Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21.697
Filtrar
1.
Arch Microbiol ; 206(7): 289, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38847838

RESUMO

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


Assuntos
Antibacterianos , Biofilmes , Diclofenaco , Staphylococcus epidermidis , Biofilmes/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Staphylococcus epidermidis/fisiologia , Diclofenaco/farmacologia , Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana , Anti-Inflamatórios não Esteroides/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Humanos , Polissacarídeos Bacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos
2.
Sci Rep ; 14(1): 13632, 2024 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-38871850

RESUMO

Helicobacter pylori is a prominent gastrointestinal pathogen associated with various gastrointestinal illnesses. It presents substantial health risks due to its antibiotic resistance. Therefore, it is crucial to identify alternative treatments for H. pylori infections. Limosilactobacillus spp exhibit probiotic properties with beneficial effects in humans; however, the mechanisms by which it counteracts H. pylori infection are unknown. This study aimed to evaluate the potential of Limosilactobacillus fermentum T0701 lyophilized cell-free supernatants (LCFS) against H. pylori. The LCFS has varying antimicrobial activities, with inhibition zones of up to 10.67 mm. The minimum inhibitory concentration and minimum bacterial concentration of LCFS are 6.25-25.00 mg/mL and 6.25 mg/mL to > 50.00 mg/mL, respectively, indicating its capability to inhibit H. pylori. There is morphological damage observed in H. pylori treated with LCFS. Additionally, H. pylori adhesion to AGS cells (human gastric adenocarcinoma epithelial cells) reduces by 74.23%, highlighting the LCFS role in preventing bacterial colonization. Moreover, LCFS exhibits no cytotoxicity or morphological changes in AGS cells, and with no detected virulence or antimicrobial resistance genes, further supporting its safety profile. L. fermentum T0701 LCFS shows promise as a safe and effective non-toxic agent against H. pylori, with the potential to prevent gastric colonization.


Assuntos
Antibacterianos , Helicobacter pylori , Limosilactobacillus fermentum , Testes de Sensibilidade Microbiana , Helicobacter pylori/efeitos dos fármacos , Limosilactobacillus fermentum/fisiologia , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Liofilização , Probióticos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/tratamento farmacológico , Linhagem Celular Tumoral
3.
Nat Commun ; 15(1): 4912, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38851738

RESUMO

Bacterial adhesion is a fundamental process which enables colonisation of niche environments and is key for infection. However, in Legionella pneumophila, the causative agent of Legionnaires' disease, these processes are not well understood. The Legionella collagen-like protein (Lcl) is an extracellular peripheral membrane protein that recognises sulphated glycosaminoglycans on the surface of eukaryotic cells, but also stimulates bacterial aggregation in response to divalent cations. Here we report the crystal structure of the Lcl C-terminal domain (Lcl-CTD) and present a model for intact Lcl. Our data reveal that Lcl-CTD forms an unusual trimer arrangement with a positively charged external surface and negatively charged solvent exposed internal cavity. Through molecular dynamics simulations, we show how the glycosaminoglycan chondroitin-4-sulphate associates with the Lcl-CTD surface via distinct binding modes. Our findings show that Lcl homologs are present across both the Pseudomonadota and Fibrobacterota-Chlorobiota-Bacteroidota phyla and suggest that Lcl may represent a versatile carbohydrate-binding mechanism.


Assuntos
Proteínas de Bactérias , Colágeno , Glicosaminoglicanos , Legionella pneumophila , Simulação de Dinâmica Molecular , Ligação Proteica , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Legionella pneumophila/metabolismo , Colágeno/metabolismo , Colágeno/química , Cristalografia por Raios X , Sulfatos de Condroitina/metabolismo , Sulfatos de Condroitina/química , Aderência Bacteriana , Domínios Proteicos , Doença dos Legionários/microbiologia , Doença dos Legionários/metabolismo , Humanos , Sequência de Aminoácidos
4.
ACS Nano ; 18(24): 16002-16010, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38837910

RESUMO

Understanding bacterial adhesion at the nanoscale is crucial for elucidating biofilm formation, enhancing biosensor performance, and designing advanced biomaterials. However, the dynamics of the critical transition from reversible to irreversible adhesion has remained elusive due to analytical constraints. Here, we probed this adhesion transition, unveiling nanoscale, step-like bacterial approaches to substrates using a plasmonic imaging technique. This method reveals the discontinuous nature of adhesion, emphasizing the complex interplay between bacterial extracellular polymeric substances (EPS) and substrates. Our findings not only deepen our understanding of bacterial adhesion but also have significant implications for the development of theoretical models for biofilm management. By elucidating these nanoscale step-like adhesion processes, our work provides avenues for the application of nanotechnology in biosensing, biofilm control, and the creation of biomimetic materials.


Assuntos
Aderência Bacteriana , Biofilmes , Nanotecnologia , Propriedades de Superfície , Escherichia coli/fisiologia
5.
J Appl Microbiol ; 135(6)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38857885

RESUMO

AIMS: Klebsiella pneumoniae, an important opportunistic pathogen of nosocomial inflection, is known for its ability to form biofilm. The purpose of the current study is to assess how co- or mono-cultured probiotics affect K. pneumoniae's ability to produce biofilms and investigate the potential mechanisms by using a polyester nonwoven chemostat and a Caco-2 cell line. METHODS AND RESULTS: Compared with pure cultures of Lactobacillus rhamnosus and Lactobacillus sake, the formation of K. pneumoniae biofilm was remarkably inhibited by the mixture of L. rhamnosus, L. sake, and Bacillus subtilis at a ratio of 5:5:1 by means of qPCR and FISH assays. In addition, Lactobacillus in combination with B. subtilis could considerably reduce the adherence of K. pneumoniae to Caco-2 cells by using inhibition, competition, and displacement assays. According to the RT-PCR assay, the adsorption of K. pneumoniae to Caco-2 cells was effectively inhibited by the co-cultured probiotics, leading to significant reduction in the expression of proinflammatory cytokines induced by K. pneumoniae. Furthermore, the HPLC and RT-PCR analyses showed that the co-cultured probiotics were able to successfully prevent the expression of the biofilm-related genes of K. pneumoniae by secreting plenty of organic acids as well as the second signal molecule (c-di-GMP), resulting in inhibition on biofilm formation. CONCLUSION: Co-culture of L. sake, L. rhamnosus, and B. subtilis at a ratio of 5:5:1 could exert an antagonistic effect on the colonization of pathogenic K. pneumoniae by down-regulating the expression of biofilm-related genes. At the same time, the co-cultured probiotics could effectively inhibit the adhesion of K. pneumoniae to Caco-2 cells and block the expression of proinflammatory cytokines induced by K. pneumoniae.


Assuntos
Biofilmes , Técnicas de Cocultura , Klebsiella pneumoniae , Probióticos , Biofilmes/crescimento & desenvolvimento , Klebsiella pneumoniae/fisiologia , Humanos , Probióticos/farmacologia , Células CACO-2 , Bacillus subtilis/fisiologia , Bacillus subtilis/genética , Lacticaseibacillus rhamnosus/fisiologia , Aderência Bacteriana , Lactobacillus/fisiologia , Citocinas/metabolismo
6.
Nat Commun ; 15(1): 4764, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38834561

RESUMO

Bacteriophage are sophisticated cellular parasites that can not only parasitize bacteria but are increasingly recognized for their direct interactions with mammalian hosts. Phage adherence to mucus is known to mediate enhanced antimicrobial effects in vitro. However, little is known about the therapeutic efficacy of mucus-adherent phages in vivo. Here, using a combination of in vitro gastrointestinal cell lines, a gut-on-a-chip microfluidic model, and an in vivo murine gut model, we demonstrated that a E. coli phage, øPNJ-6, provided enhanced gastrointestinal persistence and antimicrobial effects. øPNJ-6 bound fucose residues, of the gut secreted glycoprotein MUC2, through domain 1 of its Hoc protein, which led to increased intestinal mucus production that was suggestive of a positive feedback loop mediated by the mucus-adherent phage. These findings extend the Bacteriophage Adherence to Mucus model into phage therapy, demonstrating that øPNJ-6 displays enhanced persistence within the murine gut, leading to targeted depletion of intestinal pathogenic bacteria.


Assuntos
Infecções por Escherichia coli , Escherichia coli , Mucosa Intestinal , Mucina-2 , Animais , Escherichia coli/virologia , Camundongos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/virologia , Mucina-2/metabolismo , Humanos , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/terapia , Terapia por Fagos/métodos , Aderência Bacteriana , Feminino , Muco/metabolismo , Muco/virologia , Colífagos/fisiologia , Fucose/metabolismo , Camundongos Endogâmicos C57BL
7.
Appl Microbiol Biotechnol ; 108(1): 360, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38836914

RESUMO

In the fight against hospital-acquired infections, the challenge posed by methicillin-resistant Staphylococcus aureus (MRSA) necessitates the development of novel treatment methods. This study focused on undermining the virulence of S. aureus, especially by targeting surface proteins crucial for bacterial adherence and evasion of the immune system. A primary aspect of our approach involves inhibiting sortase A (SrtA), a vital enzyme for attaching microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) to the bacterial cell wall, thereby reducing the pathogenicity of S. aureus. Verbascoside, a phenylethanoid glycoside, was found to be an effective SrtA inhibitor in our research. Advanced fluorescence quenching and molecular docking studies revealed a specific interaction between verbascoside and SrtA, pinpointing the critical active sites involved in this interaction. This molecular interaction significantly impedes the SrtA-mediated attachment of MSCRAMMs, resulting in a substantial reduction in bacterial adhesion, invasion, and biofilm formation. The effectiveness of verbascoside has also been demonstrated in vivo, as shown by its considerable protective effects on pneumonia and Galleria mellonella (wax moth) infection models. These findings underscore the potential of verbascoside as a promising component in new antivirulence therapies for S. aureus infections. By targeting crucial virulence factors such as SrtA, agents such as verbascoside constitute a strategic and potent approach for tackling antibiotic resistance worldwide. KEY POINTS: • Verbascoside inhibits SrtA, reducing S. aureus adhesion and biofilm formation. • In vivo studies demonstrated the efficacy of verbascoside against S. aureus infections. • Targeting virulence factors such as SrtA offers new avenues against antibiotic resistance.


Assuntos
Aminoaciltransferases , Antibacterianos , Aderência Bacteriana , Proteínas de Bactérias , Biofilmes , Cisteína Endopeptidases , Glucosídeos , Staphylococcus aureus Resistente à Meticilina , Simulação de Acoplamento Molecular , Fenóis , Infecções Estafilocócicas , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Aminoaciltransferases/antagonistas & inibidores , Aminoaciltransferases/metabolismo , Cisteína Endopeptidases/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Glucosídeos/farmacologia , Animais , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Fenóis/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Antibacterianos/farmacologia , Mariposas/microbiologia , Virulência/efeitos dos fármacos , Modelos Animais de Doenças , Fatores de Virulência/metabolismo , Inibidores Enzimáticos/farmacologia , Polifenóis
8.
Vet Res ; 55(1): 70, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822378

RESUMO

Adaptation of avian pathogenic E. coli (APEC) to changing host environments including virulence factors expression is vital for disease progression. FdeC is an autotransporter adhesin that plays a role in uropathogenic Escherichia coli (UPEC) adhesion to epithelial cells. Expression of fdeC is known to be regulated by environmental conditions in UPEC and Shiga toxin-producing E. coli (STEC). The observation in a previous study that an APEC strain IMT5155 in which the fdeC gene was disrupted by a transposon insertion resulted in elevated adhesion to chicken intestinal cells prompted us to further explore the role of fdeC in infection. We found that the fdeC gene prevalence and FdeC variant prevalence differed between APEC and nonpathogenic E. coli genomes. Expression of the fdeC gene was induced at host body temperature, an infection relevant condition. Disruption of fdeC resulted in greater adhesion to CHIC-8E11 cells and increased motility at 42 °C compared to wild type (WT) and higher expression of multiple transporter proteins that increased inorganic ion export. Increased motility may be related to increased inorganic ion export since this resulted in downregulation of YbjN, a protein known to supress motility. Inactivation of fdeC in APEC strain IMT5155 resulted in a weaker immune response in chickens compared to WT in experimental infections. Our findings suggest that FdeC is upregulated in the host and contributes to interactions with the host by down-modulating motility during colonization. A thorough understanding of the regulation and function of FdeC could provide novel insights into E. coli pathogenesis.


Assuntos
Adesinas de Escherichia coli , Aderência Bacteriana , Galinhas , Infecções por Escherichia coli , Doenças das Aves Domésticas , Doenças das Aves Domésticas/microbiologia , Infecções por Escherichia coli/veterinária , Infecções por Escherichia coli/microbiologia , Animais , Adesinas de Escherichia coli/genética , Adesinas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Escherichia coli/fisiologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo
9.
Microbiol Res ; 285: 127782, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38833832

RESUMO

As a major human and animal pathogen, Staphylococcus aureus can attach to medical implants (abiotic surface) or host tissues (biotic surface), and further establish robust biofilms which enhances resistance and persistence to host immune system and antibiotics. Cell-wall-anchored proteins (CWAPs) covalently link to peptidoglycan, and largely facilitate the colonization of S. aureus on various surfaces (including adhesion and biofilm formation) and invasion into host cells (including adhesion, immune evasion, iron acquisition and biofilm formation). During biofilm formation, CWAPs function in adhesion, aggregation, collagen-like fiber network formation, and consortia formation. In this review, we firstly focus on the structural features of CWAPs, including their intracellular function and interactions with host cells, as well as the functions and ligand binding of CWAPs in different stages of S. aureus biofilm formation. Then, the roles of CWAPs in different biofilm processes with regards in development of therapeutic approaches are clarified, followed by the association between CWAPs genes and clonal lineages. By touching upon these aspects, we hope to provide comprehensive knowledge and clearer understanding on the CWAPs of S. aureus and their roles in biofilm formation, which may further aid in prevention and treatment infection and vaccine development.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias , Biofilmes , Parede Celular , Infecções Estafilocócicas , Staphylococcus aureus , Biofilmes/crescimento & desenvolvimento , Staphylococcus aureus/fisiologia , Staphylococcus aureus/genética , Humanos , Infecções Estafilocócicas/microbiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Parede Celular/metabolismo , Animais , Peptidoglicano/metabolismo
10.
Med Sci Monit ; 30: e944255, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38843112

RESUMO

Orthodontic treatments, while essential for achieving optimal oral health, present challenges in infection control due to the propensity for bacterial adhesion and biofilm formation on orthodontic appliances. Silver-coated orthodontic materials have emerged as a promising solution, leveraging the potent antimicrobial properties of silver nanoparticles (AgNPs). Antibacterial coatings are used in orthodontics to prevent the formation of bacterial biofilms. This systematic review evaluated the literature on antimicrobial silver coatings on fixed orthodontic appliances, including archwires, brackets, and microimplants. Two evaluators, working independently, rigorously conducted a comprehensive search of various databases, including PubMed, PubMed Central, Embase, Scopus and Web of Science. This systematic review comprehensively examined in vitro studies investigating the antimicrobial efficacy of silver-coated orthodontic archwires, brackets, and microimplants. The review registered in PROSPERO CRD42024509189 synthesized findings from 18 diverse studies, revealing consistent and significant reductions in bacterial adhesion, biofilm formation, and colony counts with the incorporation of AgNPs. Key studies demonstrated the effectiveness of silver-coated archwires and brackets against common oral bacteria, such as Streptococcus mutans and Staphylococcus aureus. Microimplants coated with AgNPs also exhibited notable antimicrobial activity against a range of microorganisms. The systematic review revealed potential mechanisms underlying these antimicrobial effects, highlighted implications for infection prevention in orthodontic practice, and suggested future research avenues. Despite some study heterogeneity and limitations, the collective evidence supports the potential of silver-coated orthodontic materials in mitigating bacterial complications, emphasizing their relevance in advancing infection control measures in orthodontics.


Assuntos
Biofilmes , Nanopartículas Metálicas , Braquetes Ortodônticos , Prata , Prata/farmacologia , Humanos , Biofilmes/efeitos dos fármacos , Braquetes Ortodônticos/microbiologia , Fios Ortodônticos/microbiologia , Aparelhos Ortodônticos Fixos , Anti-Infecciosos/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Antibacterianos/farmacologia , Streptococcus mutans/efeitos dos fármacos , Aderência Bacteriana/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos
11.
Int J Biol Macromol ; 271(Pt 1): 132698, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38824104

RESUMO

Ecofriendly fabrics with antibacterial and anti-adhesion properties have been attracted an increasing attention in recent years. Herein, natural menthol modified polyacrylate (PMCA) antibacterial adhesion agent was synthesized by esterification and polymerisation while natural pterostilbene-grafted-chitosan (PGC) antibacterial agent was prepared through Mannich reaction. The antibacterial and anti-adhesion cotton fabric was fabricated through durable PMCA dip finishing and then layer-by-layer self-assembly of PGC. The results showed that the antibacterial adhesion rates and antibacterial rates of the dual-function cotton fabric against Staphylococcus aureus and Escherichia coli reached up to 99.9 %. Its antibacterial adhesion rates improved by 36.1 % and 40.1 % in comparison with those of cotton fabric treated by menthol alone. Meanwhile against S. aureus, the dual-function cotton fabrics improved the antibacterial rates by 56.7 % and 36.4 %, respectively, from those of chitosan- and pterostilbene-treated fabrics. Against E. coli, the improvements were 89.4 % and 24.8 %, respectively. After 20 household washings, the dual-function cotton fabric maintained >80 % of its original anti-adhesion and antibacterial rates against both species. The dual-function cotton fabric also possessed safe and excellent wearability.


Assuntos
Antibacterianos , Quitosana , Fibra de Algodão , Escherichia coli , Staphylococcus aureus , Antibacterianos/farmacologia , Antibacterianos/química , Escherichia coli/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Quitosana/química , Quitosana/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Estilbenos/farmacologia , Estilbenos/química , Têxteis , Testes de Sensibilidade Microbiana , Resinas Acrílicas/química
12.
Appl Microbiol Biotechnol ; 108(1): 384, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38896287

RESUMO

Bacteriocins have the potential to effectively improve food-borne infections or gastrointestinal diseases and hold promise as viable alternatives to antibiotics. This study aimed to explore the antibacterial activity of three bacteriocins (nisin, enterocin Gr17, and plantaricin RX-8) and their ability to attenuate intestinal barrier dysfunction and inflammatory responses induced by Listeria monocytogenes, respectively. Bacteriocins have shown excellent antibacterial activity against L. monocytogenes without causing any cytotoxicity. Bacteriocins inhibited the adhesion and invasion of L. monocytogenes on Caco-2 cells, lactate dehydrogenase (LDH), trans-epithelial electrical resistance (TEER), and cell migration showed that bacteriocin improved the permeability of Caco-2 cells. These results were attributed to the promotion of tight junction proteins (TJP) assembly, specifically zonula occludens-1 (ZO-1), occludin, and claudin-1. Furthermore, bacteriocins could alleviate inflammation by inhibiting the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways and reducing the secretion of interleukin-6 (IL-6), interleukin-1 ß (IL-1ß) and tumor necrosis factor α (TNF-α). Among three bacteriocins, plantaricin RX-8 showed the best antibacterial activity against L. monocytogenes and the most pronounced protective effect on the intestinal barrier due to its unique structure. Based on our findings, we hypothesized that bacteriocins may inhibit the adhesion and invasion of L. monocytogenes by competing adhesion sites. Moreover, they may further enhance intestinal barrier function by inhibiting the expression of L. monocytogenes virulence factors, increasing the expression of TJP and decreasing the secretion of inflammatory factors. Therefore, bacteriocins will hopefully be an effective alternative to antibiotics, and this study provides valuable insights into food safety concerns. KEY POINTS: • Bacteriocins show excellent antibacterial activity against L. monocytogenes • Bacteriocins improve intestinal barrier damage and inflammatory response • Plantaricin RX-8 has the best protective effect on Caco-2 cells damage.


Assuntos
Antibacterianos , Bacteriocinas , Listeria monocytogenes , Listeria monocytogenes/efeitos dos fármacos , Bacteriocinas/farmacologia , Humanos , Células CACO-2 , Antibacterianos/farmacologia , Inflamação , NF-kappa B/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Proteínas de Junções Íntimas/metabolismo , Citocinas/metabolismo , Listeriose/microbiologia , Listeriose/tratamento farmacológico , Movimento Celular/efeitos dos fármacos
13.
ACS Appl Mater Interfaces ; 16(24): 30967-30979, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38857475

RESUMO

The ongoing global health has highlighted the critical issue of secondary infections, particularly antibiotic-resistant bacterial infections, which have been significant contributors to mortality rates. Orthopedic implants, while essential for trauma and orthopedic surgeries, are particularly susceptible to these infections, leading to severe complications and economic burdens. The traditional use of antibiotics in treating these infections poses further challenges including the risk of developing antibiotic-resistant bacteria. This study introduces a novel approach to combat this issue by developing nanostructured surfaces for orthopedic implants using target ion-induced plasma sputtering. Inspired by the natural design of dragonfly wings, these surfaces aim to prevent bacterial adhesion while promoting preosteoblast activity, offering a dual-function solution to the problems of bacterial infection and implant integration without relying on antibiotics. The in vitro results demonstrate the effectiveness of these bioinspired surfaces in eradicating bacteria and supporting cell proliferation and differentiation, presenting a promising alternative for the development of biomedical implants.


Assuntos
Antibacterianos , Osseointegração , Antibacterianos/farmacologia , Antibacterianos/química , Animais , Osseointegração/efeitos dos fármacos , Nanoestruturas/química , Camundongos , Propriedades de Superfície , Staphylococcus aureus/efeitos dos fármacos , Próteses e Implantes , Aderência Bacteriana/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Linhagem Celular
14.
Int J Mol Sci ; 25(11)2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38891904

RESUMO

Tooth loss during the lifetime of an individual is common. A strategy to treat partial or complete edentulous patients is the placement of dental implants. However, dental implants are subject to bacterial colonization and biofilm formation, which cause an infection named peri-implantitis. The existing long-term treatments for peri-implantitis are generally inefficient. Thus, an electrical circuit was produced with zirconia (Zr) samples using a hot-pressing technique to impregnate silver (Ag) through channels and holes to create a path by LASER texturing. The obtained specimens were characterized according to vitro cytotoxicity, to ensure ZrAg non-toxicity. Furthermore, samples were inoculated with Staphylococcus aureus using 6.5 mA of alternating current (AC). The current was delivered using a potentiostat and the influence on the bacterial concentration was assessed. Using AC, the specimens displayed no bacterial adhesion (Log 7 reduction). The in vitro results presented in this study suggest that this kind of treatment can be an alternative and promising strategy to treat and overcome bacterial adhesion around dental implants that can evolve to biofilm.


Assuntos
Aderência Bacteriana , Biofilmes , Implantes Dentários , Staphylococcus aureus , Zircônio , Implantes Dentários/microbiologia , Zircônio/química , Biofilmes/crescimento & desenvolvimento , Biofilmes/efeitos dos fármacos , Humanos , Estimulação Elétrica/métodos , Propriedades de Superfície , Peri-Implantite/microbiologia , Peri-Implantite/terapia , Prata/química , Prata/farmacologia
15.
Arch Microbiol ; 206(7): 322, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38907754

RESUMO

Limosilactobacillus reuteri DSM17938 is one of the most pivotal probiotics, whose general beneficial effects on the intestinal microbiota are well recognized. Enhancing their growth and metabolic activity can effectively regulate the equilibrium of intestinal microbiota, leading to improved physical health. A common method to promote the growth of Lactobacillus is the addition of prebiotics. Current research suggests that proteins and their hydrolysates from different sources with potential prebiotic activity can also promote the growth of probiotics. In this study, soybean proteins and peptides were effective in promoting the growth, organic acid secretion, and adhesive properties of Limosilactobacillus reuteri DSM17938 to Caco-2 cells. These results illustrate the feasibility of soybean proteins and peptides as prebiotics, providing theoretical and practical advantages for their application.


Assuntos
Aderência Bacteriana , Limosilactobacillus reuteri , Peptídeos , Probióticos , Proteínas de Soja , Limosilactobacillus reuteri/crescimento & desenvolvimento , Limosilactobacillus reuteri/metabolismo , Proteínas de Soja/farmacologia , Proteínas de Soja/metabolismo , Proteínas de Soja/química , Humanos , Células CACO-2 , Aderência Bacteriana/efeitos dos fármacos , Peptídeos/farmacologia , Prebióticos , Microbioma Gastrointestinal/efeitos dos fármacos , Glycine max/microbiologia
16.
BMC Microbiol ; 24(1): 221, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38909237

RESUMO

BACKGROUND: Group B Streptococcus (GBS) is a commensal of healthy adults and an important pathogen in newborns, the elderly and immunocompromised individuals. GBS displays several virulence factors that promote colonisation and host infection, including the ST-17 strain-specific adhesin Srr2, previously characterised for its binding to fibrinogen. Another common target for bacterial adhesins and for host colonization is fibronectin, a multi-domain glycoprotein found ubiquitously in body fluids, in the extracellular matrix and on the surface of cells. RESULTS: In this study, fibronectin was identified as a novel ligand for the Srr2 adhesin of GBS. A derivative of the ST-17 strain BM110 overexpressing the srr2 gene showed an increased ability to bind fibrinogen and fibronectin, compared to the isogenic wild-type strain. Conversely, the deletion of srr2 impaired bacterial adhesion to both ligands. ELISA assays and surface plasmon resonance studies using the recombinant binding region (BR) form of Srr2 confirmed a direct interaction with fibronectin with an estimated Kd of 92 nM. Srr2-BR variants defective in fibrinogen binding also exhibited no interaction with fibronectin, suggesting that Srr2 binds this ligand through the dock-lock-latch mechanism, previously described for fibrinogen binding. The fibronectin site responsible for recombinant Srr2-BR binding was identified and localised in the central cell-binding domain of the protein. Finally, in the presence of fibronectin, the ability of a Δsrr2 mutant to adhere to human cervico-vaginal epithelial cells was significantly lower than that of the wild-type strain. CONCLUSION: By combining genetic and biochemical approaches, we demonstrate a new role for Srr2, namely interacting with fibronectin. We characterised the molecular mechanism of this interaction and demonstrated that it plays a role in promoting the adhesion of GBS to human cervico-vaginal epithelial cells, further substantiating the role of Srr2 as a factor responsible for the hypervirulence of GBS ST-17 strains. The discovery of the previously undescribed interaction between Srr2 and fibronectin establishes this adhesin as a key factor for GBS colonisation of host tissues.


Assuntos
Adesinas Bacterianas , Aderência Bacteriana , Fibronectinas , Ligação Proteica , Streptococcus agalactiae , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidade , Fibronectinas/metabolismo , Humanos , Adesinas Bacterianas/metabolismo , Adesinas Bacterianas/genética , Fibrinogênio/metabolismo , Fibrinogênio/genética , Células Epiteliais/microbiologia , Feminino , Infecções Estreptocócicas/microbiologia , Fatores de Virulência/metabolismo , Fatores de Virulência/genética
17.
Curr Biol ; 34(11): R539-R541, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38834027

RESUMO

Strain-specific pili enable Vibrio cholerae bacteria to adhere to each other and form aggregates in liquid culture. A new study focuses on strains with less specific, promiscuous pili and suggests a role for contact-dependent bacterial killing in shaping the composition of these aggregates.


Assuntos
Fímbrias Bacterianas , Vibrio cholerae , Vibrio cholerae/fisiologia , Vibrio cholerae/genética , Fímbrias Bacterianas/fisiologia , Fímbrias Bacterianas/metabolismo , Aderência Bacteriana/fisiologia
18.
Nat Commun ; 15(1): 4582, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811534

RESUMO

The intestinal anaerobic bacterium Akkermansia muciniphila is specialized in the degradation of mucins, which are heavily O-glycosylated proteins that constitute the major components of the mucus lining the intestine. Despite that adhesion to mucins is considered critical for the persistence of A. muciniphila in the human intestinal tract, our knowledge of how this intestinal symbiont recognizes and binds to mucins is still limited. Here, we first show that the mucin-binding properties of A. muciniphila are independent of environmental oxygen concentrations and not abolished by pasteurization. We then dissected the mucin-binding properties of pasteurized A. muciniphila by use of a recently developed cell-based mucin array that enables display of the tandem repeats of human mucins with distinct O-glycan patterns and structures. We found that A. muciniphila recognizes the unsialylated LacNAc (Galß1-4GlcNAcß1-R) disaccharide selectively on core2 and core3 O-glycans. This disaccharide epitope is abundantly found on human colonic mucins capped by sialic acids, and we demonstrated that endogenous A. muciniphila neuraminidase activity can uncover the epitope and promote binding. In summary, our study provides insights into the mucin-binding properties important for colonization of a key mucin-foraging bacterium.


Assuntos
Akkermansia , Mucinas , Polissacarídeos , Akkermansia/metabolismo , Humanos , Mucinas/metabolismo , Polissacarídeos/metabolismo , Neuraminidase/metabolismo , Ligação Proteica , Glicosilação , Dissacarídeos/metabolismo , Verrucomicrobia/metabolismo , Epitopos/metabolismo , Aderência Bacteriana
19.
BMC Oral Health ; 24(1): 633, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811903

RESUMO

BACKGROUND: In this study, the antimicrobial activity of three different cleanser tablets on S. mutans and C. albicans adhesion to PMMA, polyamide and 3D printed resin was investigated. METHODS: 40 samples were prepared for PMMA (SR Triplex Hot), polyamide (Deflex) and 3D printed resin (PowerResins Denture) materials and divided into four subgroups for cleansers (Aktident™, Protefix™, Corega™ tablets and distilled water) (n = 5). After the surface preparations were completed, the samples were immersed separately in tubes containing the prepared microorganism suspension and incubated at 37˚C for 24 h. After the incubation, the samples were kept in the cleanser solutions. The samples were then transferred to sterile saline tubes. All the tubes were vortexed and 10 µl was taken from each of them. Sheep blood agar was inoculated for colony counting. The inoculated plates were incubated for 48 h for S. mutans and 24 h for C. albicans. After incubation, colonies observed on all plates were counted. Statistical analyses were done with three-way ANOVA and Tukey's multiple comparison test. RESULTS: Polyamide material registered the highest colony count of S. mutans, whereas PMMA registered the lowest. Significant differences in S. mutans adherence (p = 0.002) were found between the three denture base materials, but no such difference in C. albicans adherence (p = 0.221) was identified between the specimens. All three cleanser tablets eliminated 98% of S. mutans from all the material groups. In all these groups, as well, the antifungal effect of Corega™ on C. albicans was significantly higher than those of the other two cleanser tablets. CONCLUSIONS: According to the study's results, it may be better to pay attention to surface smoothness when using polyamide material to prevent microorganism retention. Cleanser tablets are clinically recommended to help maintain hygiene in removable denture users, especially Corega tablets that are more effective on C. albicans.


Assuntos
Candida albicans , Bases de Dentadura , Higienizadores de Dentadura , Polimetil Metacrilato , Streptococcus mutans , Candida albicans/efeitos dos fármacos , Streptococcus mutans/efeitos dos fármacos , Bases de Dentadura/microbiologia , Higienizadores de Dentadura/farmacologia , Polimetil Metacrilato/química , Nylons/farmacologia , Comprimidos , Contagem de Colônia Microbiana , Materiais Dentários/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Anti-Infecciosos/farmacologia , Teste de Materiais
20.
J Mater Chem B ; 12(23): 5711-5721, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38758163

RESUMO

The presence of a variety of bacteria is an inevitable/indispensable part of human life. In particular, for patients, the existence and spreading of bacteria lead to prolonged treatment period with many more complications. The widespread use of urinary catheters is one of the main causes for the prevalence of infections. The necessity of long-term use of indwelling catheters is unavoidable in terms of the development of bacteriuria and blockage. As is known, since a permanent solution to this problem has not yet been found, research and development activities continue actively. Herein, polyethylene glycol (PEG)-like thin films were synthesized by a custom designed plasma enhanced chemical vapor deposition (PE-CVD) method and the long-term effect of antifouling properties of PEG-like coated catheters was investigated against Escherichia coli and Proteus mirabilis. The contact angle measurements have revealed the increase of wettability with the increase of plasma exposure time. The antifouling activity of surface-coated catheters was analyzed against the Gram-negative/positive bacteria over a long-term period (up to 30 days). The results revealed that PE-CVD coated PEG-like thin films are highly capable of eliminating bacterial attachment on surfaces with relatively reduced protein attachment without having any toxic effect. Previous statements were supported with SEM, XPS, FTIR spectroscopy, and contact angle analysis.


Assuntos
Escherichia coli , Polietilenoglicóis , Proteus mirabilis , Propriedades de Superfície , Cateteres Urinários , Cateteres Urinários/microbiologia , Escherichia coli/efeitos dos fármacos , Proteus mirabilis/efeitos dos fármacos , Polietilenoglicóis/química , Aderência Bacteriana/efeitos dos fármacos , Incrustação Biológica/prevenção & controle , Humanos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...