Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.374
Filter
1.
Food Res Int ; 188: 114491, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823842

ABSTRACT

Minimum inhibitory concentrations (MIC) assays are often questioned for their representativeness. Especially when foodborne pathogens are tested, it is of crucial importance to also consider parameters of the human digestive system. Hence, the current study aimed to assess the inhibitory capacity of two antibiotics, ciprofloxacin and tetracycline, against Salmonella enterica and Listeria monocytogenes, under representative environmental conditions. More specifically, aspects of the harsh environment of the human gastrointestinal tract (GIT) were gradually added to the experimental conditions starting from simple aerobic lab conditions into an in vitro simulation of the GIT. In this way, the effects of parameters including the anoxic environment, physicochemical conditions of the GIT (low gastric pH, digestive enzymes, bile acids) and the gut microbiota were evaluated. The latter was simulated by including a representative consortium of selected gut bacteria species. In this study, the MIC of the two antibiotics against the relevant foodborne pathogens were established, under the previously mentioned environmental conditions. The results of S. enterica highlighted the importance of the anaerobic environment when conducting such studies, since the pathogen thrived under such conditions. Inclusion of physicochemical barriers led to exactly opposite results for S. enterica and L. monocytogenes since the former became more susceptible to ciprofloxacin while the latter showed lower susceptibility towards tetracycline. Finally, the inclusion of gut bacteria had a bactericidal effect against L. monocytogenes even in the absence of antibiotics, while gut bacteria protected S. enterica from the effect of ciprofloxacin.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Listeria monocytogenes , Microbial Sensitivity Tests , Salmonella enterica , Tetracycline , Ciprofloxacin/pharmacology , Listeria monocytogenes/drug effects , Salmonella enterica/drug effects , Tetracycline/pharmacology , Anti-Bacterial Agents/pharmacology , Humans , Gastrointestinal Tract/microbiology , Gastrointestinal Microbiome/drug effects , Food Microbiology , Hydrogen-Ion Concentration , Foodborne Diseases/microbiology , Foodborne Diseases/prevention & control
2.
Water Sci Technol ; 89(10): 2839-2850, 2024 May.
Article in English | MEDLINE | ID: mdl-38822618

ABSTRACT

Antibiotics release into the water environment through sewage discharge is a significant environmental concern. In the present study, we investigated the removal of ciprofloxacin (CIP) in simulated sewage by biological aeration filter (BAF) equipped with Fe3O4-modified zeolite (Fe3O4@ZF). Fe3O4@ZF were prepared with impregnation method, and the Fe3O4 particles were successfully deposited on the surface of ZF in an amorphous form according to the results of XPS and XRD analysis. The modification also increased the specific surface area (from 16.22 m²/g to 22 m²/g) and pore volume (from 0.0047 cm³/g to 0.0063 cm³/g), improving the adsorption efficiency of antibiotics. Fe3O4 modified ZF improved the treatment performance significantly, and the removal efficiency of CIP in BAF-Fe3O4@ZF was 79%±2.4%. At 10ml/L CIP, the BAF-Fe3O4@ZF reduced the relative abundances of antibiotics resistance genes (ARGs) int, mexA, qnrB and qnrS in the effluent by 57.16%, 39.59%, 60.22%, and 20.25%, respectively, which effectively mitigate the dissemination risk of ARGs. The modification of ZF increased CIP-degrading bacteria abundance, such as Rhizobium and Deinococcus-Thermus, and doubled bacterial ATP activity, promoting CIP degradation. This study offers a viable, efficient method to enhance antibiotic treatment and prevent leakage via sewage discharge.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Wastewater , Water Pollutants, Chemical , Zeolites , Zeolites/chemistry , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Wastewater/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Filtration/methods , Water Purification/methods , Waste Disposal, Fluid/methods , Adsorption , Drug Resistance, Microbial/genetics , Genes, Bacterial , Drug Resistance, Bacterial/genetics
3.
Sci Rep ; 14(1): 10406, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710736

ABSTRACT

Active pharmaceutical ingredients have emerged as an environmentally undesirable element because of their widespread exploitation and consequent pollution, which has deleterious effects on living things. In the pursuit of sustainable environmental remediation, biomedical applications, and energy production, there has been a significant focus on two-dimensional materials (2D materials) owing to their unique electrical, optical, and structural properties. Herein, we have synthesized 2D zinc oxide nanosheets (ZnO NSs) using a facile and practicable hydrothermal method and characterized them thoroughly using spectroscopic and microscopic techniques. The 2D nanosheets are used as an efficient photocatalyst for antibiotic (herein, end-user ciprofloxacin (CIP) was used as a model antibiotic) degradation under sunlight. It is observed that ZnO NSs photodegrade ~ 90% of CIP within two hours of sunlight illumination. The molecular mechanism of CIP degradation is proposed based on ex-situ IR analysis. Moreover, the 2D ZNO NSs are used as an antimicrobial agent and exhibit antibacterial qualities against a range of bacterial species, including Escherichia coli, Staphylococcus aureus, and MIC of the bacteria are found to be 5 µg/l and 10 µg/l, respectively. Despite having the biocompatible nature of ZnO, as-synthesized nanosheets have also shown cytotoxicity against two types of cancer cells, i.e. A549 and A375. Thus, ZnO nanosheets showed a nontoxic nature, which can be exploited as promising alternatives in different biomedical applications.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Nanostructures , Zinc Oxide , Zinc Oxide/chemistry , Zinc Oxide/pharmacology , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Catalysis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nanostructures/chemistry , Escherichia coli/drug effects , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Cell Line, Tumor , Photochemical Processes , Photolysis
4.
Water Sci Technol ; 89(9): 2457-2467, 2024 May.
Article in English | MEDLINE | ID: mdl-38747960

ABSTRACT

To investigate the physicochemical conditions necessary to stably remove antibiotic-resistant bacteria (ARB) via contact with activated sludge (AS), the adhesion of ciprofloxacin (CIP)-resistant and -susceptible Escherichia coli to AS was simulated by contact tests in the laboratory. The CIP-resistant E. coli and susceptible E. coli were removed by a 3 log smaller concentration by a 5 h contact test at maximum. Considering the hydraulic retention time of a reaction tank (∼5 h) and step-feeding operation, we considered the removal rate of E. coli in the current simulated contact test to be in agreement with the actual situation where 1-2 log concentrations of E. coli were reported to be removed from an AS reaction tank. With the increase in the AS concentration and/or dissolved oxygen, the removal rate of E. coli increased. The removal rate of CIP-resistant E. coli was greater than that of susceptible E. coli under all experimental conditions. Although the mechanism by which CIP-resistant E. coli preferably adhered to AS was not clearly understood in detail, finding optimum conditions under which bacteria, including ARB, were efficiently removed by the AS process may be possible.


Subject(s)
Bacterial Adhesion , Ciprofloxacin , Drug Resistance, Bacterial , Escherichia coli , Sewage , Ciprofloxacin/pharmacology , Escherichia coli/drug effects , Sewage/microbiology , Bacterial Adhesion/drug effects , Anti-Bacterial Agents/pharmacology
5.
Aquat Toxicol ; 271: 106925, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718521

ABSTRACT

Excessive antibiotic use has led to the spread of antibiotic resistance genes (ARGs), impacting gut microbiota and host health. However, the effects of antibiotics on amphibian populations remain unclear. We investigated the impact of oxytetracycline (OTC) and ciprofloxacin (CIP) on Chinese giant salamanders (Andrias davidianus), focusing on gut microbiota, ARGs, and gene expression by performing metagenome and transcriptome sequencing. A. davidianus were given OTC (20 or 40 mg/kg) or CIP (50 or 100 mg/kg) orally for 7 days. The results revealed that oral administration of OTC and CIP led to distinct changes in microbial composition and functional potential, with CIP treatment having a greater impact than OTC. Antibiotic treatment also influenced the abundance of ARGs, with an increase in fluoroquinolone and multi-drug resistance genes observed post-treatment. The construction of metagenome-assembled genomes (MAGs) accurately validated that CIP intervention enriched fish-associated potential pathogens Aeromonas hydrophila carrying an increased number of ARGs. Additionally, mobile genetic elements (MGEs), such as phages and plasmids, were implicated in the dissemination of ARGs. Transcriptomic analysis of the gut revealed significant alterations in gene expression, particularly in immune-related pathways, with differential effects observed between OTC and CIP treatments. Integration of metagenomic and transcriptomic data highlighted potential correlations between gut gene expression and microbial composition, suggesting complex interactions between the host gut and its gut microbiota in response to antibiotic exposure. These findings underscore the importance of understanding the impact of antibiotic intervention on the gut microbiome and host health in amphibians, particularly in the context of antibiotic resistance and immune function.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Gastrointestinal Microbiome , Oxytetracycline , Urodela , Animals , Oxytetracycline/toxicity , Gastrointestinal Microbiome/drug effects , Ciprofloxacin/pharmacology , Ciprofloxacin/toxicity , Urodela/genetics , Urodela/microbiology , Anti-Bacterial Agents/toxicity , Anti-Bacterial Agents/pharmacology , Transcriptome/drug effects , Metagenome , Metagenomics , Gene Expression Profiling , Water Pollutants, Chemical/toxicity , Aeromonas hydrophila/drug effects , Gene Expression Regulation/drug effects
6.
Sci Rep ; 14(1): 11706, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778123

ABSTRACT

Co-administering a low dose of colistin (CST) with ciprofloxacin (CIP) may improve the antibacterial effect against resistant Escherichia coli, offering an acceptable benefit-risk balance. This study aimed to quantify the interaction between ciprofloxacin and colistin in an in silico pharmacokinetic-pharmacodynamic model from in vitro static time-kill experiments (using strains with minimum inhibitory concentrations, MICCIP 0.023-1 mg/L and MICCST 0.5-0.75 mg/L). It was also sought to demonstrate an approach of simulating concentrations at the site of infection with population pharmacokinetic and whole-body physiologically based pharmacokinetic models to explore the clinical value of the combination when facing more resistant strains (using extrapolated strains with lower susceptibility). The combined effect in the final model was described as the sum of individual drug effects with a change in drug potency: for ciprofloxacin, concentration at half maximum killing rate (EC50) in combination was 160% of the EC50 in monodrug experiments, while for colistin, the change in EC50 was strain-dependent from 54.1% to 119%. The benefit of co-administrating a lower-than-commonly-administrated colistin dose with ciprofloxacin in terms of drug effect in comparison to either monotherapy was predicted in simulated bloodstream infections and pyelonephritis. The study illustrates the value of pharmacokinetic-pharmacodynamic modelling and simulation in streamlining rational development of antibiotic combinations.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Colistin , Computer Simulation , Escherichia coli , Microbial Sensitivity Tests , Ciprofloxacin/pharmacokinetics , Ciprofloxacin/pharmacology , Colistin/pharmacokinetics , Colistin/pharmacology , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Humans , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Drug Therapy, Combination , Models, Biological
7.
Ann Clin Microbiol Antimicrob ; 23(1): 48, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802894

ABSTRACT

BACKGROUND: Proteus mirabilis is a significant nosocomial pathogen that is frequently associated with a wide range of infections, necessitating heightened attention to mitigate potential health risks. Hence, this study was performed to investigate the impact of sub-minimum inhibitory concentrations (MICs) of ciprofloxacin (CIP) on Proteus mirabilis clinical isolates. METHODS: The sub-MICs of CIP were selected using the growth curve approach. The untreated and treated isolates with sub-MICs of CIP were assessed for their biofilm development, motilities on agar, and other virulence factors. The cell morphology of untreated and treated isolates with sub-MIC of CIP was explored using electron microscope. Moreover, the expression levels of the virulence genes in isolates were measured using quantitative real-time PCR. RESULTS: Data revealed that sub-MICs of CIP significantly (p < 0.05), in a concentration-dependent manner, inhibited biofilm formation and other virulence factors in the selected isolates. Electron microscope analysis showed cell enlargement and various abnormalities in the cell wall and membrane integrity. CONCLUSION: Sub-MICs of CIP exhibited inhibition of virulence and alterations in morphological integrity against P. mirabilis isolates.


Subject(s)
Anti-Bacterial Agents , Biofilms , Ciprofloxacin , Microbial Sensitivity Tests , Proteus Infections , Proteus mirabilis , Virulence Factors , Proteus mirabilis/drug effects , Proteus mirabilis/genetics , Ciprofloxacin/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Humans , Anti-Bacterial Agents/pharmacology , Proteus Infections/microbiology , Virulence Factors/genetics , Virulence/drug effects
8.
Front Public Health ; 12: 1365161, 2024.
Article in English | MEDLINE | ID: mdl-38807988

ABSTRACT

Introduction: Treatments that currently exist in the strategic national stockpile for acute radiation syndrome (ARS) focus on the hematopoietic subsyndrome, with no treatments on gastrointestinal (GI)-ARS. While the gut microbiota helps maintain host homeostasis by mediating GI epithelial and mucosal integrity, radiation exposure can alter gut commensal microbiota which may leave the host susceptible to opportunistic pathogens and serious sequelae such as sepsis. To mitigate the effects of hematopoietic ARS irradiation, currently approved treatments exist in the form of colony stimulating factors and antibiotics: however, there are few studies examining how these therapeutics affect GI-ARS and the gut microbiota. The aim of our study was to examine the longitudinal effects of Neulasta and/or ciprofloxacin treatment on the gut microbiota after exposure to 9.5 Gy 60Co gamma-radiation in mice. Methods: The gut microbiota of vehicle and drug-treated mice exposed to sham or gamma-radiation was characterized by shotgun sequencing with alpha diversity, beta diversity, and taxonomy analyzed on days 2, 4, 9, and 15 post-irradiation. Results: No significant alpha diversity differences were observed following radiation, while beta diversity shifts and taxonomic profiles revealed significant alterations in Akkermansia, Bacteroides, and Lactobacillus. Ciprofloxacin generally led to lower Shannon diversity and Bacteroides prevalence with increases in Akkermansia and Lactobacillus compared to vehicle treated and irradiated mice. While Neulasta increased Shannon diversity and by day 9 had more similar taxonomic profiles to sham than ciprofloxacin-or vehicle-treated irradiated animals. Combined therapy of Neulasta and ciprofloxacin induced a decrease in Shannon diversity and resulted in unique taxonomic profiles early post-irradiation, returning closer to vehicle-treated levels over time, but persistent increases in Akkermansia and Bacteroides compared to Neulasta alone. Discussion: This study provides a framework for the identification of microbial elements that may influence radiosensitivity, biodosimetry and the efficacy of potential therapeutics. Moreover, increased survival from H-ARS using these therapeutics may affect the symptoms and appearance of what may have been subclinical GI-ARS.


Subject(s)
Ciprofloxacin , Gastrointestinal Microbiome , Animals , Ciprofloxacin/pharmacology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/radiation effects , Mice , Anti-Bacterial Agents/pharmacology , Acute Radiation Syndrome/drug therapy , Gamma Rays , Male , Female
9.
Sex Transm Infect ; 100(4): 226-230, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38702191

ABSTRACT

OBJECTIVES: Antibiotic resistance in gonorrhoea is of significant public health concern with the emergence of resistance to last-line therapies such as ceftriaxone. Despite around half of Neisseria gonorrhoeae isolates tested in the UK being susceptible to ciprofloxacin, very little ciprofloxacin is used in clinical practice. Testing for the S91F mutation associated with ciprofloxacin resistance is now available in CE-marked assays and may reduce the requirement for ceftriaxone, but many patients are treated empirically, or as sexual contacts, which may limit any benefit. We describe the real-world impact of such testing on antimicrobial use and clinical outcomes in people found to have gonorrhoea in a large urban UK sexual health clinic. METHODS: Molecular ciprofloxacin resistance testing (ResistancePlus GC assay (SpeeDx)) was undertaken as an additional test after initial diagnosis (m2000 Realtime CT/NG assay (Abbott Molecular)) in those not already known to have had antimicrobial treatment. Data from a 6-month period (from March to September 2022) were analysed to determine treatment choice and treatment outcome. RESULTS: A total of 998 clinical samples tested positive for N. gonorrhoeae in 682 episodes of infection. Of the 560 (56%) samples eligible for resistance testing, 269 (48.0%) were reported as wild-type, 180 (32.1%) were predicted to be resistant, 63 (11.3%) had an indeterminate resistance profile, and in 48 (8.6%) samples, N. gonorrhoeae was not detected. Ciprofloxacin was prescribed in 172 (75%) of 228 episodes in which the wild-type strain was detected. Four (2%) of those treated with ciprofloxacin had a positive test-of-cure sample by NAAT, with no reinfection risk. All four had ciprofloxacin-susceptible infection by phenotypic antimicrobial susceptibility testing. CONCLUSIONS: In routine practice in a large UK clinic, molecular ciprofloxacin resistance testing led to a significant shift in antibiotic use, reducing use of ceftriaxone. Testing can be targeted to reduce unnecessary additional testing. Longer term impact on antimicrobial resistance requires ongoing surveillance.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Drug Resistance, Bacterial , Gonorrhea , Microbial Sensitivity Tests , Neisseria gonorrhoeae , Humans , Ciprofloxacin/therapeutic use , Ciprofloxacin/pharmacology , Gonorrhea/drug therapy , Gonorrhea/diagnosis , Gonorrhea/microbiology , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/genetics , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Male , Female , Adult , United Kingdom , Ceftriaxone/therapeutic use , Ceftriaxone/pharmacology , Mutation , Young Adult , Middle Aged
10.
Int J Pharm ; 658: 124208, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38723731

ABSTRACT

Pseudomonas aeruginosa (PA), a predominant pathogen in lung infections, poses significant challenges due to its biofilm formation, which is the primary cause of chronic and recalcitrant pulmonary infections. Bacteria within these biofilms exhibit heightened resistance to antibiotics compared to their planktonic counterparts, and their secreted toxins exacerbate lung infections. Diverging from traditional antibacterial therapy for biofilm eradication, this study introduces a novel dry powder inhalation containing muco-inert ciprofloxacin and colistin co-encapsulated liposomes (Cipro-Col-Lips) prepared using ultrasonic spray freeze drying (USFD) technique. This USFD dry powder is designed to efficiently deliver muco-inert Cipro-Col-Lips to the lungs. Once deposited, the liposomes rapidly diffuse into the airway mucus, reaching the biofilm sites. The muco-inert Cipro-Col-Lips neutralize the biofilm-secreted toxins and simultaneously trigger the release of their therapeutic payload, exerting a synergistic antibiofilm effect. Our results demonstrated that the optimal USFD liposomal dry powder formulation exhibited satisfactory in vitro aerosol performance in terms of fine particle fraction (FPF) of 44.44 ± 0.78 %, mass median aerodynamic diameter (MMAD) of 4.27 ± 0.21 µm, and emitted dose (ED) of 99.31 ± 3.31 %. The muco-inert Cipro-Col-Lips effectively penetrate the airway mucus and accumulate at the biofilm site, neutralizing toxins and safeguarding lung cells. The triggered release of ciprofloxacin and colistin works synergistically to reduce the biofilm's antibiotic resistance, impede the development of antibiotic resistance, and eliminate 99.99 % of biofilm-embedded bacteria, including persister bacteria. Using a PA-beads induced biofilm-associated lung infection mouse model, the in vivo efficacy of this liposomal dry powder aerosol was tested, and the results demonstrated that this liposomal dry powder aerosol achieved a 99.7 % reduction in bacterial colonization, and significantly mitigated inflammation and pulmonary fibrosis. The USFD dry powder inhalation containing muco-inert Cipro-Col-Lips emerges as a promising therapeutic strategy for treating PA biofilm-associated lung infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Ciprofloxacin , Colistin , Dry Powder Inhalers , Liposomes , Pseudomonas Infections , Pseudomonas aeruginosa , Ciprofloxacin/administration & dosage , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Biofilms/drug effects , Colistin/administration & dosage , Colistin/pharmacology , Administration, Inhalation , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Pseudomonas Infections/drug therapy , Mice , Aerosols , Lung/microbiology , Lung/drug effects , Powders , Female , Particle Size
11.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732134

ABSTRACT

Ciprofloxacin is a widely used antibiotic in the fluoroquinolone class. It is widely acknowledged by various researchers worldwide, and it has been documented to have a broad range of other pharmacological activities, such as anticancer, antiviral, antimalarial activities, etc. Researchers have been exploring the synthesis of ciprofloxacin derivatives with enhanced biological activities or tailored capability to target specific pathogens. The various biological activities of some of the most potent and promising ciprofloxacin derivatives, as well as the synthetic strategies used to develop them, are thoroughly reviewed in this paper. Modification of ciprofloxacin via 4-oxo-3-carboxylic acid resulted in derivatives with reduced efficacy against bacterial strains. Hybrid molecules containing ciprofloxacin scaffolds displayed promising biological effects. The current review paper provides reported findings on the development of novel ciprofloxacin-based molecules with enhanced potency and intended therapeutic activities which will be of great interest to medicinal chemists.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Animals , Structure-Activity Relationship
12.
Water Res ; 257: 121721, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38728782

ABSTRACT

Microplastics (MPs) waste is widespread globally in water systems. The opportunistic human pathogen Pseudomonas aeruginosa can cause serious acute and chronic infections that are notoriously difficult to treat. Ciprofloxacin (CIP) is broadly applied as an anti-P. aeruginosa drug. A growing evidence reveals that antibiotic-resistance genes-carrying Pseudomonas aeruginosa were detected on MPs forming plastisphere due to their adsorbability along with high occurrence of CIP in water environments. The MPs-niched CIP-resistant P. aeruginosa has been likely to emerge as an unignorable public health issue. Here, we offered a novel approach to assess the development of CIP-resistant P. aeruginosa under MPs-antibiotic coexistence at a water region scale. By combing the adsorption isotherm models used to estimate CIP condensation around MPs and a pharmacokinetic/pharmacodynamic-based microbial population dynamic model, we predicted the P. aeruginosa development on CIP-adsorbed MPs in waters. Our assessment revealed a high antibiotic resistance in the P. aeruginosa populations (∼50 %) with a wider range of waterborne total cell counts (∼10-2-104 cfu mL-1) among water regions in that the resistance proportion was primarily determined by CIP pollution level and relative abundance of various polymer type of MPs. We implicate that water region-specific MPs were highly likely to provide media for P. aeruginosa propagation. Our results highlight the importance of antibiotic-resistant pathogen colonization-emerging environmental medium interactions when addressing global threat from MPs pollution, in the context of MPs-antibiotics co-contamination assessment and for the continued provision of water system management.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Microplastics , Pseudomonas aeruginosa , Pseudomonas aeruginosa/drug effects , Ciprofloxacin/pharmacology , Anti-Bacterial Agents/pharmacology , Water Pollutants, Chemical , Drug Resistance, Bacterial
13.
Int J Biol Macromol ; 269(Pt 1): 131794, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697434

ABSTRACT

A middle ear infection occurs due to the presence of several microorganisms behind the eardrum (tympanic membrane) and is very challenging to treat due to its unique location and requires a well-designed treatment. If not treated properly, the infection can result in severe symptoms and unavoidable side effects. In this study, excellent biocompatible ethyl cellulose (EC) and biodegradable polyhydroxybutyrate (PHB) biopolymer were used to fabricate drug-loaded nanofiber scaffolds using an electrospinning technique to overcome antibiotic overdose and insufficient efficacy of drug release during treatment. PHB polymer was produced from Halomonas sp., and the purity of PHB was found to around be 90 %. Additionally, ciprofloxacin (CIP) and amoxicillin (AMX) are highly preferable since both drugs are highly effective against gram-negative and gram-positive bacteria to treat several infections. Obtained smooth nanofibers were between 116.24 and 171.82 nm in diameter and the addition of PHB polymer and antibiotics improved the morphology of the nanofiber scaffolds. Thermal properties of the nanofiber scaffolds were tested and the highest Tg temperature resulted at 229 °C. The mechanical properties of the scaffolds were tested, and the highest tensile strength resulted in 4.65 ± 6.33 MPa. Also, drug-loaded scaffolds were treated against the most common microorganisms that cause the infection, such as S.aureus, E.coli, and P.aeruginosa, and resulted in inhibition zones between 10 and 21 mm. MTT assay was performed by culturing human adipose-derived mesenchymal stem cells (hAD MSCs) on the scaffolds. The morphology of the hAD MSCs' attachment was tested with SEM analysis and hAD MSCs were able to attach, spread, and live on each scaffold even on the day of 7. The cumulative drug release kinetics of CIP and AMX from drug-loaded scaffolds were analysed in phosphate-buffered saline (pH: 7.4) within different time intervals of up to 14 days using a UV spectrophotometer. Furthermore, the drug release showed that the First-Order and Korsmeyer-Peppas models were the most suitable kinetic models. Animal testing was performed on SD rats, matrix and collagen deposition occurred on days 5 and 10, which were observed using Hematoxylin-eosin and Masson's trichrome staining. At the highest drug concentration, a better repair effect was observed. Results were promising and showed potential for novel treatment.


Subject(s)
Amoxicillin , Anti-Bacterial Agents , Cellulose , Ciprofloxacin , Nanofibers , Cellulose/chemistry , Cellulose/analogs & derivatives , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Nanofibers/chemistry , Animals , Rats , Amoxicillin/pharmacology , Amoxicillin/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacology , Humans , Otitis Media/drug therapy , Otitis Media/microbiology , Polyesters/chemistry , Drug Liberation , Tissue Scaffolds/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Prohibitins , Drug Carriers/chemistry , Male
14.
PLoS One ; 19(5): e0298746, 2024.
Article in English | MEDLINE | ID: mdl-38787890

ABSTRACT

Enterohemorrhagic E. coli (EHEC) is considered to be the most dangerous pathotype of E. coli, as it causes severe conditions such as hemorrhagic colitis (HC) and hemolytic uremic syndrome (HUS). Antibiotic treatment of EHEC infections is generally not recommended since it may promote the production of the Shiga toxin (Stx) and lead to worsened symptoms. This study explores how exposure to the fluoroquinolone ciprofloxacin reorganizes the transcriptome and proteome of EHEC O157:H7 strain EDL933, with special emphasis on virulence-associated factors. As expected, exposure to ciprofloxacin caused an extensive upregulation of SOS-response- and Stx-phage proteins, including Stx. A range of other virulence-associated factors were also upregulated, including many genes encoded by the LEE-pathogenicity island, the enterohemolysin gene (ehxA), as well as several genes and proteins involved in LPS production. However, a large proportion of the genes and proteins (17 and 8%, respectively) whose expression was upregulated upon ciprofloxacin exposure (17 and 8%, respectively) are not functionally assigned. This indicates a knowledge gap in our understanding of mechanisms involved in EHECs response to antibiotic-induced stress. Altogether, the results contribute to better understanding of how exposure to ciprofloxacin influences the virulome of EHEC and generates a knowledge base for further studies on how EHEC responds to antibiotic-induced stress. A deeper understanding on how EHEC responds to antibiotics will facilitate development of novel and safer treatments for EHEC infections.


Subject(s)
Ciprofloxacin , Proteomics , Transcriptome , Ciprofloxacin/pharmacology , Proteomics/methods , Virulence/drug effects , Transcriptome/drug effects , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology , Virulence Factors/genetics , Virulence Factors/metabolism , Proteome/metabolism , Gene Expression Profiling , Humans
15.
Microbiol Spectr ; 12(6): e0007124, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38700321

ABSTRACT

Novel antimicrobial agents are needed to combat antimicrobial resistance. This study tested novel pentafluorosulfanyl-containing triclocarban analogs for their potential antibacterial efficacy. Standard procedures were used to produce pentafluorosulfanyl-containing triclocarban analogs. Twenty new compounds were tested against seven Gram-positive and Gram-negative indicator strains as well as 10 clinical isolates for their antibacterial and antibiofilm activity. Mechanistic investigations focused on damage to cell membrane, oxidizing reduced thiols, iron-sulfur clusters, and oxidative stress to explain the compounds' activity. Safety profiles were assessed using cytotoxicity experiments in eukaryotic cell lines. Following screening, selected components had significantly better antibacterial and antibiofilm activity against Gram-positive bacteria in lower concentrations in comparison to ciprofloxacin and gentamycin. For instance, one compound had a minimum inhibitory concentration of <0.0003 mM, but ciprofloxacin had 0.08 mM. Mechanistic studies show that these novel compounds do not affect reduced thiol content, iron-sulfur clusters, or hydrogen peroxide pathways. Their impact comes from Gram-positive bacterial cell membrane damage. Tests on cell culture toxicity and host component safety showed promise. Novel diarylurea compounds show promise as Gram-positive antimicrobials. These compounds offer prospects for study and optimization. IMPORTANCE: The rise of antibiotic resistance among bacterial pathogens poses a significant threat to global health, underscoring the urgent need for novel antimicrobial agents. This study presents research on a promising class of novel compounds with potent antibacterial properties against Gram-positive bacteria, notably Staphylococcus aureus and MRSA. What sets these novel analogs apart is their superior efficacy at substantially lower concentrations compared with commonly used antibiotics like ciprofloxacin and gentamycin. Importantly, these compounds act by disrupting the bacterial cell membrane, offering a unique mechanism that could potentially circumvent existing resistance mechanisms. Preliminary safety assessments also highlight their potential for therapeutic use. This study not only opens new avenues for combating antibiotic-resistant infections but also underscores the importance of innovative chemical approaches in addressing the global antimicrobial resistance crisis.


Subject(s)
Anti-Bacterial Agents , Carbanilides , Gram-Positive Bacteria , Microbial Sensitivity Tests , Carbanilides/pharmacology , Carbanilides/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Positive Bacteria/drug effects , Humans , Biofilms/drug effects , Gram-Negative Bacteria/drug effects , Ciprofloxacin/pharmacology
16.
Microbiology (Reading) ; 170(5)2024 May.
Article in English | MEDLINE | ID: mdl-38739119

ABSTRACT

Introduction. Bacterial keratitis, particularly caused by Pseudomonas aeruginosa, is challenging to treat because of multi-drug tolerance, often associated with the formation of biofilms. Antibiotics in development are typically evaluated against planktonic bacteria in a culture medium, which may not accurately represent the complexity of infections in vivo.Hypothesis/Gap Statement. Developing a reliable, economic ex vivo keratitis model that replicates some complexity of tissue infections could facilitate a deeper understanding of antibiotic efficacy, thus aiding in the optimization of treatment strategies for bacterial keratitis.Methodology. Here we investigated the efficacy of three commonly used antibiotics (gentamicin, ciprofloxacin and meropenem) against Pseudomonas aeruginosa cytotoxic strain PA14 and invasive strain PA01 using an ex vivo porcine keratitis model.Results. Both strains of P. aeruginosa were susceptible to the MIC of the three tested antibiotics. However, significantly higher concentrations were necessary to inhibit bacterial growth in the minimum biofilm eradication concentration (MBEC) assay, with both strains tolerating concentrations greater than 512 mg l-1 of meropenem. When MIC and higher concentrations than MBEC (1024 mg l-1) of antibiotics were applied, ciprofloxacin exhibited the highest potency against both P. aeruginosa strains, followed by meropenem, while gentamicin showed the least potency. Despite this, none of the antibiotic concentrations used effectively cleared the infection, even after 18 h of continuous exposure.Conclusions. Further exploration of antibiotic concentrations and aligning dosing with clinical studies to validate the model is needed. Nonetheless, our ex vivo porcine keratitis model could be a valuable tool for assessing antibiotic efficacy.


Subject(s)
Anti-Bacterial Agents , Biofilms , Ciprofloxacin , Disease Models, Animal , Keratitis , Microbial Sensitivity Tests , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Anti-Bacterial Agents/pharmacology , Swine , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Biofilms/drug effects , Keratitis/microbiology , Keratitis/drug therapy , Ciprofloxacin/pharmacology , Gentamicins/pharmacology , Meropenem/pharmacology
17.
Int J Biol Macromol ; 270(Pt 2): 132350, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750839

ABSTRACT

Wound biofilms represent an elusive conundrum in contemporary treatment and diagnostic options, accredited to their escalating antibiotic resistance and interference in chronic wound healing processes. Here, we developed mesoporous polydopamine (mPDA) nanoparticles, and grafted with rhodamine B (Rb) as biofilm lipase responsive detection probe, followed by π - π stacking mediated ciprofloxacin (CIP) loading to create mP-Rb@CIP nanoparticles. mPDA NPs with a melanin structure could quench fluorescence emissions of Rb. Once encountering biofilm in vivo, the ester bond in Rb and mPDA is hydrolyzed by elevated lipase concentrations, triggering the liberation of Rb and restore fluorescence emissions to achieve real-time imaging of biofilm-infected wounds. Afterwards, the 808 nm near-infrared (NIR) illumination initiates a spatiotemporal controlled antibacterial photothermal therapy (PTT), boosting its effectiveness through photothermal-triggered CIP release for synergistic biofilm eradication. The mP-Rb@CIP platform exhibits dual diagnostic and therapeutic functions, efficaciously treating biofilm-infected wounds in vivo and in vitro. Particularly, the mP-Rb@CIP/NIR procedure expedites wound-healing by alleviating oxidative stress, modulating inflammatory mediators, boosting collagen synthesis, and promoting angiogenesis. Taken together, the theranostic nanosystem strategy holds significant potential for addressing wound biofilm-associated infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Indoles , Lipase , Nanoparticles , Polymers , Indoles/chemistry , Indoles/pharmacology , Biofilms/drug effects , Polymers/chemistry , Lipase/metabolism , Lipase/chemistry , Nanoparticles/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mice , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/therapy , Photothermal Therapy/methods , Rhodamines/chemistry , Wound Healing/drug effects , Humans
18.
Sci Rep ; 14(1): 9354, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38653744

ABSTRACT

Phage-antibiotic combinations to treat bacterial infections are gaining increased attention due to the synergistic effects often observed when applying both components together. Most studies however focus on a single pathogen, although in many clinical cases multiple species are present at the site of infection. The aim of this study was to investigate the anti-biofilm activity of phage-antibiotic/antifungal combinations on single- and dual-species biofilms formed by P. aeruginosa and the fungal pathogen Candida albicans. The Pseudomonas phage Motto in combination with ciprofloxacin had significant anti-biofilm activity. We then compared biofilms formed by P. aeruginosa alone with the dual-species biofilms formed by bacteria and C. albicans. Here, we found that the phage together with the antifungal fluconazole was active against 6-h-old dual-species biofilms but showed only negligible activity against 24-h-old biofilms. This study lays the first foundation for potential therapeutic approaches to treat co-infections caused by bacteria and fungi using phage-antibiotic combinations.


Subject(s)
Anti-Bacterial Agents , Antifungal Agents , Biofilms , Candida albicans , Ciprofloxacin , Pseudomonas Phages , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/virology , Antifungal Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Phages/physiology , Candida albicans/drug effects , Candida albicans/physiology , Ciprofloxacin/pharmacology , Fluconazole/pharmacology , Microbial Sensitivity Tests
19.
ACS Appl Mater Interfaces ; 16(15): 18360-18385, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38573741

ABSTRACT

Bacterial keratitis (BK) causes visual morbidity/blindness if not treated effectively. Here, ciprofloxacin (CIP)-loaded nanoparticles (NPs) using glycol chitosan (GC) and poly(lactic acid) (PLA) conjugate at three different ratios (CIP@GC(PLA) NPs (1:1,5,15)) were fabricated. CIP@GC(PLA) NPs (1:1) were more effective than other tested ratios, indicating the importance of optimal hydrophobic/hydrophilic balance for corneal penetration and preventing bacterial invasion. The CIP@GC(PLA) (NPs) (1:1) realized the highest association with human corneal epithelial cells, which were nonirritant to the hen's egg-chorioallantoic membrane test (HET-CAM test) and demonstrated significant antibacterial response in the in vitro minimum inhibitory, bactericidal, live-dead cells, zone of inhibition, and biofilm inhibition assays against the keratitis-inducing pathogen Pseudomonas aeruginosa. The antiquorum sensing activity of GC has been explored for the first time. The NPs disrupted the bacterial quorum sensing by inhibiting the production of virulence factors, including acyl homoserine lactones, pyocyanin, and motility, and caused significant downregulation of quorum sensing associated genes. In the in vivo studies, CIP@GC(PLA) NPs (1:1) displayed ocular retention in vivo (∼6 h) and decreased the opacity and the bacterial load effectively. Overall, the CIP@GC(PLA) NP (1:1) is a biofilm-disrupting antiquorum sensing treatment regimen with clinical translation potential in BK.


Subject(s)
Chitosan , Eye Infections, Bacterial , Keratitis , Nanoparticles , Animals , Female , Humans , Ciprofloxacin/pharmacology , Chickens , Biofilms , Anti-Bacterial Agents/pharmacology , Polyesters/pharmacology , Quorum Sensing , Bacteria , Pseudomonas aeruginosa
20.
Sex Transm Infect ; 100(3): 173-180, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38575313

ABSTRACT

OBJECTIVES: International travel combined with sex may contribute to dissemination of antimicrobial-resistant (AMR) Neisseria gonorrhoeae (Ng). To assess the role of travel in Ng strain susceptibility, we compared minimum inhibitory concentrations (MICs) for five antibiotics (ie, azithromycin, ceftriaxone, cefotaxime, cefixime and ciprofloxacin) in strains from clients with an exclusively Dutch sexual network and clients with an additional international sexual network. METHODS: From 2013 to 2019, we recorded recent residence of sexual partners of clients (and of their partners) with Ng at the Center for Sexual Health of Amsterdam. We categorised clients as having: (1) exclusively sexual partners residing in the Netherlands ('Dutch only') or (2) at least one partner residing outside the Netherlands. We categorised the country of residence of sexual partners by World Bank/EuroVoc regions. We analysed the difference of log-transformed MIC of Ng strains between categories using linear or hurdle regression for each antibiotic. RESULTS: We included 3367 gay and bisexual men who had sex with men (GBMSM), 516 women and 525 men who exclusively had sex with women (MSW) with Ng. Compared with GBMSM with a 'Dutch only' network, GBMSM with: (1) a Western European network had higher MICs for ceftriaxone (ß=0.19, 95% CI=0.08 to 0.29), cefotaxime (ß=0.19, 95% CI=0.08 to 0.31) and cefixime (ß=0.06, 95% CI=0.001 to 0.11); (2) a Southern European network had a higher MIC for cefixime (ß=0.10, 95% CI=0.02 to 0.17); and (3) a sub-Saharan African network had a lower MIC for ciprofloxacin (ß=-1.79, 95% CI=-2.84 to -0.74). In women and MSW, higher MICs were found for ceftriaxone in clients with a Latin American and Caribbean network (ß=0.26, 95% CI=0.02 to 0.51). CONCLUSIONS: For three cephalosporin antibiotics, we found Ng strains with slightly higher MICs in clients with partner(s) from Europe or Latin America and the Caribbean. International travel might contribute to the spread of Ng with lower susceptibility. More understanding of the emergence of AMR Ng is needed.


Subject(s)
Anti-Infective Agents , Gonorrhea , Sexual Health , Male , Female , Humans , Neisseria gonorrhoeae , Ceftriaxone/pharmacology , Ceftriaxone/therapeutic use , Cefixime/pharmacology , Gonorrhea/drug therapy , Gonorrhea/epidemiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/pharmacology , Ciprofloxacin/therapeutic use , Azithromycin/pharmacology , Cefotaxime/pharmacology , Microbial Sensitivity Tests , Anti-Infective Agents/pharmacology , Drug Resistance, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...