinct physicochemical properties, which allow them to execute diverse functions. Anabaena PCC 7120, the model photosynthetic, nitrogen-fixing cyanobacterium encodes 688 proteins (11 % of the total proteome) with at least one intrinsically disordered region (IDR). Of these, 130 proteins that showed >30 % overall disorder were designated as IDPs. Physico-chemical analysis, showed these IDPs to adopt shapes ranging from 'globular' to 'tadpole-like'. Upon exposure to NaCl, 41 IDP-encoding genes were found to be differentially expressed. Surprisingly, most of these were induced, indicating the importance of IDP-accumulation in overcoming salt stress. Subsequently, six IDPs were identified to be induced by multiple stresses (salt, ammonium and selenite). Interestingly, the presence of these 6-multiple stress-induced IDPs was conserved in filamentous cyanobacteria. Utilizing the experimental proteomic data of Anabaena, these 6 IDPs were found to interact with many proteins involved in diverse pathways, underscoring their physiological importance as protein hubs. This study lays the framework for IDP-related research in Anabaena by (a) identifying, as well as physiochemically characterizing, all the disordered proteins and (b) uncovering a subset of IDPs that are likely to be critical in adaptation to environmental stresses.


Subject(s)
Anabaena , Bacterial Proteins , Intrinsically Disordered Proteins , Stress, Physiological , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Anabaena/metabolism , Anabaena/genetics , Proteomics/methods , Cyanobacteria/metabolism , Gene Expression Regulation, Bacterial/drug effects , Proteome/metabolism , Sodium Chloride/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL