Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.118
Filter
1.
Biomaterials ; 313: 122774, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39208699

ABSTRACT

Osteomyelitis (OM) is a progressive, inflammatory infection of bone caused predominately by Staphylococcus aureus. Herein, we engineered an antibiotic-eluting collagen-hydroxyapatite scaffold capable of eliminating infection and facilitating bone healing. An iterative freeze-drying and chemical crosslinking approach was leveraged to modify antibiotic release kinetics, resulting in a layered dual-release system whereby an initial rapid release of antibiotic to clear infection was followed by a sustained controlled release to prevent reoccurrence of infection. We observed that the presence of microbial collagenase accelerated antibiotic release from the crosslinked layer of the scaffold, indicating that the material is responsive to microbial activity. As exemplar drugs, vancomycin and gentamicin-eluting scaffolds were demonstrated to be bactericidal, and supported osteogenesis in vitro. In a pilot murine model of OM, vancomycin-eluting scaffolds were observed to reduce S. aureus infection within the tibia. Finally, in a rabbit model of chronic OM, gentamicin-eluting scaffolds both facilitated radial bone defect healing and eliminated S. aureus infection. These results show that antibiotic-eluting collagen-hydroxyapatite scaffolds are a one-stage therapy for OM, which when implanted into infected bone defects simultaneously eradicate infection and facilitate bone tissue healing.


Subject(s)
Anti-Bacterial Agents , Gentamicins , Osteomyelitis , Staphylococcal Infections , Staphylococcus aureus , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcal Infections/drug therapy , Osteomyelitis/drug therapy , Rabbits , Staphylococcus aureus/drug effects , Gentamicins/pharmacology , Gentamicins/administration & dosage , Gentamicins/chemistry , Gentamicins/therapeutic use , Mice , Vancomycin/pharmacology , Vancomycin/chemistry , Vancomycin/administration & dosage , Durapatite/chemistry , Kinetics , Wound Healing/drug effects , Osteogenesis/drug effects , Collagen/chemistry , Female
2.
BMC Nephrol ; 25(1): 322, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334001

ABSTRACT

INTRODUCTION: Different initial manifestations of peritoneal dialysis-associated peritonitis (PDAP) may depend on the type of pathogenic organism. We investigated the association between the clinical characteristics of PDAP and susceptibility to vancomycin and investigated the possibility of using vancomycin monotherapy alone as an initial treatment regimen for some PDAP patients to avoid unnecessary antibiotic exposure and secondary infection. METHODS: Patients with culture-positive PDAP were retrospectively analyzed and divided into two groups: peritonitis with only cloudy effluent (PDAP-cloudy) or with cloudy effluent, abdominal pain and/or fever (PDAP-multi). The bacterial culture of PD effluent and antibiotic sensitivity test results were compared between groups. Logistic regression was used to investigate factors predicting susceptibility to vancomycin. RESULTS: Of 162 episodes of peritonitis which had a positive bacterial culture of PD fluid, 30 peritonitis were in the PDAP-cloudy group, and 132 peritonitis were in the PDAP-multi group. Thirty (100%) peritonitis in the PDAP-cloudy group had gram-positive bacterial infections, which was significantly greater than that in the PDAP-multi group (51.5%) (P < 0.001). Twenty-nine (96.7%) peritonitis in the PDAP-cloudy group were susceptible to vancomycin, compared to 67 (50.8%) in the PDAP-multi group (P < 0.001). The specificity of PDAP-cloudy for vancomycin-sensitive peritonitis was 98.48%. Only one patient (3.3%) in the PDAP-cloudy group experienced vancomycin-resistant peritonitis caused by Enterococcus gallinarum, which could neither be covered by vancomycin nor by the initial antibiotic regimen recommended by the current ISPD guidelines. The presence of only cloudy effluent was an independent predictor of susceptibility to vancomycin according to multivariate analysis (OR = 27.678, 95% CI 3.191-240.103, p = 0.003), in addition to PD effluent WBC counts (OR = 0.988, 95% CI 0.980-0.996, p = 0.004), diabetes mellitus (OR = 3.646, 95% CI 1.580-8.416, p = 0.002), first episode peritonitis (OR = 0.447, 95% CI 0.207-0.962, p = 0.039) and residual renal creatinine clearance (OR = 0.956, 95% CI 0.918-0.995, p = 0.027). Addition of these characteristics increased the AUC to 0.813 (95% CI 0.0.749-0.878, P < 0.001). The specificity of presenting with only cloudy effluent for vancomycin-sensitive peritonitis was 98.48%. CONCLUSIONS: Cloudy dialysate, as the only symptom at PDAP onset, was an independent predictor of vancomycin-sensitive PDAP, which is an important new insight that may guide the choice of initial antibiotic treatment.


Subject(s)
Anti-Bacterial Agents , Peritoneal Dialysis , Peritonitis , Vancomycin , Humans , Peritonitis/microbiology , Peritonitis/drug therapy , Peritonitis/etiology , Male , Female , Middle Aged , Peritoneal Dialysis/adverse effects , Retrospective Studies , Vancomycin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Aged , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Microbial Sensitivity Tests , Adult
3.
J Orthop Surg Res ; 19(1): 602, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342369

ABSTRACT

In this study, we developed scaffolds materials with microspheres to form a double sustained release system.Chitosan/nano-hydroxyapatite (CS-HA) was used as a drug carrier to construct a sustained-release system for Bone morphogenetic protein-2(BMP-2) and Vancomycin (VAN). Furthermore, VAN and BMP-2 loaded microspheres (Ms) were prepared by the emulsion ultrasonic method.The resultant composites were characterized by Scanning electron microscope (SEM), compressive strength, porosity, and biodegradation. The characterization results showed uniform porous and rough surface, enhanced thermal stability, and highest compressive strength ((1.912 ± 0.012) Kpa, the surface of the two microspheres was slightly folded and showed a regular spherical shape.The loading rate of BMP-2 was (59.611 × 10-4 ± 0.023 × 10-4)% and the encapsulation rate was (6.022 ± 0.005)%. The release rate of vancomycin and BMP-2 was 57.194% and 12.968% respectively. Osteogenic differentiation of Bone marrow mesenchymal stem cells (BMSCs) was confirmed by alkaline phosphatase quantification. The deposition of late osteogenic markers (calcium phosphates) detected by Alizarin red, which indicated extracellular matrix mineralization. The results showed that BMP-2/VAN in CS-HA hydrogel successfully achieved the sequential release of the double drugs, which could benefit bone regeneration.


Subject(s)
Bone Morphogenetic Protein 2 , Chitosan , Durapatite , Hydrogels , Osteomyelitis , Vancomycin , Vancomycin/administration & dosage , Vancomycin/pharmacokinetics , Bone Morphogenetic Protein 2/administration & dosage , Chitosan/administration & dosage , Chitosan/chemistry , Durapatite/administration & dosage , Osteomyelitis/drug therapy , Animals , Anti-Bacterial Agents/administration & dosage , Chronic Disease , Delayed-Action Preparations , Drug Carriers , Microspheres , Drug Liberation , Osteogenesis/drug effects , Mesenchymal Stem Cells
4.
NPJ Biofilms Microbiomes ; 10(1): 86, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39284817

ABSTRACT

Auranofin (AF), a former rheumatoid polyarthritis treatment, gained renewed interest for its use as an antimicrobial. AF is an inhibitor of thioredoxin reductase (TrxB), a thiol and protein repair enzyme, with an antibacterial activity against several bacteria including C. difficile, an enteropathogen causing post-antibiotic diarrhea. Several studies demonstrated the effect of AF on C. difficile physiology, but the crucial questions of resistance mechanisms and impact on microbiota remain unaddressed. We explored potential resistance mechanisms by studying the impact of TrxB multiplicity and by generating and characterizing adaptive mutations. We showed that if mutants inactivated for trxB genes have a lower MIC of AF, the number of TrxBs naturally present in clinical strains does not impact the MIC. All stable mutations isolated after AF long-term exposure were in the anti-sigma factor of σB and strongly affect physiology. Finally, we showed that AF has less impact on human gut microbiota than vancomycin.


Subject(s)
Anti-Bacterial Agents , Auranofin , Clostridioides difficile , Gastrointestinal Microbiome , Microbial Sensitivity Tests , Humans , Auranofin/pharmacology , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Gastrointestinal Microbiome/drug effects , Anti-Bacterial Agents/pharmacology , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxin-Disulfide Reductase/genetics , Mutation , Drug Resistance, Bacterial , Adaptation, Physiological , Sigma Factor/genetics , Sigma Factor/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Vancomycin/pharmacology
5.
Biofouling ; 40(9): 593-601, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39219014

ABSTRACT

Candida albicans invasive candidiasis is considered a global health problem. In such cases, biofilm formation on implanted devices represents a therapeutic challenge and the presence of metabolically inactive persistent cells (PCs) in these communities increases their tolerance to fungicidal drugs. This study investigated the influence of amoxicillin, AMX; cefepime, CEF; gentamicin, GEN; amikacin, AMK; vancomycin, VAN; and ciprofloxacin, CIP; on the production of PCs in biofilms of C. albicans bloodstream isolates. 48 h-mature biofilms (n = 6) grown in RPMI-1640 supplemented with antibiotics were treated with 100 µg ml-1 amphotericin B and then evaluated for PCs. Biofilms grown in the presence of antibiotics produced more PCs, up to 10×, when exposed to AMX and CIP; 5 × to CEF; and 6 × to GEN and VAN. The results indicate that antibiotics can modulate PC production in C. albicans biofilms. This scenario may have clinical repercussions in immunocompromised patients under broad-spectrum antibiotic therapy.


Biofilms are microbial communities tolerant to antifungals. Our research showed that antibiotics stimulate the formation of persistent cells within Candida albicans biofilms. These are dormant, metabolically silent cells that resist to therapy and can be related to metastatic and recalcitrant infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Candida albicans , Biofilms/drug effects , Biofilms/growth & development , Candida albicans/drug effects , Candida albicans/physiology , Anti-Bacterial Agents/pharmacology , Humans , Antifungal Agents/pharmacology , Microbial Sensitivity Tests , Ciprofloxacin/pharmacology , Gentamicins/pharmacology , Amoxicillin/pharmacology , Vancomycin/pharmacology , Amikacin/pharmacology , Cefepime/pharmacology , Amphotericin B/pharmacology , Cephalosporins/pharmacology , Candidiasis/microbiology , Candidiasis/drug therapy
6.
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
7.
Nat Commun ; 15(1): 8263, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327429

ABSTRACT

Understanding human, animal, and environmental microbiota is essential for advancing global health and combating antimicrobial resistance (AMR). We investigate the oral and gut microbiota of 48 animal species in captivity, comparing them to those of wildlife animals. Specifically, we characterize the microbiota composition, metabolic pathways, AMR genes, and biosynthetic gene clusters (BGCs) encoding the production of specialized metabolites. Our results reveal a high diversity of microbiota, with 585 novel species-level genome bins (SGBs) and 484 complete BGCs identified. Functional gene analysis of microbiomes shows diet-dependent variations. Furthermore, by comparing our findings to wildlife-derived microbiomes, we observe the impact of captivity on the animal microbiome, including examples of converging microbiome compositions. Importantly, our study identifies AMR genes against commonly used veterinary antibiotics, as well as resistance to vancomycin, a critical antibiotic in human medicine. These findings underscore the importance of the 'One Health' approach and the potential for zoonotic transmission of pathogenic bacteria and AMR. Overall, our study contributes to a better understanding of the complexity of the animal microbiome and highlights its BGC diversity relevant to the discovery of novel antimicrobial compounds.


Subject(s)
Animals, Wild , Animals, Zoo , Anti-Bacterial Agents , Gastrointestinal Microbiome , Microbiota , Animals , Animals, Wild/microbiology , Animals, Zoo/microbiology , Gastrointestinal Microbiome/genetics , Anti-Bacterial Agents/pharmacology , Microbiota/genetics , Microbiota/drug effects , Bacteria/genetics , Bacteria/metabolism , Bacteria/classification , Bacteria/drug effects , Multigene Family , Humans , Biodiversity , Drug Resistance, Bacterial/genetics , Vancomycin/pharmacology , Biosynthetic Pathways/genetics
8.
BMC Anesthesiol ; 24(1): 344, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342092

ABSTRACT

BACKGROUND: Lower gastrointestinal perforation (LGP) is a surgical emergency disease that can result in secondary bacterial peritonitis. Microbiological studies on LGP are rare. The present study aimed to ascertain the microbiological profile of LGP in patients admitted to the intensive care unit (ICU) at our institute after surgery. In addition, we investigated whether initial empirical therapy with vancomycin was associated with in-hospital mortality, duration of ICU stay, and duration of ventilator support. METHODS: This single-center, retrospective, observational study was conducted at Kobe City Medical Center General Hospital, Japan. The study population included all patients diagnosed with LGP who were admitted to the ICU after emergency surgery between 2017 and 2023. The primary outcome assessed was the microbiological profile of microorganisms isolated from ascites fluid and blood of the participants. The secondary end-points were in-hospital mortality, duration of ICU stay, and duration of ventilator support. We performed univariate and multivariate regression analyses to evaluate the end-points. RESULTS: During the study period, 89 patients were included in the analysis. The most commonly identified pathogen from the ascites cultures was Escherichia coli (65.2%), followed by Enterococcus spp. (51.7%). E. faecium was identified in 16 (18.0%) ascites samples. The microbiological profile of critically ill patients with LGP admitted to the ICU after surgery was similar to that of previous studies on intra-abdominal infection (IAI). Therefore, the initial empirical therapy in the IAI guidelines is more appropriate for LGP. Multivariate regression analysis suggested that the combination of initial empirical therapy with vancomycin was not associated with in-hospital mortality (odds ratio [OR] = 0.96, 95% confidence interval [CI] 0.23-3.00, p = 0.955), duration of ICU stay (coefficient=-0.92, 95% CI -3.04-1.21, p = 0.393), or duration of ventilator-support (coefficient=-9.03, 95%CI -49.69-31.63, p = 0.659). CONCLUSION: The microbiological profile of critically ill patients with LGP admitted to the ICU after surgery was similar to that of previous studies on IAI. However, the frequency of E. faecium in the present study was higher than that in previous studies. Initial empirical therapy with drugs such as meropenem in combination with vancomycin for E. faecium was not associated with in-hospital mortality, duration of ICU stay, and duration of ventilator support, after adjusting for confounding factors.


Subject(s)
Anti-Bacterial Agents , Critical Illness , Hospital Mortality , Intensive Care Units , Intestinal Perforation , Length of Stay , Humans , Retrospective Studies , Male , Female , Aged , Japan/epidemiology , Intestinal Perforation/surgery , Intestinal Perforation/microbiology , Middle Aged , Length of Stay/statistics & numerical data , Anti-Bacterial Agents/therapeutic use , Vancomycin/therapeutic use , Respiration, Artificial , Aged, 80 and over
9.
Medicine (Baltimore) ; 103(38): e39410, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39312358

ABSTRACT

To observe the clinical efficacy and safety of vancomycin intravenous drip combined with vancomycin intrathecal injection in the treatment of intracranial infection after severe brain injury surgery. From January 2020 to June 2022, 80 patients with intracranial infection after severe brain injury surgery were selected and randomly divided into 2 subgroups; there were 40 patients in each subgroup. All patients were treated with vancomycin. The control subgroup was medicated with intravenous drip, and the observation subgroup was treated through 2 channels (intravenous drip + intrathecal injection), with a course of 7 days. The clinical efficacy, intracranial pressure, infection control time, routine indexes of cerebrospinal fluid (white blood cell count [WBC], glucose content [Glu], and total protein content [Pro]) and the incidence of adverse reactions were contrasted between the 2 subgroups. Versus the control subgroup, the total effective rate in the observation subgroup was notably higher (95.00% vs 77.50%). After treatment, aiming at the intracranial pressure and infection control time, versus the control subgroup (146.20 ±â€…22.37) mmH2O and (9.86 ±â€…1.62) days, the observation subgroup were (125.43 ±â€…18.5) mmH2O and (7.35 ±â€…1.57) days respectively, which were notably lower. After treatment, versus the control subgroup, the concentrations of WBC and Pro in cerebrospinal fluid in the observation subgroup were lower, and the content of Glu was higher. There was no statistical distinction in the incidence of adverse reactions between the 2 subgroups (17.50% vs 10.00%). Two-channel administration of vancomycin can improve the clinical efficacy of internal infection after severe craniocerebral injury, reduce intracranial pressure, and cerebrospinal fluid WBC and Pro levels, and has high safety.


Subject(s)
Anti-Bacterial Agents , Brain Injuries , Vancomycin , Humans , Vancomycin/administration & dosage , Vancomycin/therapeutic use , Male , Female , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Middle Aged , Adult , Brain Injuries/cerebrospinal fluid , Injections, Spinal , Treatment Outcome , Intracranial Pressure/drug effects
10.
BMC Microbiol ; 24(1): 353, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294587

ABSTRACT

BACKGROUND: Clostridium innocuum, previously considered a commensal microbe, is a spore-forming anaerobic bacterium. C. innocuum displays inherent resistance to vancomycin and is associated with extra-intestinal infections, antibiotic-associated diarrhea, and inflammatory bowel disease. This study seeks to establish a multilocus sequence typing (MLST) scheme to explore the correlation between C. innocuum genotyping and its potential pathogenic phenotypes. METHODS: Fifty-two C. innocuum isolates from Linkou Chang Gung Memorial Hospital (CGMH) in Taiwan and 60 sequence-available C. innocuum isolates from the National Center for Biotechnolgy Information Genome Database were included. The concentrated sequence of housekeeping genes in C. innocuum was determined by amplicon sequencing and used for MLST and phylogenetic analyses. The biofilm production activity of the C. innocuum isolates was determined by crystal violet staining. RESULTS: Of the 112 C. innocuum isolates, 58 sequence types were identified. Maximum likelihood estimation categorized 52 CGMH isolates into two phylogenetic clades. These isolates were found to be biofilm producers, with isolates in clade I exhibiting significantly higher biofilm production than isolates in clade II. The sub-inhibitory concentration of vancomycin seemed to minimally influence biofilm production by C. innocuum isolates. Nevertheless, C. innocuum embedded in the biofilm structure demonstrated resistance to vancomycin treatments at a concentration greater than 256 µg/mL. CONCLUSIONS: This study suggests that a specific genetic clade of C. innocuum produces a substantial amount of biofilm. Furthermore, this phenotype assists C. innocuum in resisting high concentrations of vancomycin, which may potentially play undefined roles in C. innocuum pathogenesis.


Subject(s)
Anti-Bacterial Agents , Biofilms , Clostridium Infections , Clostridium , Genetic Variation , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phylogeny , Vancomycin Resistance , Vancomycin , Biofilms/growth & development , Biofilms/drug effects , Humans , Clostridium/genetics , Clostridium/drug effects , Clostridium/isolation & purification , Clostridium/classification , Anti-Bacterial Agents/pharmacology , Vancomycin/pharmacology , Vancomycin Resistance/genetics , Clostridium Infections/microbiology , Taiwan , Genotype , Genes, Essential
11.
BMC Pediatr ; 24(1): 575, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261805

ABSTRACT

BACKGROUND: Higher doses of vancomycin are currently prescribed due to the emergence of bacterial tolerance and resistance. This study aimed to evaluate the efficacy and safety of the currently adopted vancomycin dosing guide in pediatric cardiology. METHODS: This was a single-center prospective cohort study with pediatric cardiac patients, younger than 14 years, from June 2020 to March 2021. The patients received intravenous vancomycin (40 mg/kg/day divided every 6-8 h) according to the department's vancomycin medication administration guide (MAG) for at least three days. RESULTS: In total, 88 cardiac patients were included, with a median age of 0.82 years (IQR: 0.25-2.9), and 51 (58%) received cardiopulmonary bypass surgery (CPB). The majority (71.6%, n = 61) achieved a serum vancomycin level within the therapeutic range (7-20 mg/L). Infants, young children, and children exposed to CPB surgery had an increased incidence of subtherapeutic vancomycin levels, [7 (29.2%); P = 0.033], [13 (54.2%); P = 0.01], and [21 (87.5%); P = 0.009] respectively. After the treatment, 8 (10%) patients had an elevated Serum creatinine (SCr) and 2 (2.5%) developed acute kidney injury (AKI). However, no significant difference was found between the patients developing AKI or an elevated SCr and the group who did not, in terms of clinical, therapeutic, and demographic characteristics, except for the decreased incidence of SCr elevation in patients receiving an ACE inhibitor, [4 (36.4%); P = 0.036]. CONCLUSION: Our institution followed MAG recommendations; however, subtherapeutic serum concentrations were evident in infants, young children, and CPB patients. Strategies to prevent AKI should be investigated, as the possible causes have not been identified in this study.


Subject(s)
Anti-Bacterial Agents , Vancomycin , Humans , Vancomycin/administration & dosage , Vancomycin/blood , Infant , Child, Preschool , Prospective Studies , Anti-Bacterial Agents/administration & dosage , Female , Male , Child , Adolescent , Infant, Newborn , Practice Guidelines as Topic , Acute Kidney Injury
12.
World J Microbiol Biotechnol ; 40(10): 322, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283509

ABSTRACT

Staphylococcus aureus can develop antibiotic resistance and evade immune responses, causing infections in different body sites. However, the metabolic changes underlying this process are poorly understood. A variant strain, C1V, was derived from the parental strain C1 by exposing it to increasing concentrations of vancomycin in vitro. C1V exhibited a vancomycin-intermediate phenotype and physiological changes compared to C1. It showed higher survival rates than C1 when phagocytosed by Raw264.7 cells. Metabolomics analysis identified significant metabolic differences pre- and post-induction (C1 + SC1 vs. C1V + SC1V: 201 metabolites) as well as pre- and post-phagocytosis (C1 vs. SC1: 50 metabolites; C1V vs. SC1V: 95 metabolites). The variant strain had distinct morphological characteristics, decreased adhesion ability, impaired virulence, and enhanced resistance to phagocytosis compared to the parental strain. Differential metabolites may contribute to S. aureus ' resistance to antibiotics and phagocytosis, offering insights into potential strategies for altering vancomycin nonsusceptibility and enhancing phagocyte killing by manipulating bacterial metabolism.


Subject(s)
Anti-Bacterial Agents , Metabolomics , Phagocytosis , Staphylococcus aureus , Vancomycin , Vancomycin/pharmacology , Mice , Animals , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Phagocytosis/drug effects , RAW 264.7 Cells , Anti-Bacterial Agents/pharmacology , Virulence , Staphylococcal Infections/microbiology , Microbial Sensitivity Tests , Vancomycin Resistance/genetics , Metabolome/drug effects , Bacterial Adhesion/drug effects , Adaptation, Physiological
13.
ACS Appl Mater Interfaces ; 16(36): 48058-48072, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39221786

ABSTRACT

Chiral amino acids (AAs) are essential in metabolism and understanding physiological processes, and they could be used as biomarkers for the diagnosis of different diseases. In this study, chiral Cdots@Van were prepared by postmodifying an achiral Cdots core with vancomycin for recognizing and determining the enantiomeric excess (ee) of tyrosine (Tyr) enantiomers. The fluorescence response of Cdots@Van is based on an "on-off" strategy, with different quenching percentages for d- and l-tyrosine. Interestingly, the circular dichroism (CD) spectrum of Cdots@Van responded to only one form of Tyr enantiomer, specifically d-Tyr, and remained nearly unchanged upon the addition of l-Tyr. Quantum mechanical (QM) calculations were in excellent agreement with the experimental results, confirming the stronger binding affinity of Cdots@Van for d-Tyr compared to l-Tyr. We further investigated the chiral recognition ability of the interconnected vancomycin particles, which was synthesized using the EDC/NHS coupling reaction between vancomycin molecules without a Cdots core. Surprisingly, unlike free vancomycin molecules, interconnected vancomycin displayed an enantiomeric recognition ability by CD spectroscopy, similar to what was observed for Cdots@Van. Crucially, this chiral probe has been successfully utilized for cell imaging applications.


Subject(s)
Circular Dichroism , Tyrosine , Vancomycin , Tyrosine/chemistry , Vancomycin/chemistry , Humans , Stereoisomerism , Fluorescent Dyes/chemistry , Quantum Dots/chemistry , Spectrometry, Fluorescence , Optical Imaging
14.
Sci Rep ; 14(1): 21006, 2024 09 09.
Article in English | MEDLINE | ID: mdl-39251613

ABSTRACT

The emission of glyphosate and antibiotic residues from human activities threatens the diversity and functioning of the microbial community. This study examines the impact of a glyphosate-based herbicide (GBH) and common antibiotics on Gram-negative bacteria within the ESKAPEE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp. and Escherichia coli). Ten strains, including type and multidrug-resistant strains for each species were analysed and eight antibiotics (cefotaxime, meropenem, aztreonam, ciprofloxacin, gentamicin, tigecycline, sulfamethoxazole-trimethoprim, and colistin) were combined with the GBH. While most combinations yielded additive or indifferent effects in 70 associations, antagonistic effects were observed with ciprofloxacin and gentamicin in five strains. GBH notably decreased the minimum inhibitory concentration of colistin in eight strains and displayed synergistic activity with meropenem against metallo-ß-lactamase (MBL)-producing strains. Investigation into the effect of GBH properties on outer membrane permeability involved exposing strains to a combination of this GBH and vancomycin. Results indicated that GBH rendered strains sensitive to vancomycin, which is typically ineffective against Gram-negative bacteria. Furthermore, we examined the impact of GBH in combination with three carbapenem agents on 14 strains exhibiting varying carbapenem-resistance mechanisms to assess its effect on carbapenemase activity. The GBH efficiently inhibited MBL activity, demonstrating similar effects to EDTA (ethylenediaminetetraacetic acid). Chelating effect of GBH may have multifaceted impacts on bacterial cells, potentially by increasing outer membrane permeability and inactivating metalloenzyme activity.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Glycine , Glyphosate , Gram-Negative Bacteria , Herbicides , Microbial Sensitivity Tests , Glycine/analogs & derivatives , Glycine/pharmacology , Anti-Bacterial Agents/pharmacology , Herbicides/pharmacology , Gram-Negative Bacteria/drug effects , Acinetobacter baumannii/drug effects , Klebsiella pneumoniae/drug effects , Humans , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Ciprofloxacin/pharmacology , Enterococcus faecium/drug effects , Staphylococcus aureus/drug effects , Colistin/pharmacology , Vancomycin/pharmacology , Enterobacter/drug effects , Drug Synergism , Meropenem/pharmacology , Phenotype , Gentamicins/pharmacology
15.
Pol J Microbiol ; 73(3): 403-410, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39268956

ABSTRACT

Vancomycin-resistant Enterococcus faecium (VRE) has been detected in Türkiye. Only limited information is available on its dissemination in the central regions of the country. This study describes the first epidemiological characterization of VRE clinical isolates detected in patients in a hospital in the province of Aksaray. In this one-year study conducted between 2021 and 2022, stool samples from intensive care unit patients were screened for VRE using the phenotypic E-test method, and the antibiotic sensitivity test was analyzed by using the VITEK® 2 system. A molecular assay for confirmation of species level was carried out by 16S rRNA gene-based sequencing and testing for antibiotic resistance (vanA or vanB) and virulence factor-encoding genes (esp, asa1, and hyl). Further, genotypic characterization was determined by macro-restriction fragment pattern analysis (MRFPA) of genomic DNA digested with SmaI restriction enzyme. Of the total 350 Enterococcus positive patients from different hospital intensive care units, 22 (6.3%) were positive for VRE using the phenotypic E-test method. All isolates showed resistance to ampicillin, ciprofloxacin, vancomycin, and teicoplanin and positive amplification for the vanA gene. However, none of the isolates was positive for the vanB gene. The most prevalent virulence gene was esp. The results indicate that the isolates are persistent in the hospital environment and subsequently transmitted to hospitalized patients, thus representing challenges to an outbreak and infection control. These study results would also help formulate more effective strategies to reduce the transmission and propagation of VRE contamination in various hospital settings.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterococcus faecium , Genotype , Gram-Positive Bacterial Infections , Intensive Care Units , Microbial Sensitivity Tests , Vancomycin-Resistant Enterococci , Humans , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Vancomycin-Resistant Enterococci/drug effects , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Virulence Factors/genetics , Vancomycin/pharmacology , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Phenotype , Male , Female , Vancomycin Resistance/genetics , Middle Aged
16.
Proc Natl Acad Sci U S A ; 121(39): e2403510121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39288179

ABSTRACT

Multispecies microbial communities drive most ecosystems on Earth. Chemical and biological interactions within these communities can affect the survival of individual members and the entire community. However, the prohibitively high number of possible interactions within a microbial community has made the characterization of factors that influence community development challenging. Here, we report a Microbial Community Interaction (µCI) device to advance the systematic study of chemical and biological interactions within a microbial community. The µCI creates a combinatorial landscape made up of an array of triangular wells interconnected with circular wells, which each contains either a different chemical or microbial strain, generating chemical gradients and revealing biological interactions. Bacillus cereus UW85 containing green fluorescent protein provided the "target" readout in the triangular wells, and antibiotics or microorganisms in adjacent circular wells are designated the "variables." The µCI device revealed that gentamicin and vancomycin are antagonistic to each other in inhibiting the target B. cereus UW85, displaying weaker inhibitory activity when used in combination than alone. We identified three-member communities constructed with isolates from the plant rhizosphere that increased or decreased the growth of B. cereus. The µCI device enables both strain-level and community-level insight. The scalable geometric design of the µCI device enables experiments with high combinatorial efficiency, thereby providing a simple, scalable platform for systematic interrogation of three-factor interactions that influence microorganisms in solitary or community life.


Subject(s)
Bacillus cereus , Microbial Interactions/physiology , Microbiota/physiology , Anti-Bacterial Agents/pharmacology , Vancomycin/pharmacology , Rhizosphere , Gentamicins/pharmacology , Lab-On-A-Chip Devices , Green Fluorescent Proteins/metabolism
17.
Sci Rep ; 14(1): 21749, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39294268

ABSTRACT

Implant-related infections pose significant challenges to orthopedic surgeries due to the high risk of severe complications. The widespread use of bioactive prostheses in joint replacements, featuring roughened surfaces and tight integration with the bone marrow cavity, has facilitated bacterial proliferation and complicated treatment. Developing antibacterial coatings for orthopedic implants has been a key research focus in recent years to address this critical issue. Researchers have designed coatings using various materials and antibacterial strategies. In this study, we fabricated 3D-printed porous titanium rods, incorporated vancomycin-loaded mPEG750-b-PCL2500 gel, and coated them with a PCL layer. We then evaluated the antibacterial efficacy through both in vitro and in vivo experiments. Our coating passively inhibits bacterial biofilm formation and actively controls antibiotic release in response to bacterial growth, providing a practical solution for proactive and sustained infection control. This study utilized 3D printing technology to produce porous titanium rod implants simulating bioactive joint prostheses. The porous structure of the titanium rods was used to load a thermoresponsive gel, mPEG750-b-PCL2500 (PEG: polyethylene glycol; PCL: polycaprolactone), serving as a novel drug delivery system carrying vancomycin for controlled antibiotic release. The assembly was then covered with a PCL membrane that inhibits bacterial biofilm formation early in infection and degrades when exposed to lipase solutions, mimicking enzymatic activity during bacterial infections. This setup provides infection-responsive protection and promotes drug release. We investigated the coating's controlled release, antibacterial capability, and biocompatibility through in vitro experiments. We established a Staphylococcus aureus infection model in rabbits, implanting titanium rods in the femoral medullary cavity. We evaluated the efficacy and safety of the composite coating in preventing implant-related infections using imaging, hematology, and pathology. In vitro experiments demonstrated that the PCL membrane stably protects encapsulated vancomycin during PBS immersion. The PCL membrane rapidly degraded at a lipase concentration of 0.2 mg/mL. The mPEG750-b-PCL2500 gel ensured stable and sustained vancomycin release, inhibiting bacterial growth. We investigated the antibacterial effect of the 3D-printed titanium material, coated with PCL and loaded with mPEG750-b-PCL2500 hydrogel, using a rabbit Staphylococcus aureus infection model. Imaging, hematology, and histopathology confirmed that our composite antibacterial coating exhibited excellent antibacterial effects and infection prevention, with good safety in trials. Our results indicate that the composite antibacterial coating effectively protects vancomycin in the hydrogel from premature release in the absence of bacterial infection. The outer PCL membrane inhibits bacterial growth and prevents biofilm formation. Upon contact with bacterial lipase, the PCL membrane rapidly degrades, releasing vancomycin for antibacterial action. The mPEG750-b-PCL2500 gel provides stable and sustained vancomycin release, prolonging its antibacterial effects. Our composite antibacterial coating demonstrates promising potential for clinical application.


Subject(s)
Anti-Bacterial Agents , Hydrogels , Polyesters , Printing, Three-Dimensional , Titanium , Vancomycin , Titanium/chemistry , Vancomycin/pharmacology , Vancomycin/administration & dosage , Vancomycin/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Polyesters/chemistry , Animals , Hydrogels/chemistry , Rabbits , Staphylococcus aureus/drug effects , Drug Liberation , Porosity , Biofilms/drug effects , Polyethylene Glycols/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control , Drug Delivery Systems/methods , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology
19.
ACS Nano ; 18(35): 24327-24349, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39169538

ABSTRACT

A series of progress has been made in the field of antimicrobial use of nanozymes due to their superior stability and decreased susceptibility to drug resistance. However, catalytically generated reactive oxygen species (ROS) are insufficient for coping with multidrug-resistant organisms (MDROs) in complex wound environments due to their low targeting ability and insufficient catalytic activity. To address this problem, chemically stable copper-gallic acid-vancomycin (CuGA-VAN) nanoneedles were successfully constructed by a simple approach for targeting bacteria; these nanoneedles exhibit OXD-like and GSH-px-like dual enzyme activities to produce ROS and induce bacterial cuproptosis-like death, thereby eliminating MDRO infections. The results of in vitro experiments showed that the free carboxylic acid of GA could react with the free ammonia of teichoic acid in the methicillin-resistant Staphylococcus aureus (MRSA) cell wall skeleton. Thus, CuGA-VAN nanoneedles can rapidly "capture" MRSA in liquid environments, releasing ROS, VAN and Cu2+ on bacterial surfaces to break down the MRSA barrier, destroying the biofilm. In addition, CuGA-VAN effectively promoted wound repair cell proliferation and angiogenesis to facilitate wound healing while ensuring biosafety. According to transcriptome sequencing, highly internalized Cu2+ causes copper overload toxicity; downregulates genes related to the bacterial glyoxylate cycle, tricarboxylic acid cycle, and oxidative respiratory chain; and induces lipid peroxidation in the cytoplasm, leading to bacterial cuproptosis-like death. In this study, CuGA-VAN was cleverly designed to trigger a cascade reaction of targeting, drug release, ROS-catalyzed antibacterial activity and cuproptosis-like death. This provides an innovative idea for multidrug-resistant infections.


Subject(s)
Anti-Bacterial Agents , Copper , Methicillin-Resistant Staphylococcus aureus , Reactive Oxygen Species , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Copper/chemistry , Copper/pharmacology , Reactive Oxygen Species/metabolism , Vancomycin/pharmacology , Vancomycin/chemistry , Microbial Sensitivity Tests , Animals , Humans , Biofilms/drug effects , Nanostructures/chemistry , Mice
20.
J Orthop Surg Res ; 19(1): 517, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39198853

ABSTRACT

BACKGROUND: Open fractures are challenging due to susceptibility to Staphylococcus aureus infections. This study examines the impact of Vancomycin-Loaded Calcium Sulfate (VLCS) and negative pressure wound therapy (NPWT) on macrophage behavior in enhancing healing and infection resistance. Both VLCS and NPWT were evaluated individually and in combination to determine their effects on macrophage polarization and infection resistance in open fractures. METHODS: Through single-cell RNA sequencing, genomic expressions in macrophages from open fracture patients treated with VLCS and NPWT were compared to a control group. The analysis focused on MBD2 gene changes related to macrophage polarization. RESULTS: Remarkable modifications in MBD2 expression in the treatment group indicate a shift towards M2 macrophage polarization. Additionally, the combined treatment group exhibited greater improvements in infection resistance and healing compared to the individual treatments. This shift suggests a healing-promoting atmosphere with improved infection resilience. CONCLUSIONS: VLCS and NPWT demonstrate the ability to alter macrophage behavior toward M2 polarization, which is crucial for infection prevention in open fractures. The synergistic effect of their combined use shows even greater promise in enhancing outcomes in orthopedic trauma care.


Subject(s)
Calcium Sulfate , Fractures, Open , Macrophages , Negative-Pressure Wound Therapy , Vancomycin , Calcium Sulfate/administration & dosage , Calcium Sulfate/therapeutic use , Negative-Pressure Wound Therapy/methods , Humans , Vancomycin/administration & dosage , Vancomycin/therapeutic use , Fractures, Open/therapy , Male , Female , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Middle Aged , Staphylococcal Infections/prevention & control , Adult , Wound Healing/drug effects , Surgical Wound Infection/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL