Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.710
Filter
1.
Microbiol Res ; 285: 127782, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833832

ABSTRACT

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.


Subject(s)
Bacterial Adhesion , Bacterial Proteins , Biofilms , Cell Wall , Staphylococcal Infections , Staphylococcus aureus , Biofilms/growth & development , Staphylococcus aureus/physiology , Staphylococcus aureus/genetics , Humans , Staphylococcal Infections/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cell Wall/metabolism , Animals , Peptidoglycan/metabolism
2.
Nat Commun ; 15(1): 4912, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851738

ABSTRACT

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.


Subject(s)
Bacterial Proteins , Collagen , Glycosaminoglycans , Legionella pneumophila , Molecular Dynamics Simulation , Protein Binding , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Legionella pneumophila/metabolism , Collagen/metabolism , Collagen/chemistry , Crystallography, X-Ray , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/chemistry , Bacterial Adhesion , Protein Domains , Legionnaires' Disease/microbiology , Legionnaires' Disease/metabolism , Humans , Amino Acid Sequence
3.
Sci Rep ; 14(1): 13632, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871850

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Helicobacter pylori , Limosilactobacillus fermentum , Microbial Sensitivity Tests , Helicobacter pylori/drug effects , Limosilactobacillus fermentum/physiology , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Freeze Drying , Probiotics/pharmacology , Bacterial Adhesion/drug effects , Helicobacter Infections/microbiology , Helicobacter Infections/drug therapy , Cell Line, Tumor
4.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891904

ABSTRACT

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.


Subject(s)
Bacterial Adhesion , Biofilms , Dental Implants , Staphylococcus aureus , Zirconium , Dental Implants/microbiology , Zirconium/chemistry , Biofilms/growth & development , Biofilms/drug effects , Humans , Electric Stimulation/methods , Surface Properties , Peri-Implantitis/microbiology , Peri-Implantitis/therapy , Silver/chemistry , Silver/pharmacology
5.
Int J Biol Macromol ; 271(Pt 1): 132698, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38824104

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents , Chitosan , Cotton Fiber , Escherichia coli , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Bacterial Adhesion/drug effects , Stilbenes/pharmacology , Stilbenes/chemistry , Textiles , Microbial Sensitivity Tests , Acrylic Resins/chemistry
6.
ACS Nano ; 18(24): 16002-16010, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38837910

ABSTRACT

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.


Subject(s)
Bacterial Adhesion , Biofilms , Nanotechnology , Surface Properties , Escherichia coli/physiology
7.
J Appl Microbiol ; 135(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38857885

ABSTRACT

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.


Subject(s)
Biofilms , Coculture Techniques , Klebsiella pneumoniae , Probiotics , Biofilms/growth & development , Klebsiella pneumoniae/physiology , Humans , Probiotics/pharmacology , Caco-2 Cells , Bacillus subtilis/physiology , Bacillus subtilis/genetics , Lacticaseibacillus rhamnosus/physiology , Bacterial Adhesion , Lactobacillus/physiology , Cytokines/metabolism
8.
Biofouling ; 40(5-6): 333-347, 2024.
Article in English | MEDLINE | ID: mdl-38836545

ABSTRACT

The corrosion behaviors of four pure metals (Fe, Ni, Mo and Cr) in the presence of sulfate reducing bacteria (SRB) were investigated in enriched artificial seawater (EASW) after 14-day incubation. Metal Fe and metal Ni experienced weight losses of 1.96 mg cm-2 and 1.26 mg cm-2, respectively. In contrast, metal Mo and metal Cr exhibited minimal weight losses, with values of only 0.05 mg cm-2 and 0.03 mg cm-2, respectively. In comparison to Mo (2.2 × 106 cells cm-2) or Cr (1.4 × 106 cells cm-2) surface, the sessile cell counts on Fe (4.0 × 107 cells cm-2) or Ni (3.1 × 107 cells cm-2) surface was higher.


Subject(s)
Bacterial Adhesion , Sulfates , Corrosion , Sulfates/chemistry , Metals/chemistry , Seawater/microbiology , Seawater/chemistry , Biofilms/drug effects , Biofilms/growth & development , Bacteria/drug effects , Biofouling/prevention & control
9.
Sci Rep ; 14(1): 14426, 2024 06 23.
Article in English | MEDLINE | ID: mdl-38910172

ABSTRACT

Vaginitis, a prevalent gynecological condition in women, is mainly caused by an imbalance in the vaginal micro-ecology. The two most common types of vaginitis are vaginal bacteriosis and vulvovaginal candidiasis, triggered by the virulent Gardnerella vaginalis and Candida albicans, respectively. In this study, a strain capable of inhibiting G. vaginalis and C. albicans was screened from vaginal secretions and identified as Lactobacillus gasseri based on 16S rRNA sequences. The strain, named L. gasseri VHProbi E09, could inhibit the growth of G. vaginalis and C. albicans under co-culture conditions by 99.07% ± 0.26% and 99.95% ± 0.01%, respectively. In addition, it could significantly inhibit the adhesion of these pathogens to vaginal epithelial cells. The strain further showed the ability to inhibit the enteropathogenic bacteria Escherichia coli and Salmonella enteritidis, to tolerate artificial gastric and intestinal fluids and to adhere to intestinal Caco-2 cells. These results suggest that L. gasseri VHProbi E09 holds promise for clinical trials and animal studies whether administered orally or directly into the vagina. Whole-genome analysis also revealed a genome consisting of 1752 genes for L. gasseri VHProbi E09, with subsequent analyses identifying seven genes related to adhesion and three genes related to bacteriocins. These adhesion- and bacteriocin-related genes provide a theoretical basis for understanding the mechanism of bacterial inhibition of the strain. The research conducted in this study suggests that L. gasseri VHProbi E09 may be considered as a potential probiotic, and further research can delve deeper into its efficacy as an agent which can restore a healthy vaginal ecosystem.


Subject(s)
Candida albicans , Gardnerella vaginalis , Lactobacillus gasseri , Probiotics , Vagina , Female , Humans , Lactobacillus gasseri/genetics , Caco-2 Cells , Gardnerella vaginalis/genetics , Vagina/microbiology , Bacterial Adhesion , Vaginitis/microbiology , RNA, Ribosomal, 16S/genetics
10.
Nanoscale ; 16(25): 12134-12141, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38832761

ABSTRACT

Type IV pili (TFP) contribute to the ability of microbes such as Pseudomonas aeruginosa to engage with and move across surfaces. We reported previously that P. aeruginosa TFP generate retractive forces of ∼30 pN and provided indirect evidence that TFP-mediated surface attachment was enhanced in the presence of the Pel polysaccharide. Here, we use different mutants defective in flagellar, Pel production or TFP production - alone or in combination - to decipher the relative contribution of these biofilm-promoting factors for P. aeruginosa adhesion. By means of atomic force microscopy (AFM), we show that mutating the flagellum (ΔflgK mutant) results in an increase in Pel polysaccharide production, but this increase in Pel does not result in an increase in surface adhesive properties compared to those previously described for the WT strain. By blocking Pel production in the ΔflgK mutant (ΔflgKΔpel), we directly show that TFP play a major role in the adhesion of the bacteria to hydrophobic AFM tips, but that the adhesion force is only slightly impaired by the absence of Pel. Inversely, performing single-cell force spectroscopy measurements with the mutant lacking TFP (ΔflgKΔpilA) reveals that the Pel can modulate the attachment of the bacteria to a hydrophobic substrate in a time-dependent manner. Finally, little adhesion was detected for the ΔflgKΔpilAΔpelA triple mutant, suggesting that both TFP and Pel polysaccharide make a substantial contribution to bacteria-substratum interaction events. Altogether, our data allow us to decipher the relative contribution of Pel and TFP in the early attachment by P. aeruginosa.


Subject(s)
Bacterial Adhesion , Fimbriae, Bacterial , Microscopy, Atomic Force , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology , Fimbriae, Bacterial/metabolism , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/metabolism , Biofilms/growth & development , Flagella/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Mutation
11.
Front Cell Infect Microbiol ; 14: 1379206, 2024.
Article in English | MEDLINE | ID: mdl-38938878

ABSTRACT

Enteroaggregative E. coli (EAEC) is a major cause of diarrhea worldwide. EAEC are highly adherent to cultured epithelial cells and make biofilms. Both adherence and biofilm formation rely on the presence of aggregative adherence fimbriae (AAF). We compared biofilm formation from two EAEC strains of each of the five AAF types. We found that AAF type did not correlate with the level of biofilm produced. Because the composition of the EAEC biofilm has not been fully described, we stained EAEC biofilms to determine if they contained protein, carbohydrate glycoproteins, and/or eDNA and found that EAEC biofilms contained all three extracellular components. Next, we assessed the changes to the growing or mature EAEC biofilm mediated by treatment with proteinase K, DNase, or a carbohydrate cleavage agent to target the different components of the matrix. Growing biofilms treated with proteinase K had decreased biofilm staining for more than half of the strains tested. In contrast, although sodium metaperiodate only altered the biofilm in a quantitative way for two strains, images of biofilms treated with sodium metaperiodate showed that the EAEC were more spread out. Overall, we found variability in the response of the EAEC strains to the treatments, with no one treatment producing a biofilm change for all strains. Finally, once formed, mature EAEC biofilms were more resistant to treatment than biofilms grown in the presence of those same treatments.


Subject(s)
Biofilms , Deoxyribonucleases , Endopeptidase K , Escherichia coli , Biofilms/drug effects , Biofilms/growth & development , Endopeptidase K/pharmacology , Endopeptidase K/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Deoxyribonucleases/metabolism , Deoxyribonucleases/pharmacology , Fimbriae, Bacterial/metabolism , Bacterial Adhesion/drug effects , Humans , Periodic Acid/pharmacology
12.
Sci Rep ; 14(1): 14964, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38942800

ABSTRACT

Mycobacterium avium subspecies paratuberculosis (MAP) is the causative agent of Johne's Disease, a chronic granulomatous enteritis of ruminants. MAP establishes an infection in the host via the small intestine. This requires the bacterium to adhere to, and be internalised by, cells of the intestinal tract. The effector molecules expressed by MAP for this purpose remain to be fully identified and understood. Mammalian cell entry (mce) proteins have been shown to enable other Mycobacterial species to attach to and invade host epithelial cells. Here, we have expressed Mce1A, Mce1D, Mce3C and Mce4A proteins derived from MAP on the surface of a non-invasive Escherichia coli to characterise their role in the initial interaction between MAP and the host. To this end, expression of mce1A was found to significantly increase the ability of the E. coli to attach and survive intracellularly in human monocyte-like THP-1 cells, whereas expression of mce1D was found to significantly increase attachment and invasion of E. coli to bovine epithelial cell-like MDBK cells, implying cell-type specificity. Furthermore, expression of Mce1A and Mce1D on the surface of a previously non-invasive E. coli enhanced the ability of the bacterium to infect 3D bovine basal-out enteroids. Together, our data contributes to our understanding of the effector molecules utilised by MAP in the initial interaction with the host, and may provide potential targets for therapeutic intervention.


Subject(s)
Bacterial Proteins , Mycobacterium avium subsp. paratuberculosis , Paratuberculosis , Mycobacterium avium subsp. paratuberculosis/metabolism , Paratuberculosis/microbiology , Animals , Humans , Cattle , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Adhesion , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Escherichia coli/metabolism , Cell Line , THP-1 Cells
13.
Food Res Int ; 190: 114555, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945560

ABSTRACT

Cronobacter sakazakii, an opportunity foodborne pathogen, could contaminate a broad range of food materials and cause life-threatening symptoms in infants. The bacterial envelope structure contribute to bacterial environment tolerance, biofilm formation and virulence in various in Gram-negative bacteria. DsbA and PepP are two important genes related to the biogenesis and stability of bacterial envelope. In this study, the DsbA and PepP were deleted in C. sakazakii to evaluate their contribution to stress tolerance and virulence of the pathogen. The bacterial environment resistance assays showed DsbA and PepP are essential in controlling C. sakazakii resistance to heat and desiccation in different mediums, as well as acid, osmotic, oxidation and bile salt stresses. DsbA and PepP also played an important role in regulating biofilm formation and motility. Furthermore, DsbA and PepP deletion weaken C. sakazakii adhesion and invasion in Caco-2, intracellular survival and replication in RAW 264.7. qRT-PCR results showed that DsbA and PepP of C. sakazakii played roles in regulating the expression of several genes associated with environment stress tolerance, biofilm formation, bacterial motility and cellular invasion. These findings indicate that DsbA and PepP played an important regulatory role in the environment resisitance, biofilm formation and virulence of C. sakazakii, which enrich understanding of genetic determinants of adaptability and virulence of the pathogen.


Subject(s)
Biofilms , Cronobacter sakazakii , Virulence Factors , Cronobacter sakazakii/genetics , Cronobacter sakazakii/pathogenicity , Virulence Factors/genetics , Biofilms/growth & development , Humans , Mice , Virulence/genetics , Caco-2 Cells , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , RAW 264.7 Cells , Bacterial Adhesion/genetics , Stress, Physiological/genetics , Gene Expression Regulation, Bacterial , Food Microbiology
14.
Biosens Bioelectron ; 261: 116491, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38879900

ABSTRACT

As one class of molecular imprinted polymers (MIPs), surface imprinted polymer (SIP)-based biosensors show great potential in direct whole-bacteria detection. Micro-contact imprinting, that involves stamping the template bacteria immobilized on a substrate into a pre-polymerized polymer matrix, is the most straightforward and prominent method to obtain SIP-based biosensors. However, the major drawbacks of the method arise from the requirement for fresh template bacteria and often non-reproducible bacteria distribution on the stamp substrate. Herein, we developed a positive master stamp containing photolithographic mimics of the template bacteria (E. coli) enabling reproducible fabrication of biomimetic SIP-based biosensors without the need for the "real" bacteria cells. By using atomic force and scanning electron microscopy imaging techniques, respectively, the E. coli-capturing ability of the SIP samples was tested, and compared with non-imprinted polymer (NIP)-based samples and control SIP samples, in which the cavity geometry does not match with E. coli cells. It was revealed that the presence of the biomimetic E. coli imprints with a specifically designed geometry increases the sensor E. coli-capturing ability by an "imprinting factor" of about 3. These findings show the importance of geometry-guided physical recognition in bacterial detection using SIP-based biosensors. In addition, this imprinting strategy was employed to interdigitated electrodes and QCM (quartz crystal microbalance) chips. E. coli detection performance of the sensors was demonstrated with electrochemical impedance spectroscopy (EIS) and QCM measurements with dissipation monitoring technique (QCM-D).


Subject(s)
Bacterial Adhesion , Biosensing Techniques , Escherichia coli , Polymers , Escherichia coli/isolation & purification , Polymers/chemistry , Molecular Imprinting/methods , Surface Properties , Molecularly Imprinted Polymers/chemistry , Escherichia coli Infections/microbiology , Biomimetic Materials/chemistry
15.
Food Res Int ; 190: 114582, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945602

ABSTRACT

Infants have digestive environments that are more favorable for microbial proliferation and subsequent endogenous nitrite production than those of adults, but direct evidence of this has been lacking. In this study, we propose a novel epidemiology of infant methemoglobinemia by demonstrating the risk posed by nitrite-producers in the gastrointestinal tract. Nitrite-producers from vegetables (n = 323) were exposed to stress factors of the gastrointestinal environment (gastric pH, intestinal bile salts, anaerobic atmosphere) reflecting 4 different postnatal age periods (Neonate, ≤1 month; Infant A, 1-3 months; Infant B, 3-6 months; Infant C, 6-12 months). "High-risk" strains with a nitrate-to-nitrite conversion rate of ≥1.3 %, the minimum rate corresponding to nitrite overproduction, under the Neonate stress condition were analyzed for intestinal adhesion. Among all the phyla, Pseudomonadota achieved the highest survival (P < 0.05; survival rate of 51.3-71.8 %). Possible cross-protection against bile resistance due to acid shock was observed for all the phyla. All the high-risk strains exhibited moderate autoaggregation (14.0-36.4 %), whereas only a few exhibited satisfactory surface hydrophobicity (>40 %). The Pantoea agglomerans strain strongly adhered to Caco-2 cells (7.4 ± 1.1 %). This study showed the ability of the Pantoea, Enterobacter, and Klebsiella strains to survive under gastrointestinal stress for ≤12 months, to excessively produce nitrite under neonatal stress conditions, and to settle in the human intestine. To our knowledge, this is the first study to reveal the role of the natural flora of vegetables in the epidemiology of infant methemoglobinemia through a multilateral approach.


Subject(s)
Methemoglobinemia , Nitrites , Vegetables , Humans , Vegetables/microbiology , Infant , Methemoglobinemia/metabolism , Nitrites/metabolism , Infant, Newborn , Bacterial Adhesion , Bile Acids and Salts/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Hydrogen-Ion Concentration , Gastrointestinal Microbiome
16.
Arch Microbiol ; 206(7): 322, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38907754

ABSTRACT

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.


Subject(s)
Bacterial Adhesion , Limosilactobacillus reuteri , Peptides , Probiotics , Soybean Proteins , Limosilactobacillus reuteri/growth & development , Limosilactobacillus reuteri/metabolism , Soybean Proteins/pharmacology , Soybean Proteins/metabolism , Soybean Proteins/chemistry , Humans , Caco-2 Cells , Bacterial Adhesion/drug effects , Peptides/pharmacology , Prebiotics , Gastrointestinal Microbiome/drug effects , Glycine max/microbiology
17.
Sci Total Environ ; 944: 173889, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-38876335

ABSTRACT

The transport and retention of bacteria in porous media, such as aquifer, are governed by the solid-liquid interface characteristics and bacterial mobility. The secretion of extracellular polymeric substance (EPS) by bacteria modifies their surface property, and thereby has effects on their adhesion to surface. The role of EPS in bacterial mobility within saturated quartz sand media is uncertain, as both promoting and inhibitory effects have been reported, and underlying mechanisms remain unclear. In this study, the effects of EPS on bacterial transport behavior and possible underlying mechanism were investigated at 4 concentrations (0 mg L-1, 50 mg L-1, 200 mg L-1 and 1000 mg L-1) using laboratory simulation experiments in conjunction with Extend Derjaguin-Landau-Verweu-Overbeek (XDLVO) modeling. The results showed that EPS facilitated bacterial mobility at all tested concentrations. It could be partially explained by the increased energy barrier between bacterial cells and quartz sand surface in the presence of EPS. The XDLVO sphere-plate model predicted that EPS induced a higher electrostatic double layer (EDL) repulsive force, Lewis acid-base (AB) and steric stabilization (ST), as well as a lower Lifshitz-van der Waals (LW) attractive force. However, at the highest EPS concentration (1000 mg L-1), the promotion of EPS on bacterial mobility weakened as a result of lower repulsive interactions between cells, which was supported by observed enhanced bacterial aggregation. Consequently, the increased aggregation led to greater bio-colloidal straining and ripening in the sand column, weakening the positive impact of EPS on bacterial transport. These findings suggested that EPS exhibited concentration-dependent effects on bacterial surface properties and transport behavior and revealed non-intuitive dual effects of EPS on those processes.


Subject(s)
Bacteria , Extracellular Polymeric Substance Matrix , Porosity , Bacteria/metabolism , Surface Properties , Groundwater/chemistry , Bacterial Adhesion
18.
Curr Biol ; 34(11): R539-R541, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38834027

ABSTRACT

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.


Subject(s)
Fimbriae, Bacterial , Vibrio cholerae , Vibrio cholerae/physiology , Vibrio cholerae/genetics , Fimbriae, Bacterial/physiology , Fimbriae, Bacterial/metabolism , Bacterial Adhesion/physiology
19.
Vet Res ; 55(1): 70, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822378

ABSTRACT

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.


Subject(s)
Adhesins, Escherichia coli , Bacterial Adhesion , Chickens , Escherichia coli Infections , Poultry Diseases , Poultry Diseases/microbiology , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Animals , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Escherichia coli/physiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
20.
Arch Microbiol ; 206(7): 289, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847838

ABSTRACT

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


Subject(s)
Anti-Bacterial Agents , Biofilms , Diclofenac , Staphylococcus epidermidis , Biofilms/drug effects , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/physiology , Diclofenac/pharmacology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Adhesion/drug effects , Humans , Polysaccharides, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Gene Expression Regulation, Bacterial/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...