Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 638
Filtrar
1.
J Photochem Photobiol B ; 234: 112547, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36030693

RESUMO

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


Assuntos
Enterococcus faecalis , Estresse Oxidativo , Fotoquimioterapia , Vancomicina , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidade , Testes de Sensibilidade Microbiana , Estresse Oxidativo/fisiologia , Fator sigma/metabolismo , Vancomicina/farmacologia , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
2.
Methods Mol Biol ; 2427: 177-183, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35619034

RESUMO

The Enterococci, mainly Enterococcus faecalis and E. faecium, are ubiquitous members of the human gastrointestinal tract consortia but also a leading cause of opportunistic infections. The global rise in human-associated enterococcal infections, often caused by multidrug resistant strains, highlights an urgent need to identify the bacterial factors contributing to its pathogenicity such that new therapies can be devised. The use of the Galleria mellonella (greater wax moth) larvae, commonly known as wax worm, as a model to study host-pathogen interactions has allowed the identification and characterization of numerous bacterial factors that contribute to disease in humans, serving both as an alternative and complementary approach to mammalian models. Here, we describe the methods for using G. mellonella to characterize the virulence factors of E. faecalis.


Assuntos
Enterococcus faecalis , Mariposas , Animais , Modelos Animais de Doenças , Enterococcus faecalis/patogenicidade , Larva/microbiologia , Mariposas/microbiologia , Virulência , Fatores de Virulência
3.
Infect Immun ; 89(11): e0036521, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34424750

RESUMO

Second messenger nucleotides are produced by bacteria in response to environmental stimuli and play a major role in the regulation of processes associated with bacterial fitness, including but not limited to osmoregulation, envelope homeostasis, central metabolism, and biofilm formation. In this study, we uncovered the biological significance of c-di-AMP in the opportunistic pathogen Enterococcus faecalis by isolating and characterizing strains lacking genes responsible for c-di-AMP synthesis (cdaA) and degradation (dhhP and gdpP). Using complementary approaches, we demonstrated that either complete loss of c-di-AMP (ΔcdaA strain) or c-di-AMP accumulation (ΔdhhP, ΔgdpP, and ΔdhhP ΔgdpP strains) drastically impaired general cell fitness and virulence of E. faecalis. In particular, the ΔcdaA strain was highly sensitive to envelope-targeting antibiotics, was unable to multiply and quickly lost viability in human serum or urine ex vivo, and was virtually avirulent in an invertebrate (Galleria mellonella) and in two catheter-associated mouse infection models that recapitulate key aspects of enterococcal infections in humans. In addition to evidence linking these phenotypes to altered activity of metabolite and peptide transporters and inability to maintain osmobalance, we found that the attenuated virulence of the ΔcdaA strain also could be attributed to a defect in Ebp pilus production and activity that severely impaired biofilm formation under both in vitro and in vivo conditions. Collectively, these results demonstrate that c-di-AMP signaling is essential for E. faecalis pathogenesis and a desirable target for drug development.


Assuntos
Fosfatos de Dinucleosídeos/fisiologia , Enterococcus faecalis/patogenicidade , Animais , Biofilmes , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/crescimento & desenvolvimento , Fímbrias Bacterianas/fisiologia , Regulação Bacteriana da Expressão Gênica , Infecções por Bactérias Gram-Positivas/etiologia , Humanos , Virulência
4.
Infect Immun ; 89(11): e0034321, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34424752

RESUMO

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


Assuntos
Enterococcus faecalis/metabolismo , Glicosídeo Hidrolases/fisiologia , Proteínas de Bactérias/fisiologia , Biofilmes , Domínio Catalítico , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidade , Regulação Bacteriana da Expressão Gênica , Manose/metabolismo , Nutrientes/metabolismo , Polissacarídeos/metabolismo , Ribonucleases/metabolismo , Sítio de Iniciação de Transcrição
5.
Int J Mol Sci ; 22(10)2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-34063332

RESUMO

Artificial sweeteners (AS) are synthetic sugar substitutes that are commonly consumed in the diet. Recent studies have indicated considerable health risks which links the consumption of AS with metabolic derangements and gut microbiota perturbations. Despite these studies, there is still limited data on how AS impacts the commensal microbiota to cause pathogenicity. The present study sought to investigate the role of commonly consumed AS on gut bacterial pathogenicity and gut epithelium-microbiota interactions, using models of microbiota (Escherichia coli NCTC10418 and Enterococcus faecalis ATCC19433) and the intestinal epithelium (Caco-2 cells). Model gut bacteria were exposed to different concentrations of the AS saccharin, sucralose, and aspartame, and their pathogenicity and changes in interactions with Caco-2 cells were measured using in vitro studies. Findings show that sweeteners differentially increase the ability of bacteria to form a biofilm. Co-culture with human intestinal epithelial cells shows an increase in the ability of model gut bacteria to adhere to, invade and kill the host epithelium. The pan-sweet taste inhibitor, zinc sulphate, effectively blocked these negative impacts. Since AS consumption in the diet continues to increase, understanding how this food additive affects gut microbiota and how these damaging effects can be ameliorated is vital.


Assuntos
Enterococcus faecalis/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Edulcorantes/farmacologia , Aspartame/administração & dosagem , Aspartame/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Células CACO-2 , Relação Dose-Resposta a Droga , Enterococcus faecalis/patogenicidade , Escherichia coli/patogenicidade , Microbioma Gastrointestinal/fisiologia , Hemólise/efeitos dos fármacos , Humanos , Sacarina/administração & dosagem , Sacarina/farmacologia , Sacarose/administração & dosagem , Sacarose/análogos & derivados , Sacarose/farmacologia , Edulcorantes/administração & dosagem
6.
Sci Rep ; 11(1): 12577, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131273

RESUMO

The purpose of this study was to design silver nanoparticles (AgNPs) poloxamer thermoreversible gel (AgNPs-PL) and investigate whether this gel could provide sustained antibacterial activity against Enterococcus faecalis (E. faecalis) in the root canal. The gels fabricated were characterized in terms of gelatin temperature, particle size, in-vitro Ag+ release, and elemental content. Cytotoxicity of AgNPs-PL on primary human periodontal ligament fibroblasts (HPDLFs) was examined by CCK-8 assay. Characterization of AgNPs-PL gel revealed that it contained particles existing as large clumps/fused aggregates of different shapes, with a mean diameter of 21.624 ± 14.689 nm, exhibited sustained release of Ag+ for 9 days, and non-toxic to HPDLFs at a low dose (4-32 µg/mL) through 24, 48, and 72 h exposures. The antibacterial effect of 16 and 32 µg/mL concentrations of AgNPs-PL was compared with blank poloxamer gel (PL) and calcium hydroxide (CH) using three methods: (I) agar counting plate, (II) scanning electron microscope (SEM) observations, and (III) confocal laser scanning microscope (CLSM) analysis. AgNPs-PL at the two doses above was more effective than PL and CH in removing E. faecalis biofilm at 1, 3, 9 days. Thus, AgNPs-PL exhibits strong activity against E. faecalis and is easy to produce, with a continuous release profile of Ag+. AgNPs-PL gel may be a candidate for a new root canal disinfection.


Assuntos
Biofilmes/efeitos dos fármacos , Nanopartículas Metálicas/química , Poloxâmero/química , Irrigantes do Canal Radicular/administração & dosagem , Antibacterianos/química , Antibacterianos/farmacologia , Hidróxido de Cálcio/química , Cavidade Pulpar/efeitos dos fármacos , Cavidade Pulpar/microbiologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/patogenicidade , Feminino , Humanos , Masculino , Poloxâmero/administração & dosagem , Irrigantes do Canal Radicular/química , Prata/química
7.
Elife ; 102021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34032213

RESUMO

Upon exposure to harmful microorganisms, hosts engage in protective molecular and behavioral immune responses, both of which are ultimately regulated by the nervous system. Using the nematode Caenorhabditis elegans, we show that ingestion of Enterococcus faecalis leads to a fast pathogen avoidance behavior that results in aversive learning. We have identified multiple sensory mechanisms involved in the regulation of avoidance of E. faecalis. The G-protein coupled receptor NPR-1-dependent oxygen-sensing pathway opposes this avoidance behavior, while an ASE neuron-dependent pathway and an AWB and AWC neuron-dependent pathway are directly required for avoidance. Colonization of the anterior part of the intestine by E. faecalis leads to AWB and AWC mediated olfactory aversive learning. Finally, two transient receptor potential melastatin (TRPM) channels, GON-2 and GTL-2, mediate this newly described rapid pathogen avoidance. These results suggest a mechanism by which TRPM channels may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance and aversive learning by detecting changes in host physiology.


Assuntos
Aprendizagem da Esquiva , Caenorhabditis elegans/microbiologia , Enterococcus faecalis/patogenicidade , Intestinos/microbiologia , Canais de Cátion TRPM/fisiologia , Animais , Enterococcus faecalis/isolamento & purificação , Interações Hospedeiro-Patógeno , Intestinos/patologia , Neurônios/metabolismo , Virulência
8.
G3 (Bethesda) ; 11(2)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33609366

RESUMO

The nematode Caenorhabditis elegans is commonly used as a model organism in studies of the host immune response. The worm encodes twelve peroxidase-cyclooxygenase superfamily members, making it an attractive model in which to study the functions of heme peroxidases. In previous work, loss of one of these peroxidases, SKPO-1 (ShkT-containing peroxidase), rendered C. elegans more sensitive to the human, Gram-positive pathogen Enterococcus faecalis. SKPO-1 was localized to the hypodermis of the animals where it also affected cuticle development as indicated by a morphological phenotype called "dumpy." In this work, a better understanding of how loss of skpo-1 impacts both sensitivity to pathogen as well as cuticle development was sought by subjecting a deletion mutant of skpo-1 to transcriptome analysis using RNA sequencing following exposure to control (Escherichia coli) and pathogenic (E. faecalis) feeding conditions. Loss of skpo-1 caused a general upregulation of genes encoding collagens and other proteins related to cuticle development. On E. faecalis, these animals also failed to upregulate guanylyl cyclases that are often involved in environmental sensing. Hoechst straining revealed increased permeability of the cuticle and atomic force microscopy exposed the misalignment of the cuticular annuli and furrows. These findings provide a basis for better understanding of the morphological as well as the pathogen sensitivity phenotypes associated with loss of SKPO-1 function.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Enterococcus faecalis/patogenicidade , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Perfilação da Expressão Gênica , Heme , Peroxidase
9.
Molecules ; 26(3)2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-33573147

RESUMO

To determine the antibacterial effect of propolis nanoparticles (PNs) as an endodontic irrigant against Enterococcus faecalis biofilm inside the endodontic root canal system. Two-hundred-ten extracted human teeth were sectioned to obtain 6 mm of the middle third of the root. The root canal was enlarged to an internal diameter of 0.9 mm. The specimens were inoculated with E. faecalis for 21 days. Following this, specimens were randomly divided into seven groups, with 30 dentinal blocks in each group including: group I-saline; group II-propolis 100 µg/mL; group III-propolis 300 µg/mL; group IV-propolis nanoparticle 100 µg/mL; group V-propolis nanoparticle 300µg/mL; group VI-6% sodium hypochlorite; group VII-2% chlorhexidine. Dentin shavings were collected at 200 and 400 µm depths, and total numbers of CFUs were determined at the end of one, five, and ten minutes. The non-parametric Kruskal-Wallis and Mann-Whitney tests were used to compare the differences in reduction in CFUs between all groups, and probability values of p < 0.05 were set as the reference for statistically significant results. The antibacterial effect of PNs as an endodontic irrigant was also assessed against E. faecalis isolates from patients with failed root canal treatment. Scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM) were also performed after exposure to PNs. A Raman spectroscope, equipped with a Leica microscope and lenses with curve-fitting Raman software, was used for analysis. The molecular interactions between bioactive compounds of propolis (Pinocembrin, Kaempferol, and Quercetin) and the proteins Sortase A and ß-galactosidase were also understood by computational molecular docking studies. PN300 was significantly more effective in reducing CFUs compared to all other groups (p < 0.05) except 6% NaOCl and 2% CHX (p > 0.05) at all time intervals and both depths. At five minutes, 6% NaOCl and 2% CHX were the most effective in reducing CFUs (p < 0.05). However, no significant difference was found between PN300, 6% NaOCl, and 2% CHX at 10 min (p > 0.05). SEM images also showed the maximum reduction in E. faecalis with PN300, 6% NaOCl, and 2% CHX at five and ten minutes. CLSM images showed the number of dead cells in dentin were highest with PN300 compared to PN100 and saline. There was a reduction in the 484 cm-1 band and an increase in the 870 cm-1 band in the PN300 group. The detailed observations of the docking poses of bioactive compounds and their interactions with key residues of the binding site in all the three docking protocols revealed that the interactions were consistent with reasonable docking and IFD docking scores. PN300 was equally as effective as 6% NaOCl and 2% CHX in reducing the E. faecalis biofilms.


Assuntos
Antibacterianos/administração & dosagem , Enterococcus faecalis/efeitos dos fármacos , Nanopartículas/química , Própole/administração & dosagem , Adulto , Antibacterianos/química , Biofilmes/efeitos dos fármacos , Cavidade Pulpar/efeitos dos fármacos , Cavidade Pulpar/microbiologia , Dentina/microbiologia , Enterococcus faecalis/patogenicidade , Feminino , Humanos , Masculino , Microscopia Confocal , Simulação de Acoplamento Molecular , Própole/química
10.
Sci Rep ; 11(1): 3795, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589735

RESUMO

Nosocomial infections caused by enterococci are an ongoing global threat. Thus, finding therapeutic agents for the treatment of such infections are crucial. Some Enterococcus faecalis strains are able to produce antimicrobial peptides called bacteriocins. We analyzed 65 E. faecalis isolates from 43 food samples and 22 clinical samples in Egypt for 17 common bacteriocin-encoding genes of Enterococcus spp. These genes were absent in 11 isolates that showed antimicrobial activity putatively due to bacteriocins (three from food, including isolate OS13, and eight from clinical isolates). The food-isolated E. faecalis OS13 produced bacteriocin-like inhibitory substances (BLIS) named enterocin OS13, which comprised two peptides (enterocin OS13α OS13ß) that inhibited the growth of antibiotic-resistant nosocomial E. faecalis and E. faecium isolates. The molecular weights of enterocin OS13α and OS13ß were determined as 8079 Da and 7859 Da, respectively, and both were heat-labile. Enterocin OS13α was sensitive to proteinase K, while enterocin OS13ß was resistant. Characterization of E. faecalis OS13 isolate revealed that it belonged to sequence type 116. It was non-hemolytic, bile salt hydrolase-negative, gelatinase-positive, and sensitive to ampicillin, penicillin, vancomycin, erythromycin, kanamycin, and gentamicin. In conclusion, BLIS as enterocin OS13α and OS13ß represent antimicrobial agents with activities against antibiotic-resistant enterococcal isolates.


Assuntos
Bacteriocinas/farmacologia , Infecção Hospitalar/tratamento farmacológico , Farmacorresistência Bacteriana/efeitos dos fármacos , Enterococcus faecalis/química , Bacteriocinas/química , Bacteriocinas/isolamento & purificação , Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana/genética , Egito , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidade , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/patogenicidade , Microbiologia de Alimentos , Humanos , Testes de Sensibilidade Microbiana
11.
Future Microbiol ; 16(3): 175-184, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33528277

RESUMO

Aim:Enterococcus faecalis is a leading nosocomial pathogen in biofilm-associated polymicrobial infections. The study aims to understand pathogenicity and biofilm determinants of the pathogen by genome analysis. Methodology: Genome sequencing of a strong biofilm forming clinical isolate Enterococcus faecalis SK460 devoid of Fsr quorum-signaling system, was performed and comparative genomics was carried out among a set of pathogenic biofilm formers and nonpathogenic weak biofilm formers. Results: Analysis revealed a pool of virulence and adhesion related factors associated with pathogenicity. Absence of CRISPR-Cas system facilitated acquisition of pheromone responsive plasmid, pathogenicity island and phages. Comprehensive analysis identified a subset of accessory genes encoding polysaccharide lyase, sugar phosphotransferase system, phage proteins and transcriptional regulators exclusively in pathogenic biofilm formers. Conclusion: The study identified a set of genes specific to pathogenic biofilm formers and these can act as targets which in turn help to develop future treatment endeavors against enterococcal infections.


Assuntos
Proteínas de Bactérias/genética , Biofilmes , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidade , Proteínas de Bactérias/metabolismo , Sistemas CRISPR-Cas , Enterococcus faecalis/fisiologia , Genoma Bacteriano , Ilhas Genômicas , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Plasmídeos/genética , Plasmídeos/metabolismo , Virulência
12.
PLoS Genet ; 17(1): e1009204, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411815

RESUMO

Bacteriophages (phages) are being considered as alternative therapeutics for the treatment of multidrug resistant bacterial infections. Considering phages have narrow host-ranges, it is generally accepted that therapeutic phages will have a marginal impact on non-target bacteria. We have discovered that lytic phage infection induces transcription of type VIIb secretion system (T7SS) genes in the pathobiont Enterococcus faecalis. Membrane damage during phage infection induces T7SS gene expression resulting in cell contact dependent antagonism of different Gram positive bystander bacteria. Deletion of essB, a T7SS structural component, abrogates phage-mediated killing of bystanders. A predicted immunity gene confers protection against T7SS mediated inhibition, and disruption of its upstream LXG toxin gene rescues growth of E. faecalis and Staphylococcus aureus bystanders. Phage induction of T7SS gene expression and bystander inhibition requires IreK, a serine/threonine kinase, and OG1RF_11099, a predicted GntR-family transcription factor. Additionally, sub-lethal doses of membrane targeting and DNA damaging antibiotics activated T7SS expression independent of phage infection, triggering T7SS antibacterial activity against bystander bacteria. Our findings highlight how phage infection and antibiotic exposure of a target bacterium can affect non-target bystander bacteria and implies that therapies beyond antibiotics, such as phage therapy, could impose collateral damage to polymicrobial communities.


Assuntos
Bacteriófagos/genética , Farmacorresistência Bacteriana Múltipla/genética , Enterococcus faecalis/genética , Infecções por Bactérias Gram-Positivas/genética , Antibacterianos/efeitos adversos , Efeito Espectador , Enterococcus faecalis/patogenicidade , Enterococcus faecalis/virologia , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/terapia , Infecções por Bactérias Gram-Positivas/virologia , Especificidade de Hospedeiro/genética , Humanos , Sistemas de Secreção Tipo VII/genética
13.
mSphere ; 6(1)2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33408224

RESUMO

Traditionally, treatments for bacterial infection have focused on killing the microbe or preventing its growth. As antimicrobial resistance becomes more ubiquitous, the feasibility of this approach is beginning to wane and attention has begun to shift toward disrupting the host-pathogen interaction by improving the host defense. Using a high-throughput, fragment-based screen to identify compounds that alleviate Pseudomonas aeruginosa-mediated killing of Caenorhabditis elegans, we identified over 20 compounds that stimulated host defense gene expression. Five of these molecules were selected for further characterization. Four of five compounds showed little toxicity against mammalian cells or worms, consistent with their identification in a phenotypic, high-content screen. Each of the compounds activated several host defense pathways, but the pathways were generally dispensable for compound-mediated rescue in liquid killing, suggesting redundancy or that the activation of unknown pathway(s) may be driving compound effects. A genetic mechanism was identified for LK56, which required the Mediator subunit MDT-15/MED15 and NHR-49/HNF4 for its function. Interestingly, LK32, LK34, LK38, and LK56 also rescued C. elegans from P. aeruginosa in an agar-based assay, which uses different virulence factors and defense mechanisms. Rescue in an agar-based assay for LK38 entirely depended upon the PMK-1/p38 MAPK pathway. Three compounds-LK32, LK34, and LK56-also conferred resistance to Enterococcus faecalis, and the two lattermost, LK34 and LK56, also reduced pathogenesis from Staphylococcus aureus This study supports a growing role for MDT-15 and NHR-49 in immune response and identifies five molecules that have significant potential for use as tools in the investigation of innate immunity.IMPORTANCE Trends moving in opposite directions (increasing antimicrobial resistance and declining novel antimicrobial development) have precipitated a looming crisis: the nearly complete inability to safely and effectively treat bacterial infections. To avert this, new approaches are needed. One idea is to stimulate host defense pathways to improve the clearance of bacterial infection. Here, we describe five small molecules that promote resistance to infectious bacteria by activating C. elegans' innate immune pathways. Several are effective against both Gram-positive and Gram-negative pathogens. One of the compounds was mapped to the action of MDT-15/MED15 and NHR-49/HNF4, a pair of transcriptional regulators more generally associated with fatty acid metabolism, potentially highlighting a new link between these biological functions. These studies pave the way for future characterization of the anti-infective activity of the molecules in higher organisms and highlight the compounds' potential utility for further investigation of immune modulation as a novel therapeutic approach.


Assuntos
Antibacterianos/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/imunologia , Fatores Imunológicos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/imunologia , Animais , Antibacterianos/análise , Antibacterianos/imunologia , Infecções Bacterianas/tratamento farmacológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/imunologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/patogenicidade , Expressão Gênica , Regulação da Expressão Gênica , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/química , Pseudomonas aeruginosa/patogenicidade , Bibliotecas de Moléculas Pequenas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade , Fatores de Virulência
14.
J Infect Dis ; 223(3): 508-516, 2021 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32597945

RESUMO

Enterococcus  faecalis is a significant cause of hospital-acquired bacteremia. Herein, the discovery is reported that cardiac microlesions form during severe bacteremic E. faecalis infection in mice. The cardiac microlesions were identical in appearance to those formed by Streptococcus pneumoniae during invasive pneumococcal disease. However, E. faecalis does not encode the virulence determinants implicated in pneumococcal microlesion formation. Rather, disulfide bond forming protein A (DsbA) was found to be required for E. faecalis virulence in a Caenorhabditis elegans model and was necessary for efficient cardiac microlesion formation. Furthermore, E. faecalis promoted cardiomyocyte apoptotic and necroptotic cell death at sites of microlesion formation. Additionally, loss of DsbA caused an increase in proinflammatory cytokines, unlike the wild-type strain, which suppressed the immune response. In conclusion, we establish that E. faecalis is capable of forming cardiac microlesions and identify features of both the bacterium and the host response that are mechanistically involved.


Assuntos
Bacteriemia/microbiologia , Bacteriemia/patologia , Enterococcus faecalis/patogenicidade , Cardiopatias/microbiologia , Cardiopatias/patologia , Coração , Animais , Apoptose , Proteínas de Bactérias/metabolismo , Caenorhabditis elegans/microbiologia , Morte Celular , Citocinas , Modelos Animais de Doenças , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/patologia , Camundongos , Necroptose , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/patogenicidade , Tiorredoxinas , Virulência , Fatores de Virulência
15.
Arch Microbiol ; 203(1): 205-217, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32803347

RESUMO

Here, we have analysed and explored the genome sequences of three newly isolated bacteria that were recently characterised for their probiotic activities and ability to produce bacteriocins. These strains, isolated from faeces of animals living in captivity at the zoological garden of Lille (France), are Escherichia coli ICVB443, Enterococcus faecalis ICVB501 and Pediococcus pentosaceus ICVB491. Their genomes have been analysed and compared to those of their pathogenic or probiotic counterparts. The genome analyses of E. coli ICVB443 and Ent. faecalis ICVB501 displayed similarities to those of probiotics E. coli 1917 Nissle, and Ent. faecalis Symbioflor 1, respectively. Furthermore, E. coli ICVB443 shares at least 89 genes with the enteroaggregative E. coli 55989 (EAEC), and Ent. faecalis ICVB501 shares at least 315 genes with the pathogenic Ent. faecalis V583 strain. Unlike Ped. pentosaceus ICVB491, which is devoid of virulence genes, E. coli ICVB443 and Ent. faecalis ICVB501 both carry genes encoding virulence factors on their genomes. Of note, the bioinformatics analysis of these two genomes located the bsh gene, which codes for bile salt hydrolase (BSH). The presence of BSH is of major importance, as it can help to increase the viability of these two strains in the gastrointestinal tract (GIT). The genome analysis of Ped. pentosaceus ICVB491 confirmed its GRAS status (Generally Recognised As Safe), as no genomic virulence factor determinant was found.


Assuntos
Bactérias/genética , Bacteriocinas/genética , Fezes/microbiologia , Genoma Bacteriano/genética , Animais , Bactérias/patogenicidade , Simulação por Computador , Enterococcus faecalis/genética , Enterococcus faecalis/patogenicidade , Escherichia coli/genética , Escherichia coli/patogenicidade , Trato Gastrointestinal/microbiologia , Pediococcus pentosaceus/genética , Pediococcus pentosaceus/patogenicidade , Probióticos , Fatores de Virulência/genética
16.
Sci Rep ; 10(1): 21867, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33318537

RESUMO

The aim of this study was to investigate how carbohydrates (glucose or sucrose) affect the characteristics of Enterococcus faecalis (E. faecalis) planktonic and biofilm in vitro. For this study, E. faecalis was cultured in tryptone-yeast extract broth with 0% glucose + 0% sucrose, 0.5% glucose, 1% glucose, 0.5% sucrose, or 1% sucrose. Viability of E. faecalis was examined by colony forming unit counting assays. Biofilm formation was assessed by measuring extracellular DNA (eDNA), a component of the biofilm matrix. Quantitative real-time PCR (qRT-PCR) was performed to investigate the expression of virulence-associated genes. Field emission scanning electron microscopy analysis, confocal laser scanning microscopy analysis, and crystal violet colorimetric assay were conducted to study E. faecalis biofilms. E. faecalis showed the highest viability and eDNA levels in 1% sucrose medium in biofilms. The result of qRT-PCR showed that the virulence-associated genes expressed highest in 1% sucrose-grown biofilms and in 1% glucose-grown planktonic cultures. E. faecalis showed highly aggregated biofilms and higher bacteria and exopolysaccharide (EPS) bio-volume in sucrose than in 0% glucose + 0% sucrose or glucose. The results indicate that the production of eDNA and EPS and expression of virulence-associated genes in E. faecalis are affected by the concentration of carbohydrates in biofilm or planktonic culture.


Assuntos
Enterococcus faecalis , Matriz Extracelular de Substâncias Poliméricas/metabolismo , Viabilidade Microbiana , Meios de Cultura/química , Meios de Cultura/farmacologia , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/patogenicidade , Virulência
17.
Biomed Res Int ; 2020: 5465439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33150176

RESUMO

INTRODUCTION: Enterococcus faecalis (E. faecalis) is the most important species in dentistry and plays a significant role in the etiology of persistent apical lesions after root canal treatment. Up to date, the intracanal application of 2% chlorhexidine for 7 days is the best way to eliminate E. faecalis. However, due to the ability of this bacterium to persist and survive in harsh environments, many studies have been directed towards finding an alternative strategy for prevention or eradication of it. This study was conducted to investigate the effect of bismuth nanoparticles on E. faecalis, as an etiologic factor in recurrent root canal infections. METHODS: Forty patients, referred to Endodontic Ward of Shiraz University of Medical Science for endodontic pretreatment, provided root canal samples. First, all samples were transferred in Enterococcosel broth and incubated. Then, samples which showed growth were plated on blood agar plates and incubated for further PCR procedure. Nanoparticle powder was dissolved in high-purity water, and the final concentration of bismuth nanoparticles (BiNPs) was measured by the spectrophotometer. Minimum inhibitory concentration (MIC) of BiNPs against E. faecalis was determined by microbroth dilution method according to methods for antimicrobial susceptibility tests. Also, bactericidal assays were conducted in Mueller-Hinton broth medium and reported as the concentration of BiNPs that reduced the viable bacterial count by 99.9%. RESULTS: Of all samples, 77.5% revealed the presence of E. faecalis by PCR. Also, E. faecalis growth inhibition was observed at concentrations ranging from 0.625 µg/ml to 20 µg/ml (geometric mean: 2.337 µg/ml), and the MBC values were between 1.25 µg/ml and 40 µg/ml (geometric mean: 4.781 µg/ml), which in comparison with chlorhexidine, these values were about one-eighth of chlorhexidine. CONCLUSION: The experimental data suggest that bismuth nanoparticles could be an interesting alternative to combat E. faecalis, which, in view of the advantages mentioned for bismuth nanoparticle like inhibiting Streptococcus mutans biofilm formation and higher antibacterial activity compared to chlorhexidine, can be suggested to be used in different fields of dentistry.


Assuntos
Antibacterianos/farmacologia , Bismuto/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Nanopartículas Metálicas/química , Tratamento do Canal Radicular/métodos , Adolescente , Adulto , Clorexidina/farmacologia , Cavidade Pulpar/microbiologia , Cavidade Pulpar/patologia , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/patogenicidade , Feminino , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/patologia , Humanos , Masculino , Nanopartículas Metálicas/ultraestrutura , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade
18.
Biomolecules ; 10(10)2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33092156

RESUMO

Continued mining of natural products from the strain Streptomyces sp. MA37 in our laboratory led to the discovery of a minor specialized metabolite (SM) called accramycin A. Owing to its low yield (0.2 mg/L) in the wild type strain, we investigated the roles of regulatory genes in the corresponding biosynthetic gene cluster (acc BGC) through gene inactivation with the aim of improving the titer of this compound. One of the resulting mutants (∆accJ) dramatically upregulated the production of accramycin A 1 by 330-fold (66 mg/L). Furthermore, ten new metabolites, accramycins B-K 2-11, were discovered, together with two known compounds, naphthacemycin B112 and fasamycin C 13 from the mutant extract. This suggested that accJ, annotated as multiple antibiotic resistance regulator (MarR), is a negative regulator gene in the accramycin biosynthesis. Compounds 1-13 inhibited the Gram-positive pathogens (Staphylococcus aureus, Enterococcus faecalis) and clinical isolates Enterococcus faecium (K59-68 and K60-39) and Staphylococcus haemolyticus with minimal inhibitory concentration (MIC) values in the range of 1.5-12.5 µg/mL. Remarkably, compounds 1-13 displayed superior activity against K60-39 (MIC = 3.1-6.3 µg/mL) compared to ampicillin (MIC = 25 µg/mL), and offered promising potential for the development of accramycin-based antibiotics that target multidrug-resistant Enterococcus clinical isolates. Our results highlight the importance of identifying the roles of regulatory genes in natural product discovery.


Assuntos
Antibacterianos/farmacologia , Produtos Biológicos/farmacologia , Microbiologia do Solo , Streptomyces/química , Antibacterianos/química , Antibacterianos/metabolismo , Produtos Biológicos/química , Produtos Biológicos/metabolismo , Enterococcus/efeitos dos fármacos , Enterococcus/patogenicidade , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/patogenicidade , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade
19.
Int J Biol Macromol ; 165(Pt B): 2964-2973, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33086112

RESUMO

Infections are the leading cause of failure of osteogenic material implantation. Antibiotic treatment, treatment with bone cement, or collagen sponge placement can result in drug resistance and difficulties in operation. To address this, gellan gum (GG) was selected in this study and prepared as an injectable hydrogel containing chlorhexidine (CHX) and nanohydroxyapatite (nHA) that overcomes these intractable problems. Scanning electron microscopy and micro-computed tomography revealed a three-dimensional polymeric network of the hydrogel. The hydrogel had excellent biocompatibility, as detected by cell counting kit-8 and Live/Dead assay. Bone marrow mesenchymal stem cells could be encapsulated into the network, showing that the structure was suitable for cell growth. Additionally, loading the hydrogel with nHA improved its mechanical, biodegradable, and osteogenic properties. Quantitative alkaline phosphatase and Alizarin Red S staining validated its osteogenic ability. Furthermore, antibacterial activity assessment showed that the hydrogel loaded with 50 µg/mL CHX inhibited Enterococcus faecalis in a concentration-dependent manner. Thus, we report an injectable GG-based hydrogel with superior antibacterial effect against E. faecalis and osteogenesis, which holds promise for treating infectious bone defects caused by refractory periradicular periodontitis.


Assuntos
Hidrogéis/química , Osteogênese/efeitos dos fármacos , Polissacarídeos Bacterianos/química , Engenharia Tecidual , Fosfatase Alcalina/química , Animais , Interface Osso-Implante/crescimento & desenvolvimento , Interface Osso-Implante/microbiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Clorexidina/química , Clorexidina/farmacologia , Modelos Animais de Doenças , Durapatita/química , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/patogenicidade , Humanos , Hidrogéis/síntese química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Polissacarídeos Bacterianos/síntese química , Polissacarídeos Bacterianos/farmacologia , Ratos , Tecidos Suporte/química , Microtomografia por Raio-X
20.
J Immunol ; 205(10): 2763-2777, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33055280

RESUMO

Bacterial prostatitis affects 1% of men, with increased incidence in the elderly. Acute bacterial prostatitis frequently progresses to chronicity, marked by recurrent episodes interspersed with asymptomatic periods of variable duration. Antibiotic treatment is standard of care; however, dissemination of antimicrobially resistant uropathogens threatens therapy efficacy. Thus, development of nonantibiotic-based approaches to treat chronic disease is a priority. Currently, why chronic prostatitis arises is unclear, as the immune response to prostate infection is incompletely understood. As 80% of prostatitis cases are caused by Gram-negative uropathogenic Escherichia coli (UPEC) or Gram-positive Enterococcus faecalis, we used a mouse transurethral instillation model to address the hypothesis that an innate immune response fails to develop following prostate infection with these uropathogens, leading to chronic disease. Surprisingly, infection induced robust proinflammatory cytokine expression and myeloid cell infiltration. Following a second infection, cytokine responses and innate cell infiltration were largely comparable to primary infection. Characteristic of memory responses, more lymphoid cells infiltrated the prostate in a second infection compared with a first, suggesting that adaptive immunity develops to eliminate the pathogens. Unexpectedly, bacterial burden in prostates challenged with either UPEC or E. faecalis was equal or greater than primary infection despite that a protective adaptive response to UPEC infection was evident in the bladder of the same animals. Our findings support that chronic or recurrent prostatitis develops despite strong innate immune responses and may be the result of a failure to develop immune memory to infection, pointing to actionable targets for immunotherapy.


Assuntos
Infecções por Escherichia coli/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Prostatite/imunologia , Infecções Urinárias/imunologia , Animais , Doença Crônica , Citocinas/metabolismo , Modelos Animais de Doenças , Enterococcus faecalis/imunologia , Enterococcus faecalis/patogenicidade , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/terapia , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/terapia , Humanos , Imunidade Inata , Memória Imunológica , Imunoterapia , Masculino , Camundongos , Próstata/imunologia , Próstata/microbiologia , Prostatite/microbiologia , Prostatite/terapia , Recidiva , Bexiga Urinária/imunologia , Bexiga Urinária/microbiologia , Infecções Urinárias/microbiologia , Infecções Urinárias/terapia , Escherichia coli Uropatogênica/imunologia , Escherichia coli Uropatogênica/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...