Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.103
Filter
1.
Ann Clin Microbiol Antimicrob ; 23(1): 60, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965559

ABSTRACT

BACKGROUND: Gram-negative bacteria (GNB) are becoming increasingly resistant to a wide variety of antibiotics. There are currently limited treatments for GNB, and the combination of antibiotics with complementary mechanisms has been reported to be a feasible strategy for treating GNB infection. The inability to cross the GNB outer membrane (OM) is an important reason that a broad spectrum of Gram-positive only class of antibiotics (GPOAs) is lacking. Polymyxins may help GPOAs to permeate by disrupting OM of GNB. OBJECTIVE: To identify what kind of GPOAs can be aided to broaden their anti-GNB spectrum by polymyxins, we systematically investigated the synergy of eight GPOAs in combination with colistin (COL) and polymyxin B (PMB) against GNB in vitro. METHODS: The synergistic effect of COL or PMB and GPOAs combinations against GNB reference strains and clinical isolates were determined by checkerboard tests. The killing kinetics of the combinations were assessed using time-kill assays. RESULTS: In the checkerboard tests, polymyxins-GPOAs combinations exert synergistic effects characterized by species and strain specificity. The synergistic interactions on P. aeruginosa strains are significantly lower than those on strains of A. baumannii, K. pneumoniae and E. coli. Among all the combinations, COL has shown the best synergistic effect in combination with dalbavancin (DAL) or oritavancin (ORI) versus almost all of the strains tested, with FICIs from 0.16 to 0.50 and 0.13 to < 0.28, respectively. In addition, the time-kill assays demonstrated that COL/DAL and COL/ORI had sustained bactericidal activity. CONCLUSIONS: Our results indicated that polymyxins could help GPOAs to permeate the OM of specific GNB, thus showed synergistic effects and bactericidal effects in the in vitro assays. In vivo combination studies should be further conducted to validate the results of this study.


Subject(s)
Anti-Bacterial Agents , Colistin , Drug Synergism , Gram-Negative Bacteria , Microbial Sensitivity Tests , Polymyxin B , Polymyxins , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Polymyxins/pharmacology , Polymyxin B/pharmacology , Humans , Colistin/pharmacology , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Pseudomonas aeruginosa/drug effects
2.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38942450

ABSTRACT

The increasing resistance to polymyxins in Acinetobacter baumannii has made it even more urgent to develop new treatments. Anti-virulence compounds have been researched as a new solution. Here, we evaluated the modification of virulence features of A. baumannii after acquiring resistance to polymyxin B. The results showed lineages attaining unstable resistance to polymyxin B, except for Ab7 (A. baumannii polymyxin B resistant lineage), which showed stable resistance without an associated fitness cost. Analysis of virulence by a murine sepsis model indicated diminished virulence in Ab7 (A. baumannii polymyxin B resistant lineage) compared with Ab0 (A. baumannii polymyxin B susceptible lineage). Similarly, downregulation of virulence genes was observed by qPCR at 1 and 3 h of growth. However, an increase in bauE, abaI, and pgAB expression was observed after 6 h of growth. Comparison analysis of Ab0, Ab7, and Pseudomonas aeruginosa suggested no biofilm formation by Ab7. In general, although a decrease in virulence was observed in Ab7 when compared with Ab0, some virulence feature that enables infection could be maintained. In light of this, virulence genes bauE, abaI, and pgAB showed a potential relevance in the maintenance of virulence in polymyxin B-resistant strains, making them promising anti-virulence targets.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Anti-Bacterial Agents , Drug Resistance, Bacterial , Polymyxin B , Polymyxin B/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Acinetobacter baumannii/genetics , Animals , Anti-Bacterial Agents/pharmacology , Virulence , Mice , Acinetobacter Infections/microbiology , Virulence Factors/genetics , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Sepsis/microbiology , Biofilms/drug effects , Biofilms/growth & development
3.
Nat Commun ; 15(1): 4733, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830951

ABSTRACT

Polymyxins are gram-negative antibiotics that target lipid A, the conserved membrane anchor of lipopolysaccharide in the outer membrane. Despite their clinical importance, the molecular mechanisms underpinning polymyxin activity remain unresolved. Here, we use surface plasmon resonance to kinetically interrogate interactions between polymyxins and lipid A and derive a phenomenological model. Our analyses suggest a lipid A-catalyzed, three-state mechanism for polymyxins: transient binding, membrane insertion, and super-stoichiometric cluster accumulation with a long residence time. Accumulation also occurs for brevicidine, another lipid A-targeting antibacterial molecule. Lipid A modifications that impart polymyxin resistance and a non-bactericidal polymyxin derivative exhibit binding that does not evolve into long-lived species. We propose that transient binding to lipid A permeabilizes the outer membrane and cluster accumulation enables the bactericidal activity of polymyxins. These findings could establish a blueprint for discovery of lipid A-targeting antibiotics and provide a generalizable approach to study interactions with the gram-negative outer membrane.


Subject(s)
Anti-Bacterial Agents , Lipid A , Polymyxin B , Surface Plasmon Resonance , Polymyxin B/pharmacology , Polymyxin B/metabolism , Lipid A/metabolism , Lipid A/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Microbial Sensitivity Tests , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/drug effects , Kinetics
4.
World J Microbiol Biotechnol ; 40(8): 243, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38869625

ABSTRACT

It was known that UVc irradiation increases the reactive oxygen species' (ROS) levels in bacteria hence the intervention of antioxidant enzymes and causes also changes in fatty acids (FAs) composition enabling bacteria to face antibiotics. Here, we intended to elucidate an interrelationship between SOD and susceptibility to antibiotics by studying FA membrane composition of UVc-treated P. aeruginosa PAO1 and its isogenic mutants (sodM, sodB and sod MB) membrane, after treatment with antibiotics. Swarmer mutants defective in genes encoding superoxide dismutase were pre-exposed to UVc radiations and then tested by disk diffusion method for their contribution to antibiotic tolerance in comparison with the P. aeruginosa wild type (WT). Moreover, fatty acid composition of untreated and UVc-treated WT and sod mutants was examined by Gaz chromatography and correlated to antibiotic resistance. Firstly, it has been demonstrated that after UVc exposure, swarmer WT strain, sodM and sodB mutants remain resistant to polymixin B, a membrane target antibiotic, through membrane unsaturation supported by the intervention of Mn-SOD after short UVc exposure and cyclopropanation of unsaturated FAs supported by the action of Fe-SOD after longer UVc exposure. However, resistance for ciprofloxacin is correlated with increase in saturated FAs. This correlation has been confirmed by a molecular docking approach showing that biotin carboxylase, involved in the initial stage of FA biosynthesis, exhibits a high affinity for ciprofloxacin. This investigation has explored the correlation of antibiotic resistance with FA content of swarmer P.aeruginosa pre-exposed to UVc radiations, confirmed to be antibiotic target dependant.


Subject(s)
Anti-Bacterial Agents , Mutation , Pseudomonas aeruginosa , Superoxide Dismutase , Ultraviolet Rays , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Cyclopropanes/pharmacology , Drug Resistance, Bacterial/genetics , Fatty Acids/metabolism , Ciprofloxacin/pharmacology , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Computer Simulation , Polymyxin B/pharmacology
5.
Am J Physiol Renal Physiol ; 327(1): F137-F145, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38779756

ABSTRACT

Polymyxins are a last-resort treatment option for multidrug-resistant gram-negative bacterial infections, but they are associated with nephrotoxicity. Gelofusine was previously shown to reduce polymyxin-associated kidney injury in an animal model. However, the mechanism(s) of renal protection has not been fully elucidated. Here, we report the use of a cell culture model to provide insights into the mechanisms of renal protection. Murine epithelial proximal tubular cells were exposed to polymyxin B. Cell viability, lactate dehydrogenase (LDH) release, polymyxin B uptake, mitochondrial superoxide production, nuclear morphology, and apoptosis activation were evaluated with or without concomitant gelofusine. A megalin knockout cell line was used as an uptake inhibition control. Methionine was included in selected experiments as an antioxidant control. A polymyxin B concentration-dependent reduction in cell viability was observed. Increased viability was observed in megalin knockout cells following comparable polymyxin B exposures. Compared with polymyxin B exposure alone, concomitant gelofusine significantly increased cell viability as well as reduced LDH release, polymyxin B uptake, mitochondrial superoxide, and apoptosis. Gelofusine and methionine were more effective at reducing renal cell injury in combination than either agent alone. In conclusion, the mechanisms of renal protection by gelofusine involve decreasing cellular drug uptake, reducing subsequent oxidative stress and apoptosis activation. These findings would be valuable for translational research into clinical strategies to attenuate drug-associated acute kidney injury.NEW & NOTEWORTHY Gelofusine is a gelatinous saline solution with the potential to attenuate polymyxin-associated nephrotoxicity. We demonstrated that the mechanisms of gelofusine renal protection involve reducing polymyxin B uptake by proximal tubule cells, limiting subsequent oxidative stress and apoptosis activation. In addition, gelofusine was more effective at reducing cellular injury than a known antioxidant control, methionine, and a megalin knockout cell line, indicating that gelofusine likely has additional pharmacological properties besides only megalin inhibition.


Subject(s)
Anti-Bacterial Agents , Apoptosis , Polymyxin B , Animals , Polymyxin B/pharmacology , Mice , Apoptosis/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Cell Line , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Acute Kidney Injury/chemically induced , Oxidative Stress/drug effects , L-Lactate Dehydrogenase/metabolism
6.
Microbiol Immunol ; 68(7): 224-236, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797913

ABSTRACT

Pathogenic bacteria form biofilms on epithelial cells, and most bacterial biofilms show increased production of membrane vesicles (MVs), also known as outer membrane vesicles in Gram-negative bacteria. Numerous studies have investigated the MVs released under planktonic conditions; however, the impact of MVs released from biofilms on immune responses remains unclear. This study aimed to investigate the characteristics and immunomodulatory activity of MVs obtained from both planktonic and biofilm cultures of Pseudomonas aeruginosa PAO1. The innate immune responses of macrophages to planktonic-derived MVs (p-MVs) and biofilm-derived MVs (b-MVs) were investigated by measuring the mRNA expression of proinflammatory cytokines. Our results showed that b-MVs induced a higher expression of inflammatory cytokines, including Il1b, Il6, and Il12p40, than p-MVs. The mRNA expression levels of Toll-like receptor 4 (Tlr4) differed between the two types of MVs, but not Tlr2. Polymyxin B significantly neutralized b-MV-mediated cytokine induction, suggesting that lipopolysaccharide of native b-MVs is the origin of the immune response. In addition, heat-treated or homogenized b-MVs induced the mRNA expression of cytokines, including Tnfa, Il1b, Il6, and Il12p40. Heat treatment of MVs led to increased expression of Tlr2 but not Tlr4, suggesting that TLR2 ligands play a role in detecting the pathogen-associated molecular patterns in lysed MVs. Taken together, our data indicate that potent immunomodulatory MVs are produced in P. aeruginosa biofilms and that this behavior could be a strategy for the bacteria to infect host cells. Furthermore, our findings would contribute to developing novel vaccines using MVs.


Subject(s)
Biofilms , Cytokines , Macrophages , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/physiology , Biofilms/growth & development , Cytokines/metabolism , Mice , Animals , Macrophages/immunology , Macrophages/microbiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/metabolism , Immunity, Innate , Polymyxin B/pharmacology , RAW 264.7 Cells , Immunologic Factors/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Lipopolysaccharides , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Anal Chem ; 96(23): 9317-9324, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38818541

ABSTRACT

Inaccurate or cumbersome clinical pathogen diagnosis between Gram-positive bacteria (G+) and Gram-negative (G-) bacteria lead to delayed clinical therapeutic interventions. Microelectrode-based electrochemical sensors exhibit the significant advantages of rapid response and minimal sample consumption, but the loading capacity and discrimination precision are weak. Herein, we develop reversible fusion-fission MXene-based fiber microelectrodes for G+/G- bacteria analysis. During the fissuring process, the spatial utilization, loading capacity, sensitivity, and selectivity of microelectrodes were maximized, and polymyxin B and vancomycin were assembled for G+/G- identification. The surface-tension-driven reversible fusion facilitated its reusability. A deep learning model was further applied for the electrochemical impedance spectroscopy (EIS) identification in diverse ratio concentrations of G+ and G- of (1:100-100:1) with higher accuracy (>93%) and gave predictable detection results for unknown samples. Meanwhile, the as-proposed sensing platform reached higher sensitivity toward E. coli (24.3 CFU/mL) and S. aureus (37.2 CFU/mL) in 20 min. The as-proposed platform provides valuable insights for bacterium discrimination and quantification.


Subject(s)
Microelectrodes , Gram-Positive Bacteria/isolation & purification , Gram-Negative Bacteria/isolation & purification , Escherichia coli/isolation & purification , Staphylococcus aureus/isolation & purification , Electrochemical Techniques/instrumentation , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/analysis , Polymyxin B/chemistry , Polymyxin B/pharmacology , Dielectric Spectroscopy
8.
Microbiology (Reading) ; 170(5)2024 May.
Article in English | MEDLINE | ID: mdl-38739436

ABSTRACT

Endolysins are bacteriophage (or phage)-encoded enzymes that catalyse the peptidoglycan breakdown in the bacterial cell wall. The exogenous action of recombinant phage endolysins against Gram-positive organisms has been extensively studied. However, the outer membrane acts as a physical barrier when considering the use of recombinant endolysins to combat Gram-negative bacteria. This study aimed to evaluate the antimicrobial activity of the SAR-endolysin LysKpV475 against Gram-negative bacteria as single or combined therapies, using an outer membrane permeabilizer (polymyxin B) and a phage, free or immobilized in a pullulan matrix. In the first step, the endolysin LysKpV475 in solution, alone and combined with polymyxin B, was tested in vitro and in vivo against ten Gram-negative bacteria, including highly virulent strains and multidrug-resistant isolates. In the second step, the lyophilized LysKpV475 endolysin was combined with the phage phSE-5 and investigated, free or immobilized in a pullulan matrix, against Salmonella enterica subsp. enterica serovar Typhimurium ATCC 13311. The bacteriostatic action of purified LysKpV475 varied between 8.125 µg ml-1 against Pseudomonas aeruginosa ATCC 27853, 16.25 µg ml-1 against S. enterica Typhimurium ATCC 13311, and 32.50 µg ml-1 against Klebsiella pneumoniae ATCC BAA-2146 and Enterobacter cloacae P2224. LysKpV475 showed bactericidal activity only for P. aeruginosa ATCC 27853 (32.50 µg ml-1) and P. aeruginosa P2307 (65.00 µg ml-1) at the tested concentrations. The effect of the LysKpV475 combined with polymyxin B increased against K. pneumoniae ATCC BAA-2146 [fractional inhibitory concentration index (FICI) 0.34; a value lower than 1.0 indicates an additive/combined effect] and S. enterica Typhimurium ATCC 13311 (FICI 0.93). A synergistic effect against S. enterica Typhimurium was also observed when the lyophilized LysKpV475 at ⅔ MIC was combined with the phage phSE-5 (m.o.i. of 100). The lyophilized LysKpV475 immobilized in a pullulan matrix maintained a significant Salmonella reduction of 2 logs after 6 h of treatment. These results demonstrate the potential of SAR-endolysins, alone or in combination with other treatments, in the free form or immobilized in solid matrices, which paves the way for their application in different areas, such as in biocontrol at the food processing stage, biosanitation of food contact surfaces and biopreservation of processed food in active food packing.


Subject(s)
Anti-Bacterial Agents , Endopeptidases , Glucans , Polymyxin B , Salmonella Phages , Endopeptidases/pharmacology , Endopeptidases/chemistry , Endopeptidases/metabolism , Polymyxin B/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Salmonella Phages/genetics , Salmonella Phages/physiology , Salmonella Phages/chemistry , Glucans/chemistry , Glucans/pharmacology , Animals , Microbial Sensitivity Tests , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/virology , Mice , Salmonella typhimurium/virology , Salmonella typhimurium/drug effects , Bacteriophages/physiology , Bacteriophages/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/pharmacology , Viral Proteins/chemistry
9.
Int J Antimicrob Agents ; 64(1): 107189, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38697578

ABSTRACT

The main objective of this study was to assess the effect of rich artificial cation-adjusted Mueller-Hinton broth (CAMHB) on the growth of three strains of Acinetobacter baumannii (ATCC 19606 and two clinical strains), either susceptible or resistant to polymyxin B (PMB), and on PMB bactericidal activity. A pharmacokinetic (PK)/pharmacodynamic (PD) modelling approach was used to characterize the effect of PMB in various conditions. Time-kill experiments were performed using undiluted CAMHB or CAMHB diluted to 50%, 25% and 10%, with or without Ca2+ and Mg2+ compensation (known to affect PMB activity), and with PMB concentrations ranging from 0.25 to 256 mg/L based on the strain's MIC. For each strain, time-kill replicates were modelled using NONMEM. Unexpectedly, dilution of CAMHB by up to 10-fold did not affect the growth rate of any of the three strains in the absence of PMB. However, the bactericidal activity of PMB increased with medium dilution, resulting in a reduction in the apparent bacterial regrowth of the various strains observed after a few hours. Data for each strain were well characterized by a PK/PD model, with two bacterial subpopulations with different susceptibility to PMB (more susceptible and less susceptible). The impact of medium dilution and cation compensation showed relatively high, unexplained between-strain variability. Further studies are needed to characterize the mechanism underlying the medium dilution effect.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Culture Media , Microbial Sensitivity Tests , Polymyxin B , Acinetobacter baumannii/drug effects , Polymyxin B/pharmacology , Polymyxin B/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Humans , Culture Media/chemistry , Microbial Viability/drug effects
10.
Arch Microbiol ; 206(6): 272, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38772980

ABSTRACT

Phage-encoded endolysins have emerged as a potential substitute to conventional antibiotics due to their exceptional benefits including host specificity, rapid host killing, least risk of resistance. In addition to their antibacterial potency and biofilm eradication properties, endolysins are reported to exhibit synergism with other antimicrobial agents. In this study, the synergistic potency of endolysins was dissected with antimicrobial peptides to enhance their therapeutic effectiveness. Recombinantly expressed and purified bacteriophage endolysin [T7 endolysin (T7L); and T4 endolysin (T4L)] proteins have been used to evaluate the broad-spectrum antibacterial efficacy using different bacterial strains. Antibacterial/biofilm eradication studies were performed in combination with different antimicrobial peptides (AMPs) such as colistin, nisin, and polymyxin B (PMB) to assess the endolysin's antimicrobial efficacy and their synergy with AMPs. In combination with T7L, polymyxin B and colistin effectively eradicated the biofilm of Pseudomonas aeruginosa and exhibited a synergistic effect. Further, a combination of T4L and nisin displayed a synergistic effect against Staphylococcus aureus biofilms. In summary, the obtained results endorse the theme of combinational therapy consisting of endolysins and AMPs as an effective remedy against the drug-resistant bacterial biofilms that are a serious concern in healthcare settings.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Biofilms , Drug Synergism , Endopeptidases , Microbial Sensitivity Tests , Pseudomonas aeruginosa , Staphylococcus aureus , Biofilms/drug effects , Endopeptidases/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Pseudomonas aeruginosa/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Nisin/pharmacology , Nisin/chemistry , Polymyxin B/pharmacology , Bacteriophages , Colistin/pharmacology , Bacteriophage T4/drug effects , Bacteriophage T4/physiology , Bacteriophage T7/drug effects , Bacteriophage T7/genetics
11.
J Extracell Vesicles ; 13(5): e12447, 2024 May.
Article in English | MEDLINE | ID: mdl-38766978

ABSTRACT

The continuous emergence of multidrug-resistant bacterial pathogens poses a major global healthcare challenge, with Klebsiella pneumoniae being a prominent threat. We conducted a comprehensive study on K. pneumoniae's antibiotic resistance mechanisms, focusing on outer membrane vesicles (OMVs) and polymyxin, a last-resort antibiotic. Our research demonstrates that OMVs protect bacteria from polymyxins. OMVs derived from Polymyxin B (PB)-stressed K. pneumoniae exhibited heightened protective efficacy due to increased vesiculation, compared to OMVs from unstressed Klebsiella. OMVs also shield bacteria from different bacterial families. This was validated ex vivo and in vivo using precision cut lung slices (PCLS) and Galleria mellonella. In all models, OMVs protected K. pneumoniae from PB and reduced the associated stress response on protein level. We observed significant changes in the lipid composition of OMVs upon PB treatment, affecting their binding capacity to PB. The altered binding capacity of single OMVs from PB stressed K. pneumoniae could be linked to a reduction in the lipid A amount of their released vesicles. Although the amount of lipid A per vesicle is reduced, the overall increase in the number of vesicles results in an increased protection because the sum of lipid A and therefore PB binding sites have increased. This unravels the mechanism of the altered PB protective efficacy of OMVs from PB stressed K. pneumoniae compared to control OMVs. The lipid A-dependent protective effect against PB was confirmed in vitro using artificial vesicles. Moreover, artificial vesicles successfully protected Klebsiella from PB ex vivo and in vivo. The findings indicate that OMVs act as protective shields for bacteria by binding to polymyxins, effectively serving as decoys and preventing antibiotic interaction with the cell surface. Our findings provide valuable insights into the mechanisms underlying antibiotic cross-protection and offer potential avenues for the development of novel therapeutic interventions to address the escalating threat of multidrug-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents , Klebsiella pneumoniae , Polymyxin B , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/drug effects , Anti-Bacterial Agents/pharmacology , Animals , Polymyxin B/pharmacology , Bacterial Outer Membrane/metabolism , Polymyxins/pharmacology , Extracellular Vesicles/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/metabolism , Microbial Sensitivity Tests , Drug Resistance, Multiple, Bacterial/drug effects
12.
J Colloid Interface Sci ; 667: 529-542, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38653074

ABSTRACT

Diabetic wounds are characterized by chronic trauma, with long-term non-healing attributed to persistent inflammation and recurrent bacterial infections. Exacerbation of the inflammatory response is largely due to increased levels of reactive oxygen species (ROS). In this study, catalase (CAT) was used as a biological template to synthesize nanozyme-supported natural enzymes (CAT-Mn(SH)x) using a biomimetic mineralization method. Subsequently, polymyxin B (CAT-Mn(SH)x@PMB) was immobilized on its surface through electrostatic assembly. CAT-Mn(SH)x@PMB demonstrates the ability for slow and sustained release of hydrogen sulfide (H2S). Finally, CAT-Mn(SH)x@PMB loaded microneedles (MNs) substrate were synthesized using polyvinyl alcohol (PVA) and hydroxyethyl methacrylate (HEMA), and named CAT-(MnSH)x@PMB-MNs. It exhibited enhanced enzyme and antioxidant activities, along with effective antibacterial properties. Validation findings indicate that it can up-regulate the level of M2 macrophages and reduce the level of pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). Additionally, it promotes angiogenesis and rapid nerve regeneration, thereby facilitating wound healing through its dual anti-inflammatory and antibacterial effects. Hence,this study introduces a time-space tissue-penetrating and soluble microneedle patch with dual anti-inflammatory and antibacterial effects for the treatment of diabetic wounds.


Subject(s)
Anti-Bacterial Agents , Catalase , Needles , Polymyxin B , Wound Healing , Polymyxin B/pharmacology , Polymyxin B/chemistry , Polymyxin B/administration & dosage , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Animals , Catalase/metabolism , Catalase/chemistry , Wound Healing/drug effects , Mice , Escherichia coli/drug effects , Diabetes Mellitus, Experimental/drug therapy , Rats , RAW 264.7 Cells , Microbial Sensitivity Tests , Particle Size
13.
Braz J Microbiol ; 55(2): 1415-1425, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38619733

ABSTRACT

Pseudomonas aeruginosa is the main pathogen associated with pulmonary exacerbation in patients with cystic fibrosis (CF). CF is a multisystemic genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene, which mainly affects pulmonary function. P. aeruginosa isolated from individuals with CF in Brazil is not commonly associated with multidrug resistance (MDR), especially when compared to global occurrence, where the presence of epidemic clones, capable of expressing resistance to several drugs, is often reported. Due to the recent observations of MDR isolates of P. aeruginosa in our centers, combined with these characteristics, whole-genome sequencing was employed for analyses related to antimicrobial resistance, plasmid identification, search for phages, and characterization of CF clones. All isolates in this study were polymyxin B resistant, exhibiting diverse mutations and reduced susceptibility to carbapenems. Alterations in mexZ can result in the overexpression of the MexXY efflux pump. Mutations in oprD, pmrB, parS, gyrA and parC may confer reduced susceptibility to antimicrobials by affecting permeability, as observed in phenotypic tests. The phage findings led to the assumption of horizontal genetic transfer, implicating dissemination between P. aeruginosa isolates. New sequence types were described, and none of the isolates showed an association with epidemic CF clones. Analysis of the genetic context of P. aeruginosa resistance to polymyxin B allowed us to understand the different mechanisms of resistance to antimicrobials, in addition to subsidizing the understanding of possible relationships with epidemic strains that circulate among individuals with CF observed in other countries.


Subject(s)
Anti-Bacterial Agents , Cystic Fibrosis , Microbial Sensitivity Tests , Polymyxin B , Pseudomonas Infections , Pseudomonas aeruginosa , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Humans , Polymyxin B/pharmacology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/virology , Pseudomonas Infections/microbiology , Anti-Bacterial Agents/pharmacology , Mutation , Drug Resistance, Bacterial/genetics , Brazil , Bacterial Proteins/genetics , Whole Genome Sequencing , Drug Resistance, Multiple, Bacterial/genetics
14.
J Immunol ; 212(11): 1807-1818, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38639584

ABSTRACT

Drug-induced acute renal failure (ARF) is a public health concern that hinders optimal drug therapy. However, pathological mechanisms of drug-induced ARF remain to be elucidated. Here, we show that a pathological process of drug-induced ARF is mediated by proinflammatory cross-talk between kidney tubular cells and macrophages. Both polymyxin B and colistin, polypeptide antibiotics, frequently cause ARF, stimulated the ERK and NF-κB pathways in kidney tubular cells, and thereby upregulated M-CSF and MCP-1, leading to infiltration of macrophages into the kidneys. Thereafter, the kidney-infiltrated macrophages were exposed to polypeptide antibiotics, which initiated activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. Interestingly, blockade of the NLRP3 activation clearly ameliorated the pathology of ARF induced by polypeptide antibiotics, suggesting that a combination of the distinct cellular responses to polypeptide antibiotics in kidney tubular cells and macrophages plays a key role in the pathogenesis of colistin-induced ARF. Thus, our results provide a concrete example of how drugs initiate ARF, which may give insight into the underlying pathological process of drug-induced ARF.


Subject(s)
Acute Kidney Injury , Anti-Bacterial Agents , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Mice , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Polymyxin B/pharmacology , Mice, Inbred C57BL , Colistin/adverse effects , Colistin/pharmacology , Peptides/pharmacology , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Male , NF-kappa B/metabolism
15.
Arch Microbiol ; 206(4): 191, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38520490

ABSTRACT

Escherichia coli are generally resistant to the lantibiotic's action (nisin and warnerin), but we have shown increased sensitivity of E. coli to lantibiotics in the presence of subinhibitory concentrations of polymyxins. Synergistic lantibiotic-polymyxin combinations were found for polymyxins B and M. The killing of cells at the planktonic and biofilm levels was observed for two collection and four clinical multidrug-resistant E. coli strains after treatment with lantibiotic-polymyxin B combinations. Thus, 24-h treatment of E. coli mature biofilms with warnerin-polymyxin B or nisin-polymyxin B leads to five to tenfold decrease in the number of viable cells, depending on the strain. AFM revealed that the warnerin and polymyxin B combination caused the loss of the structural integrity of biofilm and the destruction of cells within the biofilm. It has been shown that pretreatment of cells with polymyxin B leads to an increase of Ca2+ and Mg2+ ions in the culture medium, as detected by atomic absorption spectroscopy. The subsequent exposure to warnerin caused cell death with the loss of K+ ions and cell destruction with DNA and protein release. Thus, polymyxins display synergy with lantibiotics against planktonic and biofilm cells of E. coli, and can be used to overcome the resistance of Gram-negative bacteria to lantibiotics.


Subject(s)
Bacteriocins , Nisin , Polymyxins/pharmacology , Polymyxin B/pharmacology , Anti-Bacterial Agents/pharmacology , Nisin/pharmacology , Escherichia coli/genetics , Plankton , Bacteriocins/pharmacology , Biofilms , Ions , Microbial Sensitivity Tests
16.
J Glob Antimicrob Resist ; 37: 108-121, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38552872

ABSTRACT

OBJECTIVES: A concern with the ESKAPE pathogen, Enterobacter bugandensis, and other species of the Enterobacter cloacae complex, is the frequent appearance of multidrug resistance against last-resort antibiotics, such as polymyxins. METHODS: Here, we investigated the responses to polymyxin B (PMB) in two PMB-resistant E. bugandensis clinical isolates by global transcriptomics and deletion mutagenesis. RESULTS: In both isolates, the genes of the CrrAB-regulated operon, including crrC and kexD, displayed the highest levels of upregulation in response to PMB. ∆crrC and ∆kexD mutants became highly susceptible to PMB and lost the heteroresistant phenotype. Conversely, heterologous expression of CrrC and KexD proteins increased PMB resistance in a sensitive Enterobacter ludwigii clinical isolate and in the Escherichia coli K12 strain, W3110. The efflux pump, AcrABTolC, and the two component regulators, PhoPQ and CrrAB, also contributed to PMB resistance and heteroresistance. Additionally, the lipid A modification with 4-L-aminoarabinose (L-Ara4N), mediated by the arnBCADTEF operon, was critical to determine PMB resistance. Biochemical experiments, supported by mass spectrometry and structural modelling, indicated that CrrC is an inner membrane protein that interacts with the membrane domain of the KexD pump. Similar interactions were modeled for AcrB and AcrD efflux pumps. CONCLUSION: Our results support a model where drug efflux potentiated by CrrC interaction with membrane domains of major efflux pumps combined with resistance to PMB entry by the L-Ara4N lipid A modification, under the control of PhoPQ and CrrAB, confers the bacterium high-level resistance and heteroresistance to PMB.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterobacter , Lipid A , Microbial Sensitivity Tests , Polymyxin B , Polymyxin B/pharmacology , Enterobacter/genetics , Enterobacter/drug effects , Enterobacter/metabolism , Anti-Bacterial Agents/pharmacology , Lipid A/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Arabinose/metabolism , Arabinose/pharmacology , Arabinose/analogs & derivatives , Humans , Gene Expression Regulation, Bacterial , Operon , Drug Resistance, Multiple, Bacterial/genetics , Enterobacteriaceae Infections/microbiology , Drug Resistance, Bacterial , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism
17.
Cornea ; 43(8): 1044-1048, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38537125

ABSTRACT

PURPOSE: Antimicrobial resistance is a global health threat, compounded by the reduction in the discovery of new antibiotics. A repurposed drugs-based approach could provide a viable alternative for the treatment of multidrug-resistant (MDR) bacterial infections. In this study, we sought to evaluate the in vitro efficacy of a novel drug combination, polymyxin B/trimethoprim (PT) + rifampin on MDR isolates from patients with bacterial keratitis in India. METHODS: Forty-three isolates, which included 20 Staphylococcus aureus , 19 Pseudomonas aeruginosa , 3 Pseudomonas stutzeri , and 1 Acinetobacter baumannii , were evaluated for their antibiotic resistance by minimum inhibitory concentration (MIC). Fractional Inhibitory Concentration Index (FICI) testing was performed to measure the antimicrobial impact of PT + rifampin in combination. RESULTS: Among S. aureus isolates, 100% were resistant to at least 1 antibiotic class, 12 (60%) were MDR, and 14 (70%) were classified as methicillin-resistant. Among the gram-negative isolates, >90% were classified as MDR. Fractional Inhibitory Concentration (FIC) testing revealed that PT + rifampin was effective in completely inhibiting growth of all isolates while also displaying additive or synergistic activity in approximately 70% of the strains. Mean FICI values were 0.753 ± 0.311 and 0.791 ± 0.369 for S. aureus and gram-negative isolates, respectively, and a >2-fold reduction in MIC was measured for both PT and rifampin when tested in combination versus alone. CONCLUSIONS: Our data demonstrate the ability of PT + rifampin to eliminate all isolates tested, even those conferring MDR, highlighting the promise of this drug combination for the treatment of bacterial keratitis.


Subject(s)
Anti-Bacterial Agents , Drug Combinations , Drug Resistance, Multiple, Bacterial , Eye Infections, Bacterial , Microbial Sensitivity Tests , Polymyxin B , Rifampin , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/drug therapy , Rifampin/pharmacology , Rifampin/therapeutic use , Polymyxin B/pharmacology , Trimethoprim/pharmacology , Trimethoprim/therapeutic use , Drug Therapy, Combination
18.
Langmuir ; 40(13): 6847-6861, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38501650

ABSTRACT

The use of an exogenous pulmonary surfactant (EPS) to deliver other relevant drugs to the lungs is a promising strategy for combined therapy. We evaluated the interaction of polymyxin B (PxB) with a clinically used EPS, the poractant alfa Curosurf (PSUR). The effect of PxB on the protein-free model system (MS) composed of four phospholipids (diC16:0PC/16:0-18:1PC/16:0-18:2PC/16:0-18:1PG) was examined in parallel to distinguish the specificity of the composition of PSUR. We used several experimental techniques (differential scanning calorimetry, small- and wide-angle X-ray scattering, small-angle neutron scattering, fluorescence spectroscopy, and electrophoretic light scattering) to characterize the binding of PxB to both EPS. Electrostatic interactions PxB-EPS are dominant. The results obtained support the concept of cationic PxB molecules lying on the surface of the PSUR bilayer, strengthening the multilamellar structure of PSUR as derived from SAXS and SANS. A protein-free MS mimics a natural EPS well but was found to be less resistant to penetration of PxB into the lipid bilayer. PxB does not affect the gel-to-fluid phase transition temperature, Tm, of PSUR, while Tm increased by ∼+ 2 °C in MS. The decrease of the thickness of the lipid bilayer (dL) of PSUR upon PxB binding is negligible. The hydrophobic tail of the PxB molecule does not penetrate the bilayer as derived from SANS data analysis and changes in lateral pressure monitored by excimer fluorescence at two depths of the hydrophobic region of the bilayer. Changes in dL of protein-free MS show a biphasic dependence on the adsorbed amount of PxB with a minimum close to the point of electroneutrality of the mixture. Our results do not discourage the concept of a combined treatment with PxB-enriched Curosurf. However, the amount of PxB must be carefully assessed (less than 5 wt % relative to the mass of the surfactant) to avoid inversion of the surface charge of the membrane.


Subject(s)
Polymyxin B , Pulmonary Surfactants , Polymyxin B/pharmacology , Polymyxin B/chemistry , Scattering, Small Angle , Lipid Bilayers , X-Ray Diffraction , Surface-Active Agents , Thermodynamics , Lung/metabolism
19.
NPJ Biofilms Microbiomes ; 10(1): 16, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429317

ABSTRACT

Pseudomonas aeruginosa is an important cause of lower respiratory tract infections, such as ventilator-associated bacterial pneumonia (VABP). Using inhaled antibiotics to treat VABP can achieve high drug concentrations at the infection site while minimizing systemic toxicities. Despite the theoretical advantages, clinical trials have failed to show a benefit for inhaled antibiotic therapy in treating VABP. A potential reason for this discordance is the presence of biofilm-embedded bacteria in lower respiratory tract infections. Drug selection and dosing are often based on data from bacteria grown planktonically. In the present study, an in vitro air-liquid interface pharmacokinetic/pharmacodynamic biofilm model was optimized to evaluate the activity of simulated epithelial lining fluid exposures of inhaled and intravenous doses of polymyxin B and tobramycin against two P. aeruginosa strains. Antibiotic activity was also determined against the P. aeruginosa strains grown planktonically. Our study revealed that inhaled antibiotic exposures were more active than their intravenous counterparts across biofilm and planktonic populations. Inhaled exposures of polymyxin B and tobramycin exhibited comparable activity against planktonic P. aeruginosa. Although inhaled polymyxin B exposures were initially more active against P. aeruginosa biofilms (through 6 h), tobramycin was more active by the end of the experiment (48 h). Together, these data slightly favor the use of inhaled tobramycin for VABP caused by biofilm-forming P. aeruginosa that are not resistant to either antibiotic. The optimized in vitro air-liquid interface pharmacokinetic/pharmacodynamic biofilm model may be beneficial for the development of novel anti-biofilm agents or to optimize antibiotic dosing for infections such as VABP.


Subject(s)
Pseudomonas Infections , Respiratory Tract Infections , Humans , Anti-Bacterial Agents , Pseudomonas aeruginosa , Polymyxin B/pharmacology , Tobramycin/pharmacology , Pseudomonas Infections/drug therapy , Biofilms
20.
Int J Pharm ; 654: 123947, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38408553

ABSTRACT

Photodynamic Therapy is a therapy based on combining a non-toxic compound, known as photosensitizer (PS), and irradiation with light of the appropriate wavelength to excite the PS molecule. The photon absorption by the PS leads to reactive oxygen species generation and a subsequent oxidative burst that causes cell damage and death. In this work, we report an antimicrobial nanodevice that uses the activity of curcumin (Cur) as a PS for antimicrobial Photodynamic Therapy (aPDT), based on mesoporous silica nanoparticles in which the action of the classical antibiotic PMB is synergistically combined with the aPDT properties of curcumin to combat bacteria. The synergistic effect of the designed gated device in combination with irradiation with blue LED light (470 nm) is evaluated against Escherichia coli, Pseudomonas aeruginosa and Staphylococcus epidermidis. The results show that the nanodevice exhibits a noteworthy antibacterial activity against these microorganisms, a much more significant effect than free Cur and PMB at equivalent concentrations. Thus, 0.1 µg/mL of MSNs-Cur-PMB eliminates a bacterial concentration of about 105 CFU/mL of E. coli, while 1 µg/mL of MSNs-Cur-PMB is required for P. aeruginosa and S. epidermidis. In addition, antibiofilm activity against the selected bacteria was also tested. We found that 0.1 mg/mL of MSNs-Cur-PMB inhibited 99 % biofilm formation for E. coli, and 1 mg/mL of MSNs-Cur-PMB achieved 90 % and 100 % inhibition of biofilm formation for S. epidermidis and P. aeruginosa, respectively.


Subject(s)
Curcumin , Nanoparticles , Photochemotherapy , Polymyxin B/pharmacology , Curcumin/pharmacology , Silicon Dioxide/pharmacology , Escherichia coli , Biofilms , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa
SELECTION OF CITATIONS
SEARCH DETAIL
...