Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 819
Filtrar
1.
Curr Microbiol ; 81(10): 349, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39254872

RESUMO

The treatment of infections caused by biofilm-forming organisms is challenging. The newly discovered antibiotic teixobactin shows activity against a wide range of biofilm-forming bacteria. However, the laborious and low-yield chemical synthesis of teixobactin complicates its further development for clinical application. The use of more easily synthesized teixobactin analogues may offer promise in this regard. In this article, three newly developed analogues were tested for efficacy against Staphylococcus aureus and Enterococcus faecalis. Minimum inhibitory and -bactericidal concentrations were investigated. MIC values for S. aureus and E. faecalis ranged from 0.5-2 and 2-4 µg/mL, respectively. Moreover, the ability of the analogues to prevent biofilm formation and to inactivate bacterial cells in already established S. aureus biofilm on medical grade materials (PVC and PTFE) used in the production of infusion tubing and catheters were also tested. The analogues showed an ability to prevent biofilm formation and inactivate bacterial cells in established biofilms at concentrations as low as 1-2 µg/mL. Confocal laser scanning microscopy showed that the most promising analogue (TB3) inactivated S. aureus cells in a preformed biofilm and gave a reduction in biovolume. The relative ease of synthesis of the analogues and their in vitro efficacy, makes them promising candidates for pharmaceutical development.


Assuntos
Antibacterianos , Biofilmes , Depsipeptídeos , Enterococcus faecalis , Testes de Sensibilidade Microbiana , Staphylococcus aureus , Biofilmes/efeitos dos fármacos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Enterococcus faecalis/crescimento & desenvolvimento , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Antibacterianos/farmacologia , Antibacterianos/química , Depsipeptídeos/farmacologia , Depsipeptídeos/química
2.
Curr Biol ; 34(18): 4129-4142.e4, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39163856

RESUMO

Despite competition for both space and nutrients, bacterial species often coexist within structured, surface-attached communities termed biofilms. While these communities play important, widespread roles in ecosystems and are agents of human infection, understanding how multiple bacterial species assemble to form these communities and what physical processes underpin the composition of multispecies biofilms remains an active area of research. Using a model three-species community composed of Pseudomonas aeruginosa, Escherichia coli, and Enterococcus faecalis, we show with cellular-scale resolution that biased dispersal of the dominant community member, P. aeruginosa, prevents competitive exclusion from occurring, leading to the coexistence of the three species. A P. aeruginosa bqsS deletion mutant no longer undergoes periodic mass dispersal, leading to the local competitive exclusion of E. coli. Introducing periodic, asymmetric dispersal behavior into minimal models, parameterized by only maximal growth rate and local density, supports the intuition that biased dispersal of an otherwise dominant competitor can permit coexistence generally. Colonization experiments show that WT P. aeruginosa is superior at colonizing new areas, in comparison to ΔbqsS P. aeruginosa, but at the cost of decreased local competitive ability against E. coli and E. faecalis. Overall, our experiments document how one species' modulation of a competition-dispersal-colonization trade-off can go on to influence the stability of multispecies coexistence in spatially structured ecosystems.


Assuntos
Biofilmes , Enterococcus faecalis , Escherichia coli , Pseudomonas aeruginosa , Biofilmes/crescimento & desenvolvimento , Pseudomonas aeruginosa/fisiologia , Escherichia coli/fisiologia , Enterococcus faecalis/fisiologia , Modelos Biológicos , Ecossistema , Interações Microbianas
3.
Biointerphases ; 19(4)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39136648

RESUMO

Liquid-infused polymers are recognized for their ability to repel foulants, making them promising for biomedical applications including catheter-associated urinary tract infections (CAUTIs). However, the impact of the quantity of free liquid layer covering the surface on protein and bacterial adhesion is not well understood. Here, we explore how the amount of free silicone liquid layer in infused silicone catheter materials influences the adhesion of bacteria and proteins relevant to CAUTIs. To alter the quantity of the free liquid layer, we either physically removed excess liquid from fully infused catheter materials or partially infused them. We then evaluated the impact on bacterial and host protein adhesion. Physical removal of the free liquid layer from the fully infused samples reduced the height of the liquid layer from 60 µm to below detection limits and silicone liquid loss into the environment by approximately 64% compared to controls, without significantly increasing the deposition of protein fibrinogen or the adhesion of the common uropathogen Enterococcus faecalis. Partially infused samples showed even greater reductions in liquid loss: samples infused to 70%-80% of their maximum capacity exhibited about an 85% decrease in liquid loss compared to fully infused controls. Notably, samples with more than 70% infusion did not show significant increases in fibrinogen or E. faecalis adhesion. These findings suggest that adjusting the levels of the free liquid layer in infused polymers can influence protein and bacterial adhesion on their surfaces. Moreover, removing the free liquid layer can effectively reduce liquid loss from these polymers while maintaining their functionality.


Assuntos
Aderência Bacteriana , Enterococcus faecalis , Aderência Bacteriana/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Enterococcus faecalis/efeitos dos fármacos , Polímeros/química , Silicones/química , Propriedades de Superfície , Fibrinogênio/química , Fibrinogênio/metabolismo , Humanos
4.
Front Cell Infect Microbiol ; 14: 1433313, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39091674

RESUMO

Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.


Assuntos
Biofilmes , Cavidade Pulpar , Enterococcus faecalis , Infecções por Bactérias Gram-Positivas , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Humanos , Cavidade Pulpar/microbiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Antibacterianos/farmacologia
5.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 15-21, 2024 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-39097900

RESUMO

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.


Assuntos
Biofilmes , Enterococcus faecalis , Staphylococcus aureus , Ácido Ursodesoxicólico , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/fisiologia , Humanos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Enterococcus faecalis/fisiologia , Ácido Ursodesoxicólico/farmacologia , Antibacterianos/farmacologia , Doença Crônica , Testes de Sensibilidade Microbiana , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Benzimidazóis/farmacologia , Tetrazóis/farmacologia , Compostos de Bifenilo/farmacologia
6.
Braz J Microbiol ; 55(3): 2119-2130, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38954220

RESUMO

Biosurfactants, sustainable alternatives to petrochemical surfactants, are gaining attention for their potential in medical applications. This study focuses on producing, purifying, and characterizing a glycolipid biosurfactant from Candida sp. UFSJ7A, particularly for its application in biofilm prevention on siliconized latex catheter surfaces. The glycolipid was extracted and characterized, revealing a critical micellar concentration (CMC) of 0.98 mg/mL, indicating its efficiency at low concentrations. Its composition, confirmed through Fourier transform infrared spectroscopy (FT-IR) and thin layer chromatography (TLC), identified it as an anionic biosurfactant with a significant ionic charge of -14.8 mV. This anionic nature contributes to its biofilm prevention capabilities. The glycolipid showed a high emulsification index (E24) for toluene, gasoline, and soy oil and maintained stability under various pH and temperature conditions. Notably, its anti-adhesion activity against biofilms formed by Escherichia coli, Enterococcus faecalis, and Candida albicans was substantial. When siliconized latex catheter surfaces were preconditioned with 2 mg/mL of the glycolipid, biofilm formation was reduced by up to 97% for E. coli and C. albicans and 57% for E. faecalis. These results are particularly significant when compared to the efficacy of conventional surfactants like SDS, especially for E. coli and C. albicans. This study highlights glycolipids' potential as a biotechnological tool in reducing biofilm-associated infections on medical devices, demonstrating their promising applicability in healthcare settings.


Assuntos
Biofilmes , Candida , Catéteres , Glicolipídeos , Tensoativos , Glicolipídeos/farmacologia , Glicolipídeos/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Tensoativos/farmacologia , Tensoativos/química , Candida/efeitos dos fármacos , Candida/fisiologia , Catéteres/microbiologia , Látex/química , Látex/farmacologia , Escherichia coli/efeitos dos fármacos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Candida albicans/efeitos dos fármacos , Candida albicans/fisiologia
7.
J Microbiol ; 62(8): 683-693, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38935316

RESUMO

Enterococcus faecalis is a Gram-positive bacterium that is frequently found in the periapical lesion of patients with apical periodontitis. Its biofilm formation in root canal is closely related to the development of refractory apical periodontitis by providing increased resistance to endodontic treatments. Phage therapy has recently been considered as an efficient therapeutic strategy in controlling various periodontal pathogens. We previously demonstrated the bactericidal capacities of Enterococcus phage vB_EfaS_HEf13 (phage HEf13) against clinically-isolated E. faecalis strains. Here, we investigated whether phage HEf13 affects biofilm formation and pre-formed biofilm of clinically-isolated E. faecalis, and its combinatory effect with endodontic treatments, including chlorhexidine (CHX) and penicillin. The phage HEf13 inhibited biofilm formation and disrupted pre-formed biofilms of E. faecalis in a dose- and time-dependent manner. Interestingly, phage HEf13 destroyed E. faecalis biofilm exopolysaccharide (EPS), which is known to be a major component of bacterial biofilm. Furthermore, combined treatment of phage HEf13 with CHX or penicillin more potently inhibited biofilm formation and disrupted pre-formed biofilm than either treatment alone. Confocal laser scanning microscopic examination demonstrated that these additive effects of the combination treatments on disruption of pre-formed biofilm are mediated by relatively enhanced reduction in thickness distribution and biomass of biofilm. Collectively, our results suggest that the effect of phage HEf13 on E. faecalis biofilm is mediated by its EPS-degrading property, and its combination with endodontic treatments more potently suppresses E. faecalis biofilm, implying that phage HEf13 has potential to be used as a combination therapy against E. faecalis infections.


Assuntos
Bacteriófagos , Biofilmes , Enterococcus faecalis , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Enterococcus faecalis/virologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Bacteriófagos/fisiologia , Humanos , Antibacterianos/farmacologia , Clorexidina/farmacologia , Terapia por Fagos , Infecções por Bactérias Gram-Positivas/microbiologia , Periodontite Periapical/terapia , Periodontite Periapical/microbiologia , Periodontite Periapical/virologia , Polissacarídeos Bacterianos/metabolismo
8.
Microbiol Spectr ; 12(8): e0056524, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38912797

RESUMO

Over 2.5 million prosthetic joint implantation surgeries occur annually in the United States. Periprosthetic joint infections (PJIs), though occurring in only 1-2% of patients receiving replacement joints, are challenging to diagnose and treat and are associated with significant morbidity. The Gram-positive bacterium Enterococcus faecalis, which can be highly antibiotic-resistant and is a robust biofilm producer on indwelling medical devices, accounts for 2-11% of PJIs. E. faecalis PJIs are understudied compared to those caused by other pathogens, such as Staphylococcus aureus. This motivates the need to generate a comprehensive understanding of E. faecalis PJIs to guide future treatments for these infections. To address this, we describe a panel of E. faecalis strains isolated from the surface of prosthetic joints in a cohort of individuals treated at the Mayo Clinic in Rochester, MN. Here, we present the first complete genome assemblage of E. faecalis PJI isolates. Comparative genomics shows differences in genome size, virulence factors, antimicrobial resistance genes, plasmids, and prophages, underscoring the genetic diversity of these strains. These isolates have strain-specific differences in in vitro biofilm biomass, biofilm burden, and biofilm morphology. We measured robust changes in biofilm architecture and aggregation for all isolates when grown in simulated synovial fluid (SSF). Finally, we evaluated the antibiotic efficacy of these isolates and found strain-specific changes across all strains when grown in SSF. Results of this study highlight the existence of genetic and phenotypic heterogeneity among E. faecalis PJI isolates which will provide valuable insight and resources for future E. faecalis PJI research. IMPORTANCE: Periprosthetic joint infections (PJIs) affect ~1-2% of those who undergo joint replacement surgery. Enterococcus faecalis is a Gram-positive opportunistic pathogen that causes ~10% of PJIs in the United States each year, but our understanding of how and why E. faecalis causes PJIs is limited. E. faecalis infections are typically biofilm-associated and can be difficult to clear with antibiotic therapy. Here, we provide complete genomes for four E. faecalis PJI isolates from the Mayo Clinic. These isolates have strain-specific differences in biofilm formation, aggregation, and antibiotic susceptibility in simulated synovial fluid. These results provide important insight into the genomic and phenotypic features of E. faecalis isolates from PJI.


Assuntos
Antibacterianos , Biofilmes , Enterococcus faecalis , Infecções por Bactérias Gram-Positivas , Infecções Relacionadas à Prótese , Enterococcus faecalis/genética , Enterococcus faecalis/isolamento & purificação , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Enterococcus faecalis/classificação , Infecções Relacionadas à Prótese/microbiologia , Biofilmes/crescimento & desenvolvimento , Humanos , Infecções por Bactérias Gram-Positivas/microbiologia , Antibacterianos/farmacologia , Genótipo , Fatores de Virulência/genética , Fenótipo , Testes de Sensibilidade Microbiana , Genoma Bacteriano , Farmacorresistência Bacteriana
9.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38892419

RESUMO

The gut microbiome plays a fundamental role in metabolism, as well as the immune and nervous systems. Microbial imbalance (dysbiosis) can contribute to subsequent physical and mental pathologies. As such, interest has been growing in the microbiota-gut-brain brain axis and the bioelectrical communication that could exist between bacterial and nervous cells. The aim of this study was to investigate the bioelectrical profile (electrome) of two bacterial species characteristic of the gut microbiome: a Proteobacteria Gram-negative bacillus Escherichia coli (E. coli), and a Firmicutes Gram-positive coccus Enterococcus faecalis (E. faecalis). We analyzed both bacterial strains to (i) validate the fluorescent probe bis-(1,3-dibutylbarbituric acid) trimethine oxonol, DiBAC4(3), as a reliable reporter of the changes in membrane potential (Vmem) for both bacteria; (ii) assess the evolution of the bioelectric profile throughout the growth of both strains; (iii) investigate the effects of two neural-type stimuli on Vmem changes: the excitatory neurotransmitter glutamate (Glu) and the inhibitory neurotransmitter γ-aminobutyric acid (GABA); (iv) examine the impact of the bioelectrical changes induced by neurotransmitters on bacterial growth, viability, and cultivability using absorbance, live/dead fluorescent probes, and viable counts, respectively. Our findings reveal distinct bioelectrical profiles characteristic of each bacterial species and growth phase. Importantly, neural-type stimuli induce Vmem changes without affecting bacterial growth, viability, or cultivability, suggesting a specific bioelectrical response in bacterial cells to neurotransmitter cues. These results contribute to understanding the bacterial response to external stimuli, with potential implications for modulating bacterial bioelectricity as a novel therapeutic target.


Assuntos
Eixo Encéfalo-Intestino , Microbioma Gastrointestinal , Eixo Encéfalo-Intestino/fisiologia , Enterococcus faecalis/fisiologia , Escherichia coli , Ácido Glutâmico/metabolismo , Ácido gama-Aminobutírico/metabolismo , Potenciais da Membrana , Humanos
10.
Int J Nanomedicine ; 19: 5879-5893, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38895145

RESUMO

Introduction: Persistent endodontic infections (PEIs) mediated by bacterial biofilm mainly cause persistent periapical inflammation, resulting in recurrent periapical abscesses and progressive bone destruction. However, conventional root canal disinfectants are highly damaging to the tooth and periodontal tissue and ineffective in treating persistent root canal infections. Antimicrobial materials that are biocompatible with apical tissues and can eliminate PEIs-associated bacteria are urgently needed. Methods: Here, ε-poly (L-lysine) derived carbon quantum dots (PL-CQDs) are fabricated using pyrolysis to remove PEIs-associated bacterial biofilms. Results: Due to their ultra-small size, high positive charge, and active reactive oxygen species (ROS) generation capacity, PL-CQDs exhibit highly effective antibacterial activity against Enterococcus faecalis (E. faecalis), which is greatly dependent on PL-CQDs concentrations. 100 µg/mL PL-CQDs could kill E. faecalis in 5 min. Importantly, PL-CQDs effectively achieved a reduction of biofilms in the isolated teeth model, disrupting the dense structure of biofilms. PL-CQDs have acceptable cytocompatibility and hemocompatibility in vitro and good biosafety in vivo. Discussion: Thus, PL-CQDs provide a new strategy for treating E. faecalis-associated PEIs.


Assuntos
Biofilmes , Carbono , Enterococcus faecalis , Infecções por Bactérias Gram-Positivas , Polilisina , Pontos Quânticos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Pontos Quânticos/química , Biofilmes/efeitos dos fármacos , Polilisina/química , Polilisina/farmacologia , Carbono/química , Carbono/farmacologia , Animais , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Antibacterianos/farmacologia , Antibacterianos/química , Humanos , Espécies Reativas de Oxigênio/metabolismo , Camundongos
11.
PLoS One ; 19(5): e0298283, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38809833

RESUMO

Biofilms make it difficult to eradicate bacterial infections through antibiotic treatments and lead to numerous complications. Previously, two periprosthetic infection-related pathogens, Enterococcus faecalis and Staphylococcus lugdunensis were reported to have relatively contrasting biofilm-forming abilities. In this study, we examined the proteomics of the two microorganisms' biofilms using LC-MS/MS. The results showed that each microbe exhibited an overall different profile for differential gene expressions between biofilm and planktonic cells as well as between each other. Of a total of 929 proteins identified in the biofilms of E. faecalis, 870 proteins were shared in biofilm and planktonic cells, and 59 proteins were found only in the biofilm. In S. lugdunensis, a total of 1125 proteins were identified, of which 1072 proteins were found in common in the biofilm and planktonic cells, and 53 proteins were present only in the biofilms. The functional analysis for the proteins identified only in the biofilms using UniProt keywords demonstrated that they were mostly assigned to membrane, transmembrane, and transmembrane helix in both microorganisms, while hydrolase and transferase were found only in E. faecalis. Protein-protein interaction analysis using STRING-db indicated that the resulting networks did not have significantly more interactions than expected. GO term analysis exhibited that the highest number of proteins were assigned to cellular process, catalytic activity, and cellular anatomical entity. KEGG pathway analysis revealed that microbial metabolism in diverse environments was notable for both microorganisms. Taken together, proteomics data discovered in this study present a unique set of biofilm-embedded proteins of each microorganism, providing useful information for diagnostic purposes and the establishment of appropriately tailored treatment strategies. Furthermore, this study has significance in discovering the target candidate molecules to control the biofilm-associated infections of E. faecalis and S. lugdunensis.


Assuntos
Proteínas de Bactérias , Biofilmes , Enterococcus faecalis , Plâncton , Proteômica , Staphylococcus lugdunensis , Biofilmes/crescimento & desenvolvimento , Enterococcus faecalis/fisiologia , Enterococcus faecalis/metabolismo , Enterococcus faecalis/genética , Proteômica/métodos , Staphylococcus lugdunensis/metabolismo , Staphylococcus lugdunensis/genética , Plâncton/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Espectrometria de Massas em Tandem , Cromatografia Líquida
12.
Appl Microbiol Biotechnol ; 108(1): 336, 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38761182

RESUMO

To investigate the cell-cell interactions of intergeneric bacterial species, the study detected the survival of Enterococcus faecalis (Ef) under monospecies or coaggregation state with Fusobacterium nucleatum subsp. polymorphum (Fnp) in environmental stress. Ef and Fnp infected the human macrophages with different forms (Ef and Fnp monospecies, Ef-Fnp coaggregates, Ef + Fnp cocultures) for exploring the immunoregulatory effects and the relevant molecular mechanisms. Meanwhile, the transcriptomic profiles of coaggregated Ef and Fnp were analyzed. Ef was shown to coaggregate with Fnp strongly in CAB within 90 min by forming multiplexes clumps. Coaggregation with Fnp reinforced Ef resistance against unfavorable conditions including alkaline, hypertonic, nutrient-starvation, and antibiotic challenges. Compared with monospecies and coculture species, the coaggregation of Ef and Fnp significantly facilitates both species to invade dTHP-1 cells and aid Ef to survive within the cells. Compared with coculture species, dual-species interaction of Ef and Fnp significantly decreased the levels of pro-inflammatory cytokines IL-6, TNF-α, and chemokines MCP-1 secreted by dTHP-1 cells and lessened the phosphorylation of p38, JNK, and p65 signaling pathways. The transcriptome sequencing results showed that 111 genes were differentially expressed or Ef-Fnp coaggregated species compared to Ef monospecies; 651 genes were differentially expressed for Fnp when coaggregation with Ef. The analysis of KEGG pathway showed that Ef differentially expressed genes (DEGs) were enriched in quorum sensing and arginine biosynthesis pathway; Fnp DEGs were differentially concentrated in lipopolysaccharide (LPS) biosynthesis, biofilm formation, and lysine degradation pathway compared to monospecies. KEY POINTS: • Coaggregated with Fnp aids Ef's survival in environmental stress, especially in root canals after endodontic treatment. • The coaggregation of Ef and Fnp may weaken the pro-inflammatory response and facilitate Ef to evade killed by macrophages. • The coaggregation between Ef and Fnp altered interspecies transcriptional profiles.


Assuntos
Enterococcus faecalis , Fusobacterium nucleatum , Macrófagos , Estresse Fisiológico , Fusobacterium nucleatum/fisiologia , Fusobacterium nucleatum/genética , Enterococcus faecalis/genética , Enterococcus faecalis/fisiologia , Humanos , Macrófagos/microbiologia , Macrófagos/imunologia , Citocinas/metabolismo , Citocinas/genética , Aderência Bacteriana , Técnicas de Cocultura , Perfilação da Expressão Gênica , Transcriptoma , Linhagem Celular , Interleucina-6/genética , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/genética , Inflamação
13.
Elife ; 132024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38767331

RESUMO

Wound infections are highly prevalent and can lead to delayed or failed healing, causing significant morbidity and adverse economic impacts. These infections occur in various contexts, including diabetic foot ulcers, burns, and surgical sites. Enterococcus faecalis is often found in persistent non-healing wounds, but its contribution to chronic wounds remains understudied. To address this, we employed single-cell RNA sequencing (scRNA-seq) on infected wounds in comparison to uninfected wounds in a mouse model. Examining over 23,000 cells, we created a comprehensive single-cell atlas that captures the cellular and transcriptomic landscape of these wounds. Our analysis revealed unique transcriptional and metabolic alterations in infected wounds, elucidating the distinct molecular changes associated with bacterial infection compared to the normal wound healing process. We identified dysregulated keratinocyte and fibroblast transcriptomes in response to infection, jointly contributing to an anti-inflammatory environment. Notably, E. faecalis infection prompted a premature, incomplete epithelial-mesenchymal transition in keratinocytes. Additionally, E. faecalis infection modulated M2-like macrophage polarization by inhibiting pro-inflammatory resolution in vitro, in vivo, and in our scRNA-seq atlas. Furthermore, we discovered macrophage crosstalk with neutrophils, which regulates chemokine signaling pathways, while promoting anti-inflammatory interactions with endothelial cells. Overall, our findings offer new insights into the immunosuppressive role of E. faecalis in wound infections.


If wounds get infected, they heal much more slowly, sometimes leading to skin damage and other complications, including disseminated infections or even amputation. Infections can happen in many types of wounds, ranging from ulcers in patients with diabetes to severe burns. If infections are not cleared quickly, the wounds can become 'chronic' and are unable to heal without intervention. Enterococcus faecalis is a type of bacteria that normally lives in the gut. Within that environment, in healthy people, it is not harmful. However, if it comes into contact with wounds ­ particularly diabetic ulcers or the site of a surgery ­ it can cause persistent infections and prevent healing. Although researchers are beginning to understand how E. faecalis initially colonises wounds, the biological mechanisms that transform these infections into chronic wounds are still largely unknown. Celik et al. therefore set out to investigate exactly how E. faecalis interferes with wound healing. To do this, Celik et al. looked at E. faecalis-infected wounds in mice and compared them to uninfected ones. Using a genetic technique called single-cell RNA sequencing, Celik et al. were able to determine which genes were switched on in individual skin and immune cells at the site of the wounds. This in turn allowed the researchers to determine how those cells were behaving in both infected and uninfected conditions. The experiments revealed that when E. faecalis was present in wounds, several important cell types in the wounds did not behave normally. For example, although the infected skin cells still underwent a change in behaviour required for healing (called an epithelial-mesenchymal transition), the change was both premature and incomplete. In other words, the skin cells in infected wounds started changing too early and did not finish the healing process properly. E. faecalis also changed the way macrophages and neutrophils worked within the wounds. These are cells in our immune system that normally promote inflammation, a process involved in both uninfected wounds or during infections and is a key part of wound healing when properly controlled. In the E. faecalis-infected wounds, these cells' inflammatory properties were suppressed, making them less helpful for healing. These results shed new light on how E. faecalis interacts with skin cells and the immune system to disrupt wound healing. Celik et al. hope that this knowledge will allow us to find new ways to target E. faecalis infections, and ultimately develop treatments to help chronic wounds heal better and faster.


Assuntos
Enterococcus faecalis , Infecções por Bactérias Gram-Positivas , Queratinócitos , Cicatrização , Enterococcus faecalis/fisiologia , Enterococcus faecalis/genética , Animais , Camundongos , Infecções por Bactérias Gram-Positivas/microbiologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Macrófagos/microbiologia , Macrófagos/metabolismo , Macrófagos/imunologia , Modelos Animais de Doenças , Infecção dos Ferimentos/microbiologia , Transcriptoma , Camundongos Endogâmicos C57BL , Análise de Célula Única , Transição Epitelial-Mesenquimal/genética , Masculino , Fibroblastos/microbiologia , Fibroblastos/metabolismo
14.
Pathog Dis ; 822024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38794885

RESUMO

Urinary tract infection (UTI), one of the most common bacterial infections worldwide, is a typical example of an infection that is often polymicrobial in nature. While the overall infection course is known on a macroscale, bacterial behavior is not fully understood at the cellular level and bacterial pathophysiology during multispecies infection is not well characterized. Here, using clinically relevant bacteria, human epithelial bladder cells and human urine, we establish co-infection models combined with high resolution imaging to compare single- and multi-species bladder cell invasion events in three common uropathogens: uropathogenic Escherichia coli (UPEC), Klebsiella pneumoniae and Enterococcus faecalis. While all three species invaded the bladder cells, under flow conditions the Gram-positive E. faecalis was significantly less invasive compared to the Gram-negative UPEC and K. pneumoniae. When introduced simultaneously during an infection experiment, all three bacterial species sometimes invaded the same bladder cell, at differing frequencies suggesting complex interactions between bacterial species and bladder cells. Inside host cells, we observed encasement of E. faecalis colonies specifically by UPEC. During subsequent dispersal from the host cells, only the Gram-negative bacteria underwent infection-related filamentation (IRF). Taken together, our data suggest that bacterial multispecies invasions of single bladder cells are frequent and support earlier studies showing intraspecies cooperation on a biochemical level during UTI.


Assuntos
Enterococcus faecalis , Células Epiteliais , Klebsiella pneumoniae , Infecções Urinárias , Escherichia coli Uropatogênica , Humanos , Infecções Urinárias/microbiologia , Enterococcus faecalis/fisiologia , Células Epiteliais/microbiologia , Escherichia coli Uropatogênica/fisiologia , Klebsiella pneumoniae/fisiologia , Bexiga Urinária/microbiologia , Bexiga Urinária/citologia , Coinfecção/microbiologia , Linhagem Celular , Interações Hospedeiro-Patógeno
15.
Future Microbiol ; 19(8): 647-654, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38661711

RESUMO

Aim: We evaluated Diacetylcurcumin (DAC), a derivative of curcumin, for its antibacterial and antibiofilm properties against Enterococcus faecalis. Methods: Minimum inhibitory concentration (MIC) and minimum bactericidal concentration were determined, along with antibiofilm potential and toxicity in Galleria mellonella. Additionally, in silico computational analysis was performed to understand its mechanisms of action. Results & conclusion: DAC demonstrated significant antibacterial effects, with MIC and MBC values of 15.6 and 31.25 µg/ml, respectively, and reduced biofilm formation. A synergistic effect, reducing biofilm by 77%, was observed when combined with calcium hydroxide. G. mellonella toxicity tests confirmed DAC's safety at tested concentrations, suggesting its potential for use in root canal disinfection products.


Diacetylcurcumin (DAC) comes from turmeric, a natural spice often used in food. DAC may have the ability to fight germs, including the bacteria Enterococcus faecalis. We tested DAC's ability to kill E. faecalis and stopping the formation of films of the bacteria. We found that a small amount of DAC did kill E. faecalis. When used with calcium hydroxide, DAC works even better to reduce the formation of bacterial films by 77%. DAC is safe to be used on teeth, so may be a useful ingredient for preserving mouth health.


Assuntos
Antibacterianos , Biofilmes , Curcumina , Enterococcus faecalis , Testes de Sensibilidade Microbiana , Mariposas , Biofilmes/efeitos dos fármacos , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Curcumina/farmacologia , Curcumina/análogos & derivados , Curcumina/química , Antibacterianos/farmacologia , Antibacterianos/química , Animais , Mariposas/efeitos dos fármacos , Mariposas/microbiologia , Desinfecção/métodos , Sinergismo Farmacológico , Hidróxido de Cálcio/farmacologia , Cavidade Pulpar/microbiologia
16.
Protein Pept Lett ; 30(10): 795-805, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37817656

RESUMO

BACKGROUND: Brevinin2 HYba5 (Peptide 29) is a novel cationic peptide identified from an endemic frog, Hydrophylax bahuvistara. Staphylococcus aureus and Enterococcus faecalis are troublesome biofilm-forming pathogens associated with nosocomial and community-acquired infections and contribute to the severity of infections associated with implanted devices and chronic wounds. Co-existence of both pathogens in biofilm mode contributes to an increased antibiotic resistance, treatment failure and hence persistent disease burden. Identifying a novel and stable, less toxic compound targeting multispecies biofilm with a lower probability of acquiring resistance in comparison to antibiotics is highly warranted. OBJECTIVE: Evaluate the activity of Brevinin2 HYba5 against S. aureus and E. faecalis mixed biofilm. METHODS: The anti-biofilm activity of peptide 29 was tested by Crystal violet assay, Confocal laser scanning Microscopy (CLSM) and MTT Assay. Cytotoxicity of the peptide was tested in RBC and L929 fibroblast cell line. Biofilm inhibitory activity of the peptide was evaluated at different temperatures, pH, serum and plasma concentrations. The antibiofilm potential of the peptide was tested against polymicrobial biofilm by Fluorescent in situ hybridisation (FISH) and plate counting on HiCromeTM UTI Agar media. RESULTS: The peptide 29 could inhibit biofilm formation of S. aureus and E. faecalis individually as well as in polymicrobial biofilm at 75 µM concentration. The peptide maintained its antibiofilm potential at different temperatures, serum and plasma concentrations. Activity of the peptide was high at acidic and neutral pH but found to get reduced towards alkaline pH. The peptide is nonhemolytic and does not exhibit significant cytotoxicity against the L929 fibroblast cell line (92.80% cell viability). CONCLUSION: The biofilm inhibition property makes peptide 29 a promising candidate for the management of S. aureus and E. faecalis biofilm, especially in catheter-associated devices to prevent the initial colonization and thus can ease the burden of pathogenic biofilm-associated infections.


Assuntos
Enterococcus faecalis , Staphylococcus aureus , Enterococcus faecalis/fisiologia , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Biofilmes , Peptídeos
17.
Beijing Da Xue Xue Bao Yi Xue Ban ; 55(1): 38-43, 2023 Feb 18.
Artigo em Chinês | MEDLINE | ID: mdl-36718687

RESUMO

OBJECTIVE: To construct a model of Enterococcus faecalis (E. faecalis) infection in dentinal tubules by gradient centrifugation and to evaluate the antibacterial effect of low-temperature plasma on E. faecalis in dentinal tubules. METHODS: Standard dentin blocks of 4 mm×4 mm×2 mm size were prepared from single root canal isolated teeth without caries, placed in the E. faecalis bacterial solution, centrifuged in gradient and incubated for 24 h to establish the model of dentinal tubule infection with E. faecalis. The twenty dentin blocks of were divided into five groups, low-temperature plasma jet treatment for 0, 5 and 10 min, calcium hydroxide paste sealing for 7 d and 2% chlorhexidine gel sealing for 7 d. Scanning electron microscopy and confocal laser scanning microscope were used to assess the infection in the dentinal tubules and the antibacterial effect of low-temperature plasma. RESULTS: The results of scanning electron microscopy and confocal laser scanning microscopy showed that after 24 h of incubation by gradient centrifugation, E. faecalis could fully enter the dentinal tubules to a depth of more than 600µm indicating that this method was time-saving and efficient and could successfully construct a model of E. faecalis infection in dentinal tubules. Low-temperature plasma could enter the dentinal tubules and play a role, the structure of E. faecalis was still intact after 5 min of low-temperature plasma treatment, with no obvious damage, and after 10 min of low-temperature plasma treatment, the surface morphology of E. faecalis was crumpled and deformed, the cell wall was seriously collapsed, and the normal physiological morphology was damaged indicating that the majority of E. faecalis was killed in the dentinal tubules. The antibacterial effect of low-temperature plasma treatment for 10 min exceeded that of the calcium hydroxide paste sealing for 7 d and the 2% chlorhexidine gel sealing for 7 d. These two chemicals had difficulty entering deep into the dentinal tubules, and therefore only had a few of antibacterial effect on the bacterial biofilm on the root canal wall, and there was also no significant damage to the E. faecalis bacterial structure. CONCLUSION: Gradient centrifugation could establish the model of E. faecalis dentin infection successfully. Low-temperature plasma treatment for 10 min could kill E. faecalis in dentinal tubules effectively, which is superior to the calcium hydroxide paste sealing for 7 d and the 2% chlorhexidine gel sealing for 7 d.


Assuntos
Hidróxido de Cálcio , Clorexidina , Clorexidina/farmacologia , Hidróxido de Cálcio/farmacologia , Enterococcus faecalis/fisiologia , Temperatura , Dentina , Biofilmes , Antibacterianos/farmacologia , Irrigantes do Canal Radicular/farmacologia , Cavidade Pulpar
18.
J Endod ; 49(2): 198-204, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36509168

RESUMO

INTRODUCTION: The purpose of this study was to evaluate the antimicrobial efficacy of a novel irrigation strategy using synchronized microbubble photodynamic activation (SYMPA) in a minimally prepared single canal. METHODS: Single-canal mandibular incisors were inoculated with Enterococcus faecalis for 3 weeks and randomly allocated to 4 groups based on the irrigation protocols: (1) control (saline), (2) conventional needle irrigation (CI), (3) ultrasonic-assisted irrigation (UI), and (4) irrigation with SYMPA. The first 3 groups were instrumented to size 25.07v (WaveOne Gold Primary; Dentsply Sirona, Johnson City, TN), and the SYMPA group was minimally prepared to size 20.07v (WaveOne Gold Small, Dentsply Sirona). The apical 5 mm was resected for microbiological assessment using the culture technique (colony-forming unit), adenosine-5'-triphosphate-based viability assay (relative luminescence units), and the percentage of live bacteria using confocal laser scanning microscopy. RESULTS: Log colony-forming units from the UI (2.37 ± 0.66) and SYMPA (2.21 ± 0.86) groups showed a reduction compared with the control (5.16 ± 0.75) and CI (4.08 ± 1.19) groups. Relative luminescence unit reduction was significant for UI (619.08 ± 352.78) and SYMPA (415.25 ± 329.51) compared with the control (1213.2 ± 880.03) (P < .05). The percentage of live bacteria was significantly lower in the UI and SYMPA groups compared with the control and CI groups. Although higher microbial reduction was observed in SYMPA compared with UI, there was no statistical significance (P > .05). CONCLUSION: SYMPA in minimally prepared canals showed significant antimicrobial efficacy. The novel irrigation strategy using SYMPA could be an effective disinfection strategy for minimally prepared root canals.


Assuntos
Anti-Infecciosos , Irrigantes do Canal Radicular , Preparo de Canal Radicular , Cavidade Pulpar/microbiologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Microbolhas , Irrigantes do Canal Radicular/farmacologia , Preparo de Canal Radicular/métodos , Hipoclorito de Sódio , Humanos
19.
Biofouling ; 38(9): 903-915, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36451605

RESUMO

The biofilm lifestyle plays a major role in the resistance and virulence of Pseudomonas aeruginosa and Enterococcus faecalis. In this study, two microencapsulated proteases (pepsin ME-PEP and trypsin ME-TRYP) were evaluated for their biofilm dispersal activity and their synergistic effect with microencapsulated carvacrol (ME-CARV). Spray-drying was used to protect enzymes and essential oil and enhance their activities. Cell count analysis proved the synergistic activity of enzymes and carvacrol treatment as biofilms were further reduced after combined treatment in comparison to ME-CARV or enzymes alone. Furthermore, results showed that sequential treatment in the order ME-TRYP - ME-PEP - ME-CARV resulted in more efficient biofilm removal with a maximum reduction of 5 log CFU mL-1 for P. aeruginosa and 4 log CFU mL-1 for E. faecalis. This study proposes that the combination of microencapsulated proteases with ME-CARV could be useful for the effective control of P. aeruginosa and E. faecalis biofilms.


Assuntos
Antibacterianos , Biofilmes , Enterococcus faecalis , Pseudomonas aeruginosa , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Enterococcus faecalis/fisiologia , Pepsina A , Pseudomonas aeruginosa/fisiologia , Tripsina , Composição de Medicamentos
20.
Mol Oral Microbiol ; 37(6): 276-291, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36102211

RESUMO

Enterococcus faecalis, a Gram-positive bacterium, is known to be a key player in several chronic infections as well as nosocomial, heart valve, urinary tract, surgical wound, and dental root canal infections. The capability to sense different transition metal levels and tune its response accordingly endows it with the potential to thrive and cause infections in several host niches. Over the past decade, our knowledge of how transition metals play a critical role in maintaining homeostasis of E. faecalis has improved significantly. The aim of this review is to elucidate the roles of metals such as iron, manganese, zinc, and copper in the physiology, metabolism, and pathogenicity of E. faecalis. These essential micronutrients contribute to energy production, redox stress response, expression of virulence determinants, and cooperation in polymicrobial communities. The review also highlights metal homeostasis systems in E. faecalis, which respond to fluctuations in extracellular metal levels, and regulate the intracellular metal content. Regulation of intracellular metallome secures the tolerance of E. faecalis to oxidative stress and host-mediated metal sequestration strategies. Therapeutic interventions which deprive E. faecalis of its essential metal requirements or disrupt its homeostatic control have been proposed to combat E. faecalis infections.


Assuntos
Enterococcus faecalis , Manganês , Enterococcus faecalis/fisiologia , Virulência , Homeostase , Manganês/metabolismo , Ferro/metabolismo , Metais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...