Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 830
Filter
1.
Sci Rep ; 14(1): 21269, 2024 09 11.
Article in English | MEDLINE | ID: mdl-39261496

ABSTRACT

Staphylococcus aureus infections are hard to treat due to the emergence of antibiotic resistant strains, as well as their ability to form biofilms. The MazEF toxin-antitoxin system is thought play a role in bacterial biofilm phenotype as well as antibiotic resistance. In S. aureus, the physiologic function of the mazEF gene in the disease transition from acute to chronic infection is not well understood. In methicillin resistant S. aureus (MRSA), loss of mazF expression results in loss of resistance to first generation cephalosporins. mazF::tn displayed sensitivity while the isogenic wild type (WT) remained resistant. mazF::tn displayed significantly increased growth of biofilms on metal implants over 48 h compared to WT and the complemented transposon mutant. mazF::tn biofilms displayed significantly decreased antibiotic tolerance to vancomycin and cefazolin in comparison to WT and complement biofilms. Mice given mazF::tn in a sepsis model displayed less abscess burden and increased survival (100%) when treated with cefazolin compared to WT bacteremia treated with cefazolin (20%). mazF::tn periprosthetic joint infections displayed increased biofilm burden at acute time points and decreased biofilm burden at chronic time points. Our data suggests MazEF in MRSA is responsible for controlling growth of biofilms, antibiotic tolerance, and influence chronic infections in vivo.


Subject(s)
Anti-Bacterial Agents , Biofilms , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Animals , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Mice , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbial Sensitivity Tests , Disease Models, Animal , Vancomycin/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Cefazolin/pharmacology , Female
2.
BMC Res Notes ; 17(1): 252, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252055

ABSTRACT

OBJECTIVE: The purpose of this study is a new update on the resistance profile, Macrolide-Lincosamide-Streptogramin B resistance mechanisms and biofilm formation in the Staphylococcus aureus isolated from health care workers (HCWs) nasal carriage at a children's teaching hospital in Babol (Northern Iran). RESULTS: A total of 143 non-repetitive nasal swab samples were collected from volunteers, where 53.8% (n; 77/143) were HCWs, 33.6% (n; 48/143) medical students, and 12.6% (n; 18/143) resident students. The prevalence of nasal carriers of S. aureus was 22.4% (n; 32/143), among them, 40.6% (n; 13/32) were identified as methicillin-resistant Staphylococcus aureus (MRSA( carriers. Antimicrobial susceptibility testing showed that erythromycin (68.8%, n; 22/32) and ciprofloxacin (15.6%, n; 5/32) had the highest and lowest resistance rate, respectively. The frequency of resistance genes in the strains was as follows; ermC (n; 17/32, 53.1%), ermA (n; 11/32, 34.4%), ermB (n; 6/32, 18.7%), ereA (n; 3/32, 9.4%). Moreover, 50.0% (n; 16/32), 28.1% (n; 9/32) and 21.8% (n; 7/32) of isolates were strongly, weakly and moderately biofilm producer, respectively. Macrolides-lincosamides-streptogramins B (MLSB) antibiotic resistance among S. aureus isolates from HCWs nasal carriage have found significant prevalence rates throughout the globe. It is crucial to remember that the development of biofilms and MLS B antibiotic resistance are both dynamic processes.


Subject(s)
Anti-Bacterial Agents , Biofilms , Carrier State , Clindamycin , Health Personnel , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Staphylococcal Infections , Staphylococcus aureus , Humans , Biofilms/drug effects , Biofilms/growth & development , Clindamycin/pharmacology , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/physiology , Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/epidemiology , Carrier State/microbiology , Iran , Male , Adult , Female , Erythromycin/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Drug Resistance, Bacterial/genetics
3.
Ecotoxicol Environ Saf ; 283: 116853, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39137468

ABSTRACT

The effect and underlying mechanism of tetrabromobisphenol A (TBBPA), a plastic additive, on biofilm formation of methicillin-resistant Staphylococcus aureus (MRSA USA300) remain unknown. This study first investigated the impact of different concentrations of TBBPA on the growth and biofilm formation of USA300. The results indicated that a low concentration (0.5 mg/L) of TBBPA promoted the growth and biofilm formation of USA300, whereas high concentrations (5 mg/L and 10 mg/L) of TBBPA had inhibitory effects. Further exploration revealed that the low concentration of TBBPA enhance biofilm formation by promoting the synthesis of extracellular proteins, release of extracellular DNA (eDNA), and production of staphyloxanthin. RTqPCR analysis demonstrated that the low concentration of TBBPA upregulated genes associated with extracellular protein synthesis (sarA, fnbA, fnbB, aur) and eDNA formation (atlA) and increased the expression of genes involved in staphyloxanthin biosynthesis (crtM), suggesting a potential mechanism for enhanced resistance of USA300 to adverse conditions. These findings shed light on how low concentrations of TBBPA facilitate biofilm formation in USA300 and highlight the indirect impact of plastic additives on pathogenic bacteria in terms of human health. In the future, in-depth studies about effects of plastic additives on pathogenicity of pathogenic bacteria should be conducted. CAPSULE: The protein and eDNA contents in biofilms of methicillin-resistant Staphylococcus aureus are increased by low concentrations of TBBPA.


Subject(s)
Biofilms , Methicillin-Resistant Staphylococcus aureus , Polybrominated Biphenyls , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Polybrominated Biphenyls/toxicity , Xanthophylls , Bacterial Proteins/genetics
4.
PLoS One ; 19(8): e0308282, 2024.
Article in English | MEDLINE | ID: mdl-39102390

ABSTRACT

Biofilm development significantly enhances the virulence of methicillin-resistant Staphylococcus aureus (MRSA), leading to severe infections and decreased susceptibility to antibiotics, especially in strains associated with hospital environments. This study examined the occurrence of MRSA, their ability to form biofilms, agr typing, and the antibiotic resistance profiles of biofilm-forming MRSA strains isolated from environmental surfaces at Mymensingh Medical College Hospital (MMCH). From 120 swab samples, 86 (71.67%) tested positive for S. aureus. MRSA was identified in 86 isolates using the disk diffusion technique, and by polymerase chain reaction (PCR), 56 (65.1%) isolates were confirmed to carry the mecA gene. The Crystal Violet Microtiter Plate (CVMP) test revealed that 80.35% (45 isolates) were biofilm-forming and 19.6% (11 isolates) were non-biofilm-forming. Out of 45 biofilm producer isolates 37.5% and 42.9% isolates exhibited strong and intermediate biofilm-forming characteristics, respectively. Molecular analysis revealed that 17.78% of MRSA isolates carried at least one gene related to biofilm formation, specifically icaA, icaB, and icaD genes were discovered in 13.33%, 8.89%, 6.67% of the MRSA isolates, respectively. In agr typing, the most prevalent group was agr I (71.11%), followed by group III (17.78%) and group II (11.11%). Group IV was not detected. The distribution of agr gene groups showed a significant difference among biofilm-forming isolates (p < 0.05). In agr group I, 18.75% of isolates carried the icaA gene, 12.5% carried the icaB gene, and 9.37% carried the icaD gene. Biofilm-forming genes were not detected in any of the isolates from agr groups II or III. There are no statistically significant differences between agr groups and the presence of these genes (p > 0.05). Antibiotic resistance varied significantly among agr groups, with agr group I displaying the highest resistance, agr group II, and agr group III exhibiting the least resistance (p < 0.05). Seventy-three (73.3%) of the isolates were multi-drug resistant, with agr group I displaying nineteen MDR patterns. The occurrence of MRSA in hospital environments and their capacity to form biofilm raises concerns for public health. These findings support the importance of further research focused on agr quorum sensing systems as a basis for developing novel antibacterial agents.


Subject(s)
Bacterial Proteins , Biofilms , Methicillin-Resistant Staphylococcus aureus , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/physiology , Bacterial Proteins/genetics , Humans , Anti-Bacterial Agents/pharmacology , Hospitals , Microbial Sensitivity Tests , Trans-Activators/genetics , Staphylococcal Infections/microbiology
5.
Microb Pathog ; 194: 106838, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39111368

ABSTRACT

Staphylococcus aureus (S. aureus), a versatile Gram-positive bacterium, is implicated in a spectrum of infections, and its resilience is often attributed to biofilm formation. This study investigates the effect of sub-inhibitory doses of oxacillin on biofilm formation by methicillin-resistant S. aureus (MRSA). Specifically, it examines how these doses influence biofilms' development, maturation, and dispersal. The biofilm's zenith reached 48 h of incubation, followed by a noteworthy decline at 96 h and a distinctive clearance zone around biofilm-positive cells exposed to oxacillin. Scanning electron micrographs unveiled an intriguing active biofilm dispersal mechanism, a rarity in this species. Among 180 isolates, only three carrying the elusive icaD gene exhibited this phenomenon. icaD gene was absent in their counterparts. Notably, the icaD gene emerges as a distinctive marker, crucial in regulating biofilm dispersion and setting these isolates apart. The captivating interplay of oxacillin, biofilm dynamics, and genetic signatures disintegrate novel dimensions in understanding MRSA's adaptive strategies and underscores the importance of the icaD gene in engineering biofilm resilience.


Subject(s)
Anti-Bacterial Agents , Biofilms , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Oxacillin , Biofilms/drug effects , Biofilms/growth & development , Oxacillin/pharmacology , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcal Infections/microbiology , Microscopy, Electron, Scanning , Humans , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology
6.
BMC Microbiol ; 24(1): 241, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961344

ABSTRACT

BACKGROUND: Staphylococcus aureus, a commensal bacterium, colonizes the skin and mucous membranes of approximately 30% of the human population. Apart from conventional resistance mechanisms, one of the pathogenic features of S. aureus is its ability to survive in a biofilm state on both biotic and abiotic surfaces. Due to this characteristic, S. aureus is a major cause of human infections, with Methicillin-Resistant Staphylococcus aureus (MRSA) being a significant contributor to both community-acquired and hospital-acquired infections. RESULTS: Analyzing non-repetitive clinical isolates of MRSA collected from seven provinces and cities in China between 2014 and 2020, it was observed that 53.2% of the MRSA isolates exhibited varying degrees of ability to produce biofilm. The biofilm positivity rate was notably high in MRSA isolates from Guangdong, Jiangxi, and Hubei. The predominant MRSA strains collected in this study were of sequence types ST59, ST5, and ST239, with the biofilm-producing capability mainly distributed among moderate and weak biofilm producers within these ST types. Notably, certain sequence types, such as ST88, exhibited a high prevalence of strong biofilm-producing strains. The study found that SCCmec IV was the predominant type among biofilm-positive MRSA, followed by SCCmec II. Comparing strains with weak and strong biofilm production capabilities, the positive rates of the sdrD and sdrE were higher in strong biofilm producers. The genetic determinants ebp, icaA, icaB, icaC, icaD, icaR, and sdrE were associated with strong biofilm production in MRSA. Additionally, biofilm-negative MRSA isolates showed higher sensitivity rates to cefalotin (94.8%), daptomycin (94.5%), mupirocin (86.5%), teicoplanin (94.5%), fusidic acid (81.0%), and dalbavancin (94.5%) compared to biofilm-positive MRSA isolates. The biofilm positivity rate was consistently above 50% in all collected specimen types. CONCLUSIONS: MRSA strains with biofilm production capability warrant increased vigilance.


Subject(s)
Biofilms , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/physiology , China/epidemiology , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Genes, Bacterial/genetics , Humans
7.
ACS Appl Mater Interfaces ; 16(29): 37795-37805, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39008846

ABSTRACT

Biofilm-mediated wound infections pose a significant challenge due to the limitations of conventional antibiotics, which often exhibit narrow-spectrum activity, fail to eliminate recurrent bacterial contamination, and are unable to penetrate the biofilm matrix. While the search for alternatives has explored the use of metal nanoparticles and synthetic biocides, these solutions often suffer from unintended toxicity to surrounding tissues and lack controlled administration and release. In this study, we engineered a pH-responsive release-active dressing film based on carboxymethyl cellulose, incorporating a synthetic antibacterial molecule (SAM-17). The dressing film exhibited optimal mechanical stability for easy application and demonstrated excellent fluid absorption properties, allowing for prolonged moisturization at the site of injury. The film exhibited pH-dependent release of cargo, with 78% release within 24 h at acidic pH, enabling targeted antibacterial drug delivery within the wound microenvironment. Furthermore, the release-active film effectively eliminated repeated challenges of bacterial contamination. Remarkably, the film demonstrated a minimal toxicity profile in both in vitro and in vivo models. The film eliminated preformed bacterial biofilms, achieving a reduction of 2.5 log against methicillin-resistant Staphylococcus aureus (MRSA) and 4.1 log against vancomycin-resistant S. aureus (VRSA). In a biofilm-mediated MRSA wound infection model, this release-active film eradicated the biofilm-embedded bacteria by over 99%, resulting in accelerated wound healing. These findings highlight the potential of this film as an effective candidate for tackling biofilm-associated wound infections.


Subject(s)
Anti-Bacterial Agents , Bandages , Biofilms , Methicillin-Resistant Staphylococcus aureus , Wound Infection , Biofilms/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Wound Infection/drug therapy , Wound Infection/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Animals , Mice , Hydrogen-Ion Concentration , Staphylococcal Infections/drug therapy , Microbial Sensitivity Tests , Humans , Carboxymethylcellulose Sodium/chemistry , Carboxymethylcellulose Sodium/pharmacology
8.
BMC Biotechnol ; 24(1): 47, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978013

ABSTRACT

The threat of methicillin-resistant Staphylococcus aureus (MRSA) is increasing worldwide, making it significantly necessary to discover a novel way of dealing with related infections. The quick spread of MRSA isolates among infected individuals has heightened public health concerns and significantly limited treatment options. Vancomycin (VAN) can be applied to treat severe MRSA infections, and the indiscriminate administration of this antimicrobial agent has caused several concerns in medical settings. Owing to several advantageous characteristics, a niosomal drug delivery system may increase the potential of loaded antimicrobial agents. This work aims to examine the antibacterial and anti-biofilm properties of VAN-niosome against MRSA clinical isolates with emphasis on cytotoxicity and stability studies. Furthermore, we aim to suggest an effective approach against MRSA infections by investigating the inhibitory effect of formulated niosome on the expression of the biofilm-associated gene (icaR). The thin-film hydration approach was used to prepare the niosome (Tween 60, Span 60, and cholesterol), and field emission scanning electron microscopy (FE-SEM), an in vitro drug release, dynamic light scattering (DLS), and entrapment efficiency (EE%) were used to investigate the physicochemical properties. The physical stability of VAN-niosome, including hydrodynamic size, polydispersity index (PDI), and EE%, was analyzed for a 30-day storage time at 4 °C and 25 °C. In addition, the human foreskin fibroblast (HFF) cell line was used to evaluate the cytotoxic effect of synthesized niosome. Moreover, minimum inhibitory and bactericidal concentrations (MICs/MBCs) were applied to assess the antibacterial properties of niosomal VAN formulation. Also, the antibiofilm potential of VAN-niosome was investigated by microtiter plate (MTP) and real-time PCR methods. The FE-SEM result revealed that synthesized VAN-niosome had a spherical morphology. The hydrodynamic size and PDI of VAN-niosome reported by the DLS method were 201.2 nm and 0.301, respectively. Also, the surface zeta charge of the prepared niosome was - 35.4 mV, and the EE% ranged between 58.9 and 62.5%. Moreover, in vitro release study revealed a sustained-release profile for synthesized niosomal formulation. Our study showed that VAN-niosome had acceptable stability during a 30-day storage time. Additionally, the VAN-niosome had stronger antibacterial and anti-biofilm properties against MRSA clinical isolates compared with free VAN. In conclusion, the result of our study demonstrated that niosomal VAN could be promising as a successful drug delivery system due to sustained drug release, negligible toxicity, and high encapsulation capacity. Also, the antibacterial and anti-biofilm studies showed the high capacity of VAN-niosome against MRSA clinical isolates. Furthermore, the results of real-time PCR exhibited that VAN-niosome could be proposed as a powerful strategy against MRSA biofilm via down-regulation of icaR gene expression.


Subject(s)
Anti-Bacterial Agents , Biofilms , Drug Delivery Systems , Liposomes , Methicillin-Resistant Staphylococcus aureus , Vancomycin , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Vancomycin/pharmacology , Vancomycin/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Liposomes/chemistry , Humans , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Drug Liberation
9.
Biofouling ; 40(8): 483-498, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39069795

ABSTRACT

Using existing adrentimicrobials with essential oil components to prevent antimicrobial resistance is an alternative strategy. This study aimed to evaluate the resistance status, synergistic combinations, and in vitro biofilm formation activities of clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA), Stenotrophomonas maltophilia and Candida albicans against antimicrobial agents and cinnamaldehyde, carvacrol, eugenol, limonene and eucalyptol. Antimicrobial activities were evaluated by microdilution, cytotoxicity by XTT, synergy by checkerboard and time-kill, and biofilm inhibition by microplate methods. Cinnamaldehyde and carvacrol showed strong antimicrobial activity. Synergistic effects were observed when using all essential oils with antimicrobials. Only two C. albicans isolates showed antagonism with cinnamaldehyde and fluconazole. The constituents showed cytotoxic effects in the L929 cell line (except limonene). A time-kill analysis revealed a bacteriostatic effect on S. maltophilia and MRSA isolates and a fungicidal effect on C. albicans isolates. These results are important for further research to improve antimicrobial efficacy or to develop new agents.


Subject(s)
Anti-Infective Agents , Biofilms , Candida albicans , Drug Synergism , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Oils, Volatile , Stenotrophomonas maltophilia , Biofilms/drug effects , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Candida albicans/drug effects , Candida albicans/physiology , Stenotrophomonas maltophilia/drug effects , Stenotrophomonas maltophilia/physiology , Anti-Infective Agents/pharmacology , Limonene/pharmacology , Acrolein/analogs & derivatives , Acrolein/pharmacology , Cymenes/pharmacology , Cell Line , Monoterpenes/pharmacology , Anti-Bacterial Agents/pharmacology , Terpenes/pharmacology , Eucalyptol/pharmacology , Eugenol/pharmacology , Cyclohexenes/pharmacology , Mice
10.
J Appl Microbiol ; 135(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39066496

ABSTRACT

AIMS: Staphylococcus aureus is an opportunistic pathogen whose treatment is further complicated by its ability to form biofilms. In this study, we examine the impact of growing S. aureus biofilms on different polymerizing surfaces, specifically agar and agarose, on the pathogen's tolerance to fluoroquinolones. METHODS AND RESULTS: Biofilms of two methicillin-resistant strains of S. aureus were grown on agar or agarose in the presence of the same added nutrients, and their antibiotic susceptibility to two fluoroquinolones, moxifloxacin (MXF) and delafloxacin (DLX), were measured. We also compared the metabolism and extracellular polymeric substances (EPS) production of biofilms that were grown on agar and agarose. CONCLUSIONS: Biofilms that were grown on agarose were consistently more susceptible to antibiotics than those grown on agar. We found that in biofilms that were grown on agar, extracellular protein composition was higher, and adding EPS to agarose-grown biofilms increased their tolerance to DLX to levels that were comparable to agar-grown biofilms.


Subject(s)
Agar , Anti-Bacterial Agents , Biofilms , Fluoroquinolones , Microbial Sensitivity Tests , Sepharose , Staphylococcus aureus , Biofilms/drug effects , Biofilms/growth & development , Fluoroquinolones/pharmacology , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Staphylococcus aureus/growth & development , Culture Media/chemistry , Moxifloxacin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology
11.
Curr Microbiol ; 81(9): 272, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014046

ABSTRACT

Biofilms formed by Pseudomonas aeruginosa and Staphylococcus aureus, along with their antibiotic tolerance have posed challenges to treatment strategies for lung, wound, and other infections, particularly when co-infecting. In the present study, the inhibitory effect of xylitol on biofilm formation, as well as its eradication potential on pre-established biofilms formed by P. aeruginosa strain PAO1, methicillin-resistant S. aureus, and a mix of both species in an alginate bead model were tested. Xylitol concentrations of 2, 1, and 0.5 M reduced biofilm formation by P. aeruginosa strain PAO1, methicillin-resistant S. aureus, and the mixed-species biofilm in a concentration-dependent manner. Additionally, biofilms formed by these species were subjected to treatment with xylitol. Xylitol was also capable of eradicating biofilms established by P. aeruginosa strain PAO1, methicillin-resistant S. aureus, and the mixed-species biofilm by at least 20%, with the most effective eradication observed for P. aeruginosa strain PAO1. The present study indicates the effectiveness of xylitol as both an inhibitory and eradicating agent against biofilms formed by P. aeruginosa strain PAO1, methicillin-resistant S. aureus, and a mix of both species in an alginate bead model, which mimics the in vivo characteristics of P. aeruginosa aggregates.


Subject(s)
Alginates , Anti-Bacterial Agents , Biofilms , Methicillin-Resistant Staphylococcus aureus , Pseudomonas aeruginosa , Xylitol , Biofilms/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Alginates/pharmacology , Xylitol/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology
12.
PLoS One ; 19(6): e0296140, 2024.
Article in English | MEDLINE | ID: mdl-38900759

ABSTRACT

Implant-associated osteomyelitis remains a major orthopaedic problem. As neutrophil swarming to the surgical site is a critical host response to prevent infection, visualization and quantification of this dynamic behavior at the native microenvironment of infection will elucidate previously unrecognized mechanisms central to understanding the host response. We recently developed longitudinal intravital imaging of the bone marrow (LIMB) to visualize host cells and fluorescent S. aureus on a contaminated transfemoral implant in live mice, which allows for direct visualization of bacteria colonization of the implant and host cellular responses using two-photon laser scanning microscopy. To the end of rigorous and reproducible quantitative outcomes of neutrophil swarming kinetics in this model, we developed a protocol for robust segmentation, tracking, and quantifications of neutrophil dynamics adapted from Trainable Weka Segmentation and TrackMate, two readily available Fiji/ImageJ plugins. In this work, Catchup mice with tdTomato expressing neutrophils received a transfemoral pin with or without ECFP/EGFP-expressing USA300 methicillin-resistant Staphylococcus aureus (MRSA) to obtain 30-minute LIMB videos at 2-, 4-, and 6-hours post-implantation. The developed semi-automated neutrophil tracking protocol was executed independently by two users to quantify the distance, displacement, speed, velocity, and directionality of the target cells. The results revealed high inter-user reliability for all outcomes (ICC > 0.96; p > 0.05). Consistent with the established paradigm on increased neutrophil swarming during active infection, the results also demonstrated increased neutrophil speed and velocity at all measured time points, and increased displacement at later time points (6 hours) in infected versus uninfected mice (p < 0.05). Neutrophils and bacteria also exhibit directionality during migration in the infected mice. The semi-automated cell tracking protocol provides a streamlined approach to robustly identify and track individual cells across diverse experimental settings and eliminates inter-observer variability.


Subject(s)
Cell Tracking , Femur , Neutrophils , Animals , Neutrophils/immunology , Mice , Femur/microbiology , Cell Tracking/methods , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Disease Models, Animal , Osteomyelitis/microbiology , Methicillin-Resistant Staphylococcus aureus/physiology , Prosthesis-Related Infections/microbiology , Prostheses and Implants/microbiology , Staphylococcus aureus/physiology , Female
13.
Arch Microbiol ; 206(7): 301, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874781

ABSTRACT

Biofilm formation by methicillin-resistant Staphylococcus aureus (MRSA) on indwelling medical devices complicates the treatment of infection. Tetrabromobisphenol A (TBBPA), a synthetic, lipophilic, halogenated aromatic compound widely used as an additive in plastics and electronic products, has raised environmental concerns due to its potential for bioaccumulation. This study investigated the impact of sub-inhibitory concentrations of TBBPA on MRSA biofilm formation. Crystal violet staining and confocal laser scanning microscopy analysis demonstrated that 1/8 MIC (0.5 µg/mL) of TBBPA significantly stimulated MRSA biofilm formation (P < 0.0001). MTT assays indicated that the metabolic activity within the biofilms increased by 15.60-40.85% compared to untreated controls. Dot blot immunoassay, autolysis assay, and extracellular DNA (eDNA) quantification further revealed TBBPA enhanced the production of polysaccharide intercellular adhesin (PIA) and eDNA, which are key biofilm components. Additionally, TBBPA was found to enhance the production of staphyloxanthin, facilitating MRSA survival under oxidative conditions and in human whole blood. RT-qPCR analysis showed that TBBPA significantly upregulated genes associated with biofilm formation (icaA, atlA, sarA), staphyloxanthin biosynthesis (crtM and sigB), and oxidative stress responses (sodA and katA). These findings suggest that TBBPA promotes MRSA biofilm development and enhances bacterial resistance to adverse conditions, thereby potentially exacerbating risks to human health.


Subject(s)
Biofilms , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Polybrominated Biphenyls , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Polybrominated Biphenyls/pharmacology , Humans , Xanthophylls/metabolism , Xanthophylls/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects
14.
Macromol Rapid Commun ; 45(16): e2400170, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936823

ABSTRACT

A challenge remains in the development of anti-infectious coatings for the inert surfaces of biomedical devices that are prone to bacterial colonization and biofilm formation. Here, a facile photocuring method to construct functionalized polymeric coatings on inert polydimethylsiloxane (PDMS) surfaces, is developed. Using atom transfer radical polymerization (ATRP) initiator bearing thymol group, hydrophilic DMAEMA and benzophenone (BP)-containing monomers are copolymerized to form polymers with end functional groups. An end-functionalized biocidal coating is then constructed on the inert PDMS surface in one step using a photocuring reaction. The functionalized PDMS surfaces show excellent antibacterial and antifouling properties, are capable of completely eradiating MRSA within ≈6 h, and effectively inhibit the growth of biofilms. In addition, they have good stability and long-lasting antibacterial activity in body fluid environments such as 0.9% saline and urine. According to bladder model experiments, the catheter's lifespan can be extended from ≈7 to 35 days by inhibiting the growth and migration of bacteria along its inner surface. The photocuring technique is therefore very promising in terms of surface functionalization of inert biomedical devices in order to minimize the spread of infection.


Subject(s)
Anti-Bacterial Agents , Biofilms , Dimethylpolysiloxanes , Surface Properties , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Dimethylpolysiloxanes/chemistry , Biofilms/drug effects , Microbial Sensitivity Tests , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Polymers/chemistry , Polymers/pharmacology , Photochemical Processes , Benzophenones/chemistry , Benzophenones/pharmacology , Polymerization , Molecular Structure
15.
Acta Biomater ; 184: 313-322, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38897337

ABSTRACT

Antimicrobial phototherapy has gained recognition as a promising approach for addressing bacterial biofilms, however, its effectiveness is often impeded by the robust physical and chemical defenses of the biofilms. Traditional antibacterial nanoplatforms face challenges in breaching the extracellular polymeric substances barrier to efficiently deliver photosensitizers deep into biofilms. Moreover, the prevalent hypoxia within biofilms restricts the success of oxygen-reliant phototherapy. In this study, we engineered a soft mesoporous organosilica nanoplatform (SMONs) by incorporating polyethylene glycol (PEG), catalase (CAT), and indocyanine green (ICG), forming SMONs-PEG-CAT-ICG (SPCI). We compared the antimicrobial efficacy of SPCI with more rigid nanoplatforms. Our results demonstrated that unique flexible mechanical properties of SPCI enable it to navigate through biofilm barriers, markedly enhancing ICG penetration in methicillin-resistant Staphylococcus aureus (MRSA) biofilms. Notably, in a murine subcutaneous MRSA biofilm infection model, SPCI showed superior biofilm penetration and pharmacokinetic benefits over its rigid counterparts. The embedded catalase in SPCI effectively converts excess H2O2 present in infected tissues into O2, alleviating hypoxia and significantly boosting the antibacterial performance of phototherapy. Both in vitro and in vivo experiments confirmed that SPCI surpasses traditional rigid nanoplatforms in overcoming biofilm barriers, offering improved treatment outcomes for infections associated with bacterial biofilms. This study presents a viable strategy for managing bacterial biofilm-induced diseases by leveraging the unique attributes of a soft mesoporous organosilica-based nanoplatform. STATEMENT OF SIGNIFICANCE: This research introduces an innovative antimicrobial phototherapy soft nanoplatform that overcomes the inherent limitations posed by the protective barriers of bacterial biofilms. By soft nanoplatform with flexible mechanical properties, we enhance the penetration and delivery of photosensitizers into biofilms. The inclusion of catalase within this soft nanoplatform addresses the hypoxia in biofilms by converting hydrogen peroxide into oxygen in infected tissues, thereby amplifying the antibacterial effectiveness of phototherapy. Compared to traditional rigid nanoplatforms, this flexible nanoplatform not only promotes the delivery of therapeutic agents but also sets a new direction for treating bacterial biofilm infections, offering significant implications for future antimicrobial therapies.


Subject(s)
Anti-Bacterial Agents , Biofilms , Catalase , Indocyanine Green , Photosensitizing Agents , Biofilms/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/chemistry , Catalase/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Phototherapy , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Nanoparticles/chemistry , Staphylococcal Infections/drug therapy , Permeability , Female , Mice, Inbred BALB C
16.
Colloids Surf B Biointerfaces ; 239: 113940, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744081

ABSTRACT

AIM: The challenging properties of biofilm-associated infections and the rise of multidrug-resistant bacteria are prompting the exploration of alternative treatment options. This study investigates the efficacy of different bioactive glass (BAG) formulations - alone or combined with vancomycin - to eradicate biofilm. Further, we study the influence of BAG on pH and osmotic pressure as important factors limiting bacterial growth. METHOD: Different BAG S53P4 formulations were used for this study, including (a) powder (<45 µm), (b) granules (500-800 µm), (c) a cone-shaped scaffold and (d) two putty formulations containing granules with no powder (putty A) or with additional powder (putty B) bound together by a synthetic binder. Inert glass beads (1.0-1.3 mm) were included as control. All formulations were tested in a concentration of 1750 mg/ml in Müller-Hinton-Broth against methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-resistant Staphylococcus epidermidis (MRSE). Vancomycin was tested at the minimum-inhibitory concentration for each strain. Changes in pH and osmolality over time were assessed at 0 h, 24 h, 72 h and 168 h. RESULTS: All tested BAG formulations showed antibiofilm activity against MRSA and MRSE. Powder and putty B were the most effective formulations suppressing biofilm leading to its complete eradication after up to 168 h of co-incubation, followed by granules, scaffold and putty A. In general, MRSE appeared to be more susceptible to bioactive glass compared to MRSA. The addition of vancomycin had no substantial impact on biofilm eradication. We observed a positive correlation between a higher pH and higher antibiofilm activity. CONCLUSIONS: BAG S53P4 has demonstrated efficient biofilm antibiofilm activity against MRSA and MRSE, especially in powder-containing formulations, resulting in complete eradication of biofilm. Our data indicate neither remarkable increase nor decrease in antimicrobial efficacy with addition of vancomycin. Moreover, high pH appears to have a direct antimicrobial impact; the role of high osmolality needs further investigation.


Subject(s)
Anti-Bacterial Agents , Biofilms , Glass , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Osmotic Pressure , Staphylococcus epidermidis , Vancomycin , Biofilms/drug effects , Glass/chemistry , Hydrogen-Ion Concentration , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/physiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Vancomycin/pharmacology , Vancomycin/chemistry , Osmotic Pressure/drug effects , Drug Compounding
17.
mSphere ; 9(5): e0012624, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38695568

ABSTRACT

Biofilm formation is an important virulence factor for methicillin-resistant Staphylococcus aureus (MRSA). The extracellular matrix of MRSA biofilms contains significant amounts of double-stranded DNA that hold the biofilm together. MRSA cells secrete micrococcal nuclease (Nuc1), which degrades double-stranded DNA. In this study, we used standard methodologies to investigate the role of Nuc1 in MRSA biofilm formation and dispersal. We quantified biofilm formation and extracellular DNA (eDNA) levels in broth and agar cultures. In some experiments, cultures were supplemented with sub-MIC amoxicillin to induce biofilm formation. Biofilm erosion was quantitated by culturing biofilms on rods and enumerating detached colony-forming units (CFUs), and biofilm sloughing was investigated by perfusing biofilms cultured in glass tubes with fresh broth and measuring the sizes of the detached cell aggregates. We found that an MRSA nuc1- mutant strain produced significantly more biofilm and more eDNA than a wild-type strain, both in the absence and presence of sub-MIC amoxicillin. nuc1- mutant biofilms grown on rods detached significantly less than wild-type biofilms. Detachment was restored by exogenous DNase or complementing the nuc1- mutant. In the sloughing assay, nuc1- mutant biofilms released cell aggregates that were significantly larger than those released by wild-type biofilms. Our results suggest that Nuc1 modulates biofilm formation, biofilm detachment, and the sizes of detached cell aggregates. These processes may play a role in the spread and subsequent survival of MRSA biofilms during biofilm-related infections.IMPORTANCEInfections caused by antibiotic-resistant bacteria known as methicillin-resistant Staphylococcus aureus (MRSA) are a significant problem in hospitals. MRSA forms adherent biofilms on implanted medical devices such as catheters and breathing tubes. Bacteria can detach from biofilms on these devices and spread to other parts of the body such as the blood or lungs, where they can cause life-threatening infections. In this article, researchers show that MRSA secretes an enzyme known as thermonuclease that causes bacteria to detach from the biofilm. This is important because understanding the mechanism by which MRSA detaches from biofilms could lead to the development of procedures to mitigate the problem.


Subject(s)
Biofilms , Methicillin-Resistant Staphylococcus aureus , Micrococcal Nuclease , Biofilms/growth & development , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Micrococcal Nuclease/genetics , Micrococcal Nuclease/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/genetics , Virulence Factors/genetics , Microbial Sensitivity Tests , Amoxicillin/pharmacology
18.
J Med Microbiol ; 73(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38567642

ABSTRACT

Introduction. Staphylococcus aureus is the leading cause of acute medical implant infections, representing a significant modern medical concern. The success of S. aureus as a pathogen in these cases resides in its arsenal of virulence factors, resistance to multiple antimicrobials, mechanisms of immune modulation, and ability to rapidly form biofilms associated with implant surfaces. S. aureus device-associated, biofilm-mediated infections are often persistent and notoriously difficult to treat, skewing innate immune responses to promote chronic reoccurring infections. While relatively little is known of the role neutrophils play in response to acute S. aureus biofilm infections, these effector cells must be efficiently recruited to sites of infection via directed chemotaxis. Here we investigate the effects of modulating CXC chemokine receptor 2 (CXCR2) activity, predominantly expressed on neutrophils, during S. aureus implant-associated infection.Hypothesis. We hypothesize that modulation of CXCR2 expression and/or signalling activities during S. aureus infection, and thus neutrophil recruitment, extravasation and antimicrobial activity, will affect infection control and bacterial burdens in a mouse model of implant-associated infection.Aim. This investigation aims to elucidate the impact of altered CXCR2 activity during S. aureus biofilm-mediated infection that may help develop a framework for an effective novel strategy to prevent morbidity and mortality associated with implant infections.Methodology. To examine the role of CXCR2 during S. aureus implant infection, we employed a mouse model of indwelling subcutaneous catheter infection using a community-associated methicillin-resistant S. aureus (MRSA) strain. To assess the role of CXCR2 induction or inhibition during infection, treatment groups received daily intraperitoneal doses of either Lipocalin-2 (Lcn2) or AZD5069, respectively. At the end of the study, catheters and surrounding soft tissues were analysed for bacterial burdens and dissemination, and Cxcr2 transcription within the implant-associated tissues was quantified.Results. Mice treated with Lcn2 developed higher bacterial burdens within the soft tissue surrounding the implant site, which was associated with increased Cxcr2 expression. AZD5069 treatment also resulted in increased implant- and tissues-associated bacterial titres, as well as enhanced Cxcr2 expression.Conclusion. Our results demonstrate that CXCR2 plays an essential role in regulating the severity of S. aureus implant-associated infections. Interestingly, however, perturbation of CXCR2 expression or signalling both resulted in enhanced Cxcr2 transcription and elevated implant-associated bacterial burdens. Thus, CXCR2 appears finely tuned to efficiently recruit effector cells and mediate control of S. aureus biofilm-mediated infection.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Pyrimidines , Staphylococcal Infections , Sulfonamides , Mice , Animals , Staphylococcus aureus/physiology , Methicillin-Resistant Staphylococcus aureus/physiology , Receptors, Interleukin-8B/genetics , Staphylococcal Infections/microbiology , Biofilms
19.
Int J Nanomedicine ; 19: 3697-3714, 2024.
Article in English | MEDLINE | ID: mdl-38681091

ABSTRACT

Introduction: Over 75% of clinical microbiological infections are caused by bacterial biofilms that grow on wounds or implantable medical devices. This work describes the development of a new poly(diallyldimethylammonium chloride) (PDADMAC)/alginate-coated gold nanorod (GNR/Alg/PDADMAC) that effectively disintegrates the biofilms of Staphylococcus aureus (S. aureus), a prominent pathogen responsible for hospital-acquired infections. Methods: GNR was synthesised via seed-mediated growth method, and the resulting nanoparticles were coated first with Alg and then PDADMAC. FTIR, zeta potential, transmission electron microscopy, and UV-Vis spectrophotometry analysis were performed to characterise the nanoparticles. The efficacy and speed of the non-coated GNR and GNR/Alg/PDADMAC in disintegrating S. aureus-preformed biofilms, as well as their in vitro biocompatibility (L929 murine fibroblast) were then studied. Results: The synthesised GNR/Alg/PDADMAC (mean length: 55.71 ± 1.15 nm, mean width: 23.70 ± 1.13 nm, aspect ratio: 2.35) was biocompatible and potent in eradicating preformed biofilms of methicillin-resistant (MRSA) and methicillin-susceptible S. aureus (MSSA) when compared to triclosan, an antiseptic used for disinfecting S. aureus colonisation on abiotic surfaces in the hospital. The minimum biofilm eradication concentrations of GNR/Alg/PDADMAC (MBEC50 for MRSA biofilm = 0.029 nM; MBEC50 for MSSA biofilm = 0.032 nM) were significantly lower than those of triclosan (MBEC50 for MRSA biofilm = 10,784 nM; MBEC50 for MRSA biofilm 5967 nM). Moreover, GNR/Alg/PDADMAC was effective in eradicating 50% of MRSA and MSSA biofilms within 17 min when used at a low concentration (0.15 nM), similar to triclosan at a much higher concentration (50 µM). Disintegration of MRSA and MSSA biofilms was confirmed by field emission scanning electron microscopy and confocal laser scanning microscopy. Conclusion: These findings support the potential application of GNR/Alg/PDADMAC as an alternative agent to conventional antiseptics and antibiotics for the eradication of medically important MRSA and MSSA biofilms.


Subject(s)
Alginates , Anti-Bacterial Agents , Biofilms , Gold , Nanotubes , Polyethylenes , Quaternary Ammonium Compounds , Staphylococcus aureus , Biofilms/drug effects , Gold/chemistry , Gold/pharmacology , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Alginates/chemistry , Alginates/pharmacology , Nanotubes/chemistry , Animals , Mice , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Polyethylenes/chemistry , Polyethylenes/pharmacology , Staphylococcal Infections/drug therapy , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Cell Line , Microbial Sensitivity Tests , Metal Nanoparticles/chemistry
20.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 1-7, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678634

ABSTRACT

 Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main causes of community- and hospital-acquired infections. The expression of virulence genes in S. aureus is arranged by regulators like the accessory gene regulator (agr). The present study aims to estimate phenotypic characteristics of S. aureus and investigate the occurrence of agr genes with their correlation to biofilm formation. In this study, 34 MRSA strains out of 100 S. aureus isolates were recovered in a variety of clinical samples. Phenotypic characterization and ability of biofilm formation were assessed. About 8(24%) of isolates were biofilm producers. The percentages of biofilm production among isolates were 3(37.5%), 2(25%), 3(37.5%) as strong, moderate, and weak, respectively. Furthermore, the resistance rates for all antibiotics were higher in biofilm producers and 76% of the isolates were staphyloxanthin producers, around 82% of the strains showed resistance to H2O2. Hemolytic activity was detected in 74% of the total isolates. The activity of the protease enzyme was 68%. The lipase enzyme was active in 79% of the tested S. aureus isolates. The majority of isolates were established to be agrI 84%, followed by agrII 53%, agrIII 32%, and 30% of the isolates have agr IV. Our study indicated that the majority of MRSA isolates were non-biofilm producers and the agr I is the most dominant type. Thus, agr I is not correlated with biofilm production.


Subject(s)
Bacterial Proteins , Biofilms , Methicillin-Resistant Staphylococcus aureus , Xanthophylls , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Biofilms/drug effects , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Lipase/genetics , Lipase/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/physiology , Microbial Sensitivity Tests , Polymorphism, Genetic , Staphylococcal Infections/microbiology , Trans-Activators/genetics , Trans-Activators/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL