Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
1.
Appl Microbiol Biotechnol ; 108(1): 17, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38170316

RESUMO

Polymyxins are cationic peptide antibiotics and regarded as the "final line of defense" against multidrug-resistant bacterial infections. Meanwhile, some polymyxin-resistant strains and the corresponding resistance mechanisms have also been reported. However, the response of the polymyxin-producing strain Paenibacillus polymyxa to polymyxin stress remains unclear. The purpose of this study was to investigate the stress response of gram-positive P. polymyxa SC2 to polymyxin B and to identify functional genes involved in the stress response process. Polymyxin B treatment upregulated the expression of genes related to basal metabolism, transcriptional regulation, transport, and flagella formation and increased intracellular ROS levels, flagellar motility, and biofilm formation in P. polymyxa SC2. Adding magnesium, calcium, and iron alleviated the stress of polymyxin B on P. polymyxa SC2, furthermore, magnesium and calcium could improve the resistance of P. polymyxa SC2 to polymyxin B by promoting biofilm formation. Meanwhile, functional identification of differentially expressed genes indicated that an ABC superfamily transporter YwjA was involved in the stress response to polymyxin B of P. polymyxa SC2. This study provides an important reference for improving the resistance of P. polymyxa to polymyxins and increasing the yield of polymyxins. KEY POINTS: • Phenotypic responses of P. polymyxa to polymyxin B was performed and indicated by RNA-seq • Forming biofilm was a key strategy of P. polymyxa to alleviate polymyxin stress • ABC transporter YwjA was involved in the stress resistance of P. polymyxa to polymyxin B.


Assuntos
Paenibacillus polymyxa , Paenibacillus , Paenibacillus polymyxa/genética , Polimixina B/farmacologia , Polimixina B/metabolismo , Paenibacillus/genética , Paenibacillus/metabolismo , Cálcio/metabolismo , Magnésio , Polimixinas/farmacologia
2.
J Biochem Mol Toxicol ; 37(6): e23344, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36992629

RESUMO

Polymyxin B (PMB) is a polypeptide antibiotic widely used in treating multidrug-resistant Gram-negative bacteria. However, nephrotoxicity is a serious adverse effect that limits its clinical use. Therefore, clarification of the molecular mechanism of PMB-induced renal injury is essential. Our study aimed to explore possible mechanisms of PMB-induced nephrotoxicity in vivo and in vitro. Mice were treated with PMB to construct the kidney injury model. The antioxidant capacity was assessed by measuring the superoxide dismutase (SOD) and catalase (CAT) activities and the glutathione (GSH) and malondialdehyde (MDA) contents. The pathway of the nuclear factor erythroid 2-related factor 2/NADH quinone oxidoreductase 1 (Nrf2/NQO1) was examined after PMB treatment in NRK-52E cells and mice. Finally, the expressions of genes and proteins (Bax, Bcl-2, Caspase-3, Caspase-9) related to apoptosis were evaluated through quantitative polymerase chain reaction and western blot assay. The study verified PMB-induced nephrotoxicity in mice and NRK-52E cells in a dose- and time-dependent manner. PMB treatment significantly decreased the expression of Nrf2 and its downstream target gene NQO1 and increased the apoptosis-related proteins expression. In summary, our results suggested that PMB-induced oxidative stress damage by inhibiting the Nrf2/NQO1 pathway and promoting apoptosis in kidney tissues.


Assuntos
Antioxidantes , Polimixina B , Camundongos , Animais , Antioxidantes/farmacologia , Polimixina B/metabolismo , Polimixina B/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Glutationa/metabolismo , Apoptose
3.
mSphere ; 8(1): e0053722, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36622250

RESUMO

Outer membrane vesicles (OMVs) secreted by Gram-negative bacteria serve as transporters for the delivery of cargo such as virulence and antibiotic resistance factors. OMVs play a key role in the defense against membrane-targeting antibiotics such as the polymyxin B. Herein, we conducted comparative proteomics of OMVs from paired Klebsiella pneumoniae ATCC 700721 polymyxin-susceptible (polymyxin B MIC = 0.5 mg/L) and an extremely resistant (polymyxin B MIC ≥128 mg/L), following exposure to 2 mg/L of polymyxin B. Comparative profiling of the OMV subproteome of each strain revealed proteins from multiple perturbed pathways, particularly in the polymyxin-susceptible strain, including outer membrane assembly (lipopolysaccharide, O-antigen, and peptidoglycan biosynthesis), cationic antimicrobial peptide resistance, ß-lactam resistance, and quorum sensing. In the polymyxin-susceptible strain, polymyxin B treatment reduced the expression of OMV proteins in the pathways related to adhesion, virulence, and the cell envelope stress responses, whereas, in the polymyxin-resistant strain, the proteins involved in LPS biosynthesis, RNA degradation, and nucleotide excision repair were significantly overexpressed in response to polymyxin B treatment. Intriguingly, the key polymyxin resistance enzymes 4-amino-4-deoxy-l-arabinose transferase and the PhoPQ two-component protein kinase were significantly downregulated in the OMVs of the polymyxin-susceptible strain. Additionally, a significant reduction in class A ß-lactamase proteins was observed following polymyxin B treatment in the OMVs of both strains, particularly the OMVs of the polymyxin-susceptible strain. These findings shed new light on the OMV subproteome of extremely polymyxin resistant K. pneumoniae, which putatively may serve as active decoys to make the outer membrane more impervious to polymyxin attack. IMPORTANCE OMVs can help bacteria to fight antibiotics not only by spreading antibiotic resistance genes but also by acting as protective armor against antibiotics. By employing proteomics, we found that OMVs have a potential role in shielding K. pneumoniae and acting as decoys to polymyxin attack, through declining the export of proteins (e.g., 4-amino-4-deoxy-l-arabinose transferase) involved in polymyxin resistance. Furthermore, polymyxin B treatment of both strains leads to shedding of the OMVs with perturbed proteins involved in outer membrane remodeling (e.g., LPS biosynthesis) as well as pathogenic potential of K. pneumoniae (e.g., quorum sensing). The problematic extended spectrum beta-lactamases SHV and TEM were significantly reduced in both strains, suggesting that polymyxin B may act as a potentiator to sensitize the bacterium to ß-lactam antibiotics. This study highlights the importance of OMVs as "molecular mules" for the intercellular transmission and delivery of resistance and cellular repair factors in the bacterial response to polymyxins.


Assuntos
Polimixina B , Polimixinas , Polimixina B/farmacologia , Polimixina B/metabolismo , Polimixinas/farmacologia , Klebsiella pneumoniae/genética , Preparações Farmacêuticas , Lipopolissacarídeos/metabolismo , Proteômica , Antibacterianos/farmacologia , Antibacterianos/metabolismo
4.
Res Microbiol ; 174(1-2): 103993, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36184018

RESUMO

Aspergillus fumigatus and Pseudomonas aeruginosa biofilms are associated to the recalcitrant and persistent infections due to resistance to antimicrobials. Here, we evaluated the effect of antimicrobials on single and mixed biofilms of A. fumigatus and P. aeruginosa (carbapenem-resistant and susceptible strains) determining total biomass by crystal violet, cell viability by colony forming unit count, and microscopy. Polymyxin B (PMB) had the best action on P. aeruginosa biofilms inhibiting the biomass (2-4 µg/mL) and it was efficient reducing the viable bacterial cells. Amphotericin B (AMB) and caspofungin (CAS) were the best antifungal at inhibiting A. fumigatus biofilms and reducing fungal viability at concentration ≥1 and ≥ 16 µg/mL, respectively. In addition, CAS was able to significantly reduce P. aeruginosa viability in mixed biofilms. CAS combined with PMB also significantly reduced the mixed biofilm biomass and fungal and bacterial viability mainly against carbapenem-resistant bacterium. The light and fluorescence microscopy showed alterations on hyphae morphology and confirmed the increase of fungal and bacterial death cells after combined therapy of mixed biofilms. Taken together, our work showed that CAS alone and its combination with PMB showed better potential in reducing mixed biofilm biomass and fungal and bacterial viability, even for the carbapenem-resistant P. aeruginosa strain.


Assuntos
Anti-Infecciosos , Polimixina B , Caspofungina/farmacologia , Caspofungina/metabolismo , Polimixina B/farmacologia , Polimixina B/metabolismo , Aspergillus fumigatus , Pseudomonas aeruginosa , Anti-Infecciosos/farmacologia , Biofilmes , Carbapenêmicos/farmacologia , Carbapenêmicos/metabolismo , Testes de Sensibilidade Microbiana
5.
J Biomed Sci ; 29(1): 89, 2022 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-36310165

RESUMO

BACKGROUND: Understanding the mechanism of antimicrobial action is critical for improving antibiotic therapy. For the first time, we integrated correlative metabolomics and transcriptomics of Pseudomonas aeruginosa to elucidate the mechanism of synergistic killing of polymyxin-rifampicin combination. METHODS: Liquid chromatography-mass spectrometry and RNA-seq analyses were conducted to identify the significant changes in the metabolome and transcriptome of P. aeruginosa PAO1 after exposure to polymyxin B (1 mg/L) and rifampicin (2 mg/L) alone, or in combination over 24 h. A genome-scale metabolic network was employed for integrative analysis. RESULTS: In the first 4-h treatment, polymyxin B monotherapy induced significant lipid perturbations, predominantly to fatty acids and glycerophospholipids, indicating a substantial disorganization of the bacterial outer membrane. Expression of ParRS, a two-component regulatory system involved in polymyxin resistance, was increased by polymyxin B alone. Rifampicin alone caused marginal metabolic perturbations but significantly affected gene expression at 24 h. The combination decreased the gene expression of quorum sensing regulated virulence factors at 1 h (e.g. key genes involved in phenazine biosynthesis, secretion system and biofilm formation); and increased the expression of peptidoglycan biosynthesis genes at 4 h. Notably, the combination caused substantial accumulation of nucleotides and amino acids that last at least 4 h, indicating that bacterial cells were in a state of metabolic arrest. CONCLUSION: This study underscores the substantial potential of integrative systems pharmacology to determine mechanisms of synergistic bacterial killing by antibiotic combinations, which will help optimize their use in patients.


Assuntos
Polimixina B , Pseudomonas aeruginosa , Humanos , Pseudomonas aeruginosa/genética , Polimixina B/farmacologia , Polimixina B/metabolismo , Rifampina/farmacologia , Rifampina/metabolismo , Transcriptoma , Polimixinas/farmacologia , Polimixinas/metabolismo , Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana
6.
Biosensors (Basel) ; 12(9)2022 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-36140103

RESUMO

An in vitro human renal proximal tubule model that represents the proper transporter expression and pronounced epithelial polarization is necessary for the accurate prediction of nephrotoxicity. Here, we constructed a high-throughput human renal proximal tubule model based on an integrated biomimetic array chip (iBAC). Primary human renal proximal tubule epithelial cells (hRPTECs) cultured on this microfluidic platform were able to form a tighter barrier, better transporter function and more sensitive nephrotoxicity prediction than those on the static Transwell. Compared with the human immortalized HK2 model, the hRPTECs model on the chip gained improved apical-basolateral polarization, barrier function and transporter expression. Polymyxin B could induce nephrotoxicity not only from the apical of the hRPTECs, but also from the basolateral side on the iBAC. However, other chemotherapeutic agents, such as doxorubicin and sunitinib, only induced nephrotoxicity from the apical surface of the hRPTECs on the iBAC. In summary, our renal proximal tubule model on the chip exhibits improved epithelial polarization and membrane transporter activity, and can be implemented as an effective nephrotoxicity-screening toolkit.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Dispositivos Lab-On-A-Chip , Doxorrubicina , Células Epiteliais/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/farmacologia , Polimixina B/metabolismo , Polimixina B/farmacologia , Sunitinibe/metabolismo , Sunitinibe/farmacologia
7.
Front Immunol ; 13: 927017, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36159837

RESUMO

The exploration of therapies combining antimicrobial lung proteins and conventional antibiotics is important due to the growing problem of multidrug-resistant bacteria. The aim of this study was to investigate whether human SP-A and a recombinant trimeric fragment (rfhSP-A) have cooperative antimicrobial activity with antibiotics against pathogenic Gram-negative bacteria. We found that SP-A bound the cationic peptide polymyxin B (PMB) with an apparent dissociation constant (K D) of 0.32 ± 0.04 µM. SP-A showed synergistic microbicidal activity with polymyxin B and E, but not with other antibiotics, against three SP-A-resistant pathogenic bacteria: Klebsiella pneumoniae, non-typable Haemophilus influenzae (NTHi), and Pseudomonas aeruginosa. SP-A was not able to bind to K. pneumoniae, NTHi, or to mutant strains thereof expressing long-chain lipopolysaccharides (or lipooligosaccharides) and/or polysaccharide capsules. In the presence of PMB, SP-A induced the formation of SP-A/PMB aggregates that enhance PMB-induced bacterial membrane permeabilization. Furthermore, SP-A bound to a molecular derivative of PMB lacking the acyl chain (PMBN) with a K D of 0.26 ± 0.02 µM, forming SP-A/PMBN aggregates. PMBN has no bactericidal activity but can bind to the outer membrane of Gram-negative bacteria. Surprisingly, SP-A and PMBN showed synergistic bactericidal activity against Gram-negative bacteria. Unlike native supratrimeric SP-A, the trimeric rfhSP-A fragment had small but significant direct bactericidal activity against K. pneumoniae, NTHi, and P. aeruginosa. rfhSP-A did not bind to PMB under physiological conditions but acted additively with PMB and other antibiotics against these pathogenic bacteria. In summary, our results significantly improve our understanding of the antimicrobial actions of SP-A and its synergistic action with PMB. A peptide based on SP-A may aid the therapeutic use of PMB, a relatively cytotoxic antibiotic that is currently being reintroduced into clinics due to the global problem of antibiotic resistance.


Assuntos
Polimixina B , Polimixinas , Antibacterianos/química , Antibacterianos/farmacologia , Antibióticos Antineoplásicos , Bactérias , Bactérias Gram-Negativas/metabolismo , Humanos , Klebsiella pneumoniae , Polimixina B/metabolismo , Polimixina B/farmacologia , Polimixinas/química , Polimixinas/metabolismo , Polimixinas/farmacologia , Pseudomonas aeruginosa , Proteína A Associada a Surfactante Pulmonar
8.
Microbiol Spectr ; 10(5): e0135622, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-35972274

RESUMO

Bacteria frequently encounter selection by both phages and antibiotics. However, our knowledge on the evolutionary interactions between phages and antibiotics are still limited. Here, we characterized a phage-resistant Pseudomonas aeruginosa variant PAO1-R1 that shows increased sensitivity to gentamicin and polymyxin B. Using whole genome sequencing, significant genome differences were observed between the reference P. aeruginosa PAO1 and PAO1-R1. Compared to PAO1, 64 gene-encoding proteins with nonsynonymous single nucleotide polymorphisms (SNPs) and 31 genes with insertion/deletion (indel) mutations were found in PAO1-R1. We observed a significant reduction in phage adsorption rate for both phage vB_Pae_QDWS and vB_Pae_W3 against PAO1-R1 and proposed that disruption of phage adsorption is likely the main cause for evolving resistance. Because the majority of spontaneous mutations are closely related to membrane components, alterations in the cell envelope may explain the antibiotic-sensitive phenotype of PAO1-R1. Collectively, we demonstrate that the evolution of phage resistance comes with fitness defects resulting in antibiotic sensitization. Our finding provides new insights into the evolutionary interactions between resistance to the phage and sensitivity to antibiotics, which may have implications for the future clinical use of steering in phage therapies. IMPORTANCE Bacteria frequently encounter the selection pressure from both antibiotics and lytic phages. Little is known about the evolutionary interactions between antibiotics and phages. Our study provides new insights into the trade-off mechanism between resistance to the phage and sensitivity to antibiotics. This evolutionary trade-off is not dependent on the outer membrane proteins (OMPs) of the multidrug efflux pumps. The disruption of phage adsorption that induced phage resistance and the changes in structure or composition of membranes are presumably one of the major causes for antibiotic sensitivity. Our finding may fill some gaps in the field of phage-host interplay and have implications for the future clinical use of steering in phage therapies.


Assuntos
Bacteriófagos , Fagos de Pseudomonas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Fagos de Pseudomonas/genética , Antibacterianos/farmacologia , Polimixina B/farmacologia , Polimixina B/metabolismo , Gentamicinas/metabolismo
9.
Biocontrol Sci ; 27(2): 57-64, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35753794

RESUMO

Vibrio vulnificus, an opportunistic human pathogen responsible for primary septicemia, initiates pathogenesis by attachment to the intestinal epithelial cells for which the motility by the polar flagellum plays an essential role. The proteomic analysis of outer membrane proteins showed that the treatment with the 1/2 minimum inhibitory concentration (MIC) of polymyxin B (a bacterial antimicrobial peptide) led to the reduced production of flagellin (a major component of the polar flagellum). Furthermore, the bacterial motility was inhibited in the presence of 1/2 MIC of polymyxin B. V. vulnificus has six flagellin genes organized into the flaFBA and flaCDE loci. The flaA was found to be expressed higher than flaC, and its expression was significantly decreased by polymyxin B. As well as polymyxin B, the 1/2 MIC of LL-37 (a human intestinal antimicrobial peptide) reduced the expression of flaA. In addition, among four fragments of LL-37, KI-18 and FK-13 containing F17KRIVQRIKDELR29 could lead to the decreased expression of flaA. Because the motility closely relates to virulence of V. vulnificus, the findings obtained herein indicate that LL-37 may reduce the bacterial virulence through inhibition of the motility via the polar flagellum.


Assuntos
Vibrio vulnificus , Flagelos/genética , Flagelos/metabolismo , Flagelina/genética , Flagelina/metabolismo , Flagelina/farmacologia , Humanos , Polimixina B/metabolismo , Polimixina B/farmacologia , Proteômica , Vibrio vulnificus/genética
10.
FEBS J ; 289(12): 3550-3567, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35038363

RESUMO

The outer membrane (OM) is an essential component of the Gram-negative bacterial cell envelope. Restricted diffusion of integral OM proteins and lipopolysaccharide (LPS) that constitute the outer leaflet of the OM support a model in which the OM is in a semi-crystalline state. The low fluidity of the OM has been suggested to be an important property of this membrane that even contributes to cell rigidity. The LPS characteristics strongly determine the properties of the OM and the LPS layer fluidity has been measured using different techniques that require specific conditions or are technically challenging. Here, we characterize the Escherichia coli LPS fluidity by evaluating the lateral diffusion of the styryl dye FM4-64FX in fluorescence recovery after photobleaching experiments. This technique allowed us to determine the effect of different conditions and genetic backgrounds on the LPS fluidity. Our results show that a fraction of the LPS can slowly diffuse and that the fluidity of the LPS layer adapts by modifying the diffusion of the LPS and the fraction of mobile LPS molecules.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Cátions Bivalentes/metabolismo , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Lipopolissacarídeos/metabolismo , Fluidez de Membrana , Polimixina B/análise , Polimixina B/metabolismo , Temperatura
11.
Environ Microbiol ; 24(3): 1133-1149, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34490971

RESUMO

Antimicrobials are commonly used in prevention of infections including in aquaculture, agriculture and medicine. Subinhibitory concentrations of antimicrobial peptides can modulate resistance, virulence and persistence effectors in Gram-negative pathogens. In this study, we investigated the effect of subinhibitory concentrations of polymyxin B (PmB) on the secretome of Vibrio cholerae, a natural inhabitant of aquatic environments and the pathogen responsible for the cholera disease. Our proteomic approach revealed that the abundance of many extracellular proteins is affected by PmB and some of them are detected only either in the presence or in the absence of PmB. The type VI secretion system (T6SS) secreted hemolysin-coregulated protein (Hcp) displayed an increased abundance in the presence of PmB. Hcp is also more abundant in the bacterial cells in the presence of PmB and hcp expression is upregulated upon PmB supplementation. No effect of the T6SS on antimicrobial resistance was observed. Conversely, PmB increases the T6SS-dependent cytotoxicity of V. cholerae towards the amoeba Dictyostelium discoideum and its ability to compete with Escherichia coli.


Assuntos
Dictyostelium , Sistemas de Secreção Tipo VI , Vibrio cholerae , Proteínas de Bactérias/metabolismo , Escherichia coli/metabolismo , Polimixina B/metabolismo , Polimixina B/farmacologia , Proteômica , Secretoma , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/metabolismo , Vibrio cholerae/metabolismo
12.
Biomolecules ; 11(2)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562877

RESUMO

BACKGROUND: Infectious diseases are one of the main causes of morbidity and mortality worldwide. Nuclear molecular imaging would be of great help to non-invasively discriminate between septic and sterile inflammation through available radiopharmaceuticals, as none is currently available for clinical practice. Here, we describe the radiolabeling procedure and in vitro and in vivo studies of 99mTc-polymyxin B sulfate (PMB) as a new single photon emission imaging agent for the characterization of infections due to Gram-negative bacteria. RESULTS: Labeling efficiency was 97 ± 2% with an average molar activity of 29.5 ± 0.6 MBq/nmol. The product was highly stable in saline and serum up to 6 h. In vitro binding assay showed significant displaceable binding to Gram-negative bacteria but not to Gram-positive controls. In mice, 99mTc-HYNIC-PMB was mainly taken up by liver and kidneys. Targeting studies confirmed the specificity of 99mTc-HYNIC-PMB obtained in vitro, showing significantly higher T/B ratios for Gram-negative bacteria than Gram-positive controls. CONCLUSIONS: In vitro and in vivo results suggest that 99mTc-HYNIC-PMB has a potential for in vivo identification of Gram-negative bacteria in patients with infections of unknown etiology. However, further investigations are needed to deeply understand the mechanism of action and behavior of 99mTc-HYNIC-PMB in other animal models and in humans.


Assuntos
Infecções por Bactérias Gram-Negativas/diagnóstico por imagem , Marcação por Isótopo/métodos , Polimixina B/química , Compostos Radiofarmacêuticos/química , Tecnécio/química , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Acinetobacter baumannii/crescimento & desenvolvimento , Acinetobacter baumannii/metabolismo , Animais , Reagentes de Ligações Cruzadas/química , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/metabolismo , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Feminino , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Rim/diagnóstico por imagem , Rim/metabolismo , Rim/microbiologia , Klebsiella pneumoniae/crescimento & desenvolvimento , Klebsiella pneumoniae/metabolismo , Fígado/diagnóstico por imagem , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Polimixina B/metabolismo , Polimixina B/farmacocinética , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/metabolismo , Succinimidas/química , Tecnécio/metabolismo , Tecnécio/farmacocinética
13.
Microbiology (Reading) ; 166(12): 1149-1159, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33205745

RESUMO

Colonization of the gastrointestinal tract by enterohaemorrhagic Escherichia coli (EHEC) is critically dependent on its ability to sense and respond to various microenvironments within the host. EHEC exposure to physiologically relevant levels of bile salts upregulates the two-component system, pmrAB, and the arnBCADTEF operon, resulting in lipopolysaccharide modification and increased resistance to the cationic antimicrobial peptide, polymyxin B (PMB). A similar pmrAB- and arn-dependent PMB resistance has been observed in Salmonella enterica in the presence of ferric iron. Limiting magnesium levels and mild acid can also induce Salmonella resistance to PMB through another two-component system, PhoPQ and the connector protein, PmrD. This study aims to evaluate the relative contributions of a bile-salt mix (BSM), iron, limiting magnesium as well as the roles of pmrAB, phoPQ and pmrD to EHEC's resistance to PMB. Killing assays show that EHEC treatment with the BSM or iron under excess magnesium and neutral pH conditions induces a pmrAB-dependent, phoP-independent PMB resistance. By contrast, exposure to limiting magnesium triggers a pmrB-, phoP- and pmrD-dependent PMB resistance. The iron-induced PMB resistance is independent of phoP and pmrD under limiting magnesium conditions while the bile-salt-induced PMB resistance is independent of pmrD only under non-PhoP-inducing conditions. GFP-pmrD transcriptional reporter studies reveal that the limiting magnesium enhances pmrD expression, which is repressed upon additional exposure to either BSM or iron. Our results also show that exposure to mild acid enhances PMB resistance in a pmrD-independent manner and GFP reporter results confirm minimal expression of pmrD at this pH regardless of the magnesium level. This study provides novel insights into how EHEC differentially employs PmrAB, PhoPQ and PmrD to monitor and respond to bile salts, iron, acidic pH and magnesium typically encountered within the gastrointestinal tract in order to modulate its survival against cationic antimicrobial peptides.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Ácidos e Sais Biliares/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Escherichia coli Êntero-Hemorrágica/fisiologia , Ferro/farmacologia , Antibacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Farmacorresistência Bacteriana/genética , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Magnésio/metabolismo , Polimixina B/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Life Sci ; 259: 118250, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32791152

RESUMO

AIMS: Several microbial toll-like receptor (TLR) ligands, bacterial DNA and bacterial cell wall fragments have been identified in the synovium of rheumatoid arthritis (RA) patients, proving bacterial involvement in the pathogenesis of RA. The current study aimed to verify that low dose polymyxin B could prevent the development of chronic inflammatory arthritis. METHODS: Twelve days post adjuvant injection, Sprague-Dawley rats were treated twice weekly with methotrexate (0.5 mg/kg) or daily with polymyxin B (1 mg/kg) or with combination of both for 1 or 2 weeks. Arthritis progression was assessed by hind paw swelling, serum levels of tumor growth factor-1ß (TGF-1ß), tumor necrosis factor-alpha (TNF-α), high sensitivity C-reactive protein (HS-CRP) and nuclear factor kappa B (NF-κB) were measured using ELISA. Cyclooxygenase-1 (Cox-1) and Cox-2 activities, as well as mRNA expression of TLR-2 and TLR-4 were determined. Histopathological examination of the ankle joint was performed as well as immunohistochemistry for anti-TLR-4. Histopathological assessment of toxic effects on the kidney was performed. KEY FINDINGS: Adjuvant arthritis led to a significant swelling of the hind paw and alteration in all serum parameters, TLR-2 and TLR-4 expression, as well as Cox-2 activity. These alterations were associated with histopathological changes of the joints. Polymyxin B reduced significantly all biomarkers of inflammation, showing better effect of the combination in most of the studied parameters, with minimal signs of nephrotoxicity. SIGNIFICANCE: In conclusion, results showed that polymyxin B possesses significant anti-arthritic activity which may be attributed to inhibition of the TLR-4, NF-κB and Cox-2 signaling pathway.


Assuntos
Artrite Experimental/tratamento farmacológico , Polimixina B/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/tratamento farmacológico , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Ciclo-Oxigenase 2/fisiologia , Adjuvante de Freund/farmacologia , Inflamação/tratamento farmacológico , Masculino , NF-kappa B/metabolismo , Polimixina B/metabolismo , Polimixina B/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Membrana Sinovial/metabolismo , Receptores Toll-Like/metabolismo , Receptores Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Chemosphere ; 259: 127449, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32622246

RESUMO

Polymyxin B is increasingly employed all over the world to treat patients who affected by multidrug-resistant Gram-negative bacteria. Although the mechanism of resistance to polymyxin B is well known, the metabolic role of bacteria in stress response to polymyxin B remains an important task and may help to better understand polymyxin B-related stress response. In this study, the proteome changes of Escherichia coli (E. coli) continuously induced in concentrations of 1.0 mg/L and 10.0 mg/L polymyxin B were revealed. Compared to E. coli (PMB0), E. coli exposed to polymyxin B at 1.0 mg/L (PMB1) and 10.0 mg/L (PMB10) resulted in 89 and 314 differentially expressed proteins (DEPs), respectively. Such differences related to fatty acid degradation, quorum sensing and two-component regulatory system pathways. Based on absolute quantitative (iTRAQ) proteomics analysis, this study comprehensively studied the changes of E. coli proteome in culture with concentrations of 1.0 mg/L and 10.0 mg/L polymyxin B through confocal laser scanning microscopy observation, cell viability detection and reactive oxygen species analysis. The results showed that E. coli cultured at concentration of 10.0 mg/L polymyxin B increased the expression levels of multidrug-resistant efflux transporters and efflux pump membrane transporters, which might further improve the pathogens of polymyxin B-resistant bacteria lastingness and evolution. It has emerged globally to resist polymyxin B. The reuse of polymyxin B should be aroused public attention to avoid causing more serious environmental pollution. These findings could provide new insights into polymyxin B-related stress.


Assuntos
Antibacterianos/toxicidade , Escherichia coli/efeitos dos fármacos , Polimixina B/toxicidade , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Proteínas de Escherichia coli/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Polimixina B/metabolismo , Polimixinas/análogos & derivados , Polimixinas/metabolismo , Polimixinas/farmacologia , Proteoma/metabolismo , Proteômica
16.
Biochem Biophys Res Commun ; 528(1): 85-91, 2020 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-32451086

RESUMO

Pseudomonas aeruginosa is a widely found opportunistic pathogen. The emergence of multidrug-resistant strains and persistent chronic infections have increased. The protein encoded by the pa0423 gene in P. aeruginosa is proposed to be critical for pathogenesis and could be a virulence-promoting protease or a bacterial lipocalin that binds a lipid-like antibiotic for drug resistance. Although two functions of proteolysis and antibiotic resistance are mutually related to bacterial survival in the host, it is very unusual for a single-domain protein to target unrelated ligand molecules such as protein substrates and lipid-like antibiotics. To clearly address the biological role of the PA0423 protein, we performed structural and biochemical studies. We found that PA0423 adopts a single-domain ß-barrel structure and belongs to the lipocalin family. The PA0423 structure houses an internal tubular cavity, which accommodates a ubiquinone-8 molecule. Furthermore, we reveal that PA0423 can directly interact with the polymyxin B antibiotic using the internal cavity, suggesting that PA0423 has a physiological function in the antibiotic resistance of P. aeruginosa.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Lipocalinas/química , Modelos Moleculares , Polimixina B/química , Polimixina B/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidade , Homologia Estrutural de Proteína , Ubiquinona/química , Ubiquinona/metabolismo
17.
J Med Microbiol ; 69(1): 63-71, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31904320

RESUMO

Introduction. The worldwide emergence of carbapenem resistance in Gram-negative bacteria makes the development of simple tests mandatory to identify antimicrobial resistance mechanisms. Enzymatic and membrane barriers are the prominent resistance mechanisms described in these bacteria. Several tests are currently used to detect carbapenemase activities.Aim. However, a simple test for the identification of membrane-associated mechanisms of resistance is not yet available and this mechanism is often inferred after the exclusion of a carbapenemase in carbapenem-resistant Gram-negative bacteria.Methodology. Different media (liquid and solid) containing a membrane permeabilizer were tested to identify the existence of a membrane barrier. Here, polymyxin B nonapeptide (PMBN) was selected to bypass the role of impermeability in clinical carbapenem-resistant Enterobacteriaceae, including Escherichia coli, Enterobacter cloacae , Klebsiella pneumoniae and Klebsiella aerogenes isolates. In parallel, the expression of porins (OmpC and OmpF types) was checked in the various bacterial strains in order to search for a correlation between the restoration of susceptibility and the expression of porin.Results. Using a large number of clinical isolates, PMBN associated with a carbapenem allowed us to detect porin-deficient isolates with a sensitivity ranging from 89 to 93 % and a specificity ranging from 86 to 100 %.Conclusion. This paves the way for a diagnostic assay allowing the detection of this membrane-associated mechanism of resistance in Enterobacteriaceae.


Assuntos
Antibacterianos/metabolismo , Membrana Externa Bacteriana/fisiologia , Enterobacteriáceas Resistentes a Carbapenêmicos/efeitos dos fármacos , Permeabilidade , Polimixina B/metabolismo , Enterobacteriáceas Resistentes a Carbapenêmicos/genética , Porinas/genética , Porinas/metabolismo
18.
Am J Vet Res ; 80(12): 1099-1106, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31763943

RESUMO

OBJECTIVE: To determine whether therapeutic concentrations (> 0.5 to 1.0 µg/mL) of polymyxin B (PB) were achieved in the tarsocrural joint of horses when the drug was administered by IV regional limb perfusion (IV-RLP) via a saphenous vein at doses of 25, 50, and 300 mg and to describe any adverse systemic or local effects associated with such administration. ANIMALS: 9 healthy adult horses. PROCEDURES: In the first of 2 experiments, 6 horses each received 25 and 50 mg of PB by IV-RLP via a saphenous vein with at least 2 weeks between treatments. For each treatment, a tourniquet was placed at the midmetatarsus and another was placed midway between the stifle joint and tarsus. Both tourniquets were removed 30 minutes after the assigned dose was administered. Blood and tarsocrural joint fluid samples were collected for determination of PB concentration before and at predetermined times after drug administration. In experiment 2, 4 horses were administered 300 mg of PB by IV-RLP in 1 randomly selected pelvic limb in a manner identical to that used in experiment 1. RESULTS: For all 3 doses, the mean synovial fluid PB concentration was > 10 times the therapeutic concentration and below the level of quantification at 30 and 1,440 minutes after drug administration, respectively. No adverse systemic or local effects were observed following PB administration. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggested that IV-RLP of PB might be a viable alternative for treatment of horses with synovial infections caused by gram-negative bacteria.


Assuntos
Administração Intravenosa/veterinária , Cavalos , Polimixina B/administração & dosagem , Polimixina B/análise , Veia Safena , Líquido Sinovial/química , Animais , Antibacterianos/administração & dosagem , Antibacterianos/análise , Antibacterianos/metabolismo , Membro Posterior , Polimixina B/metabolismo , Distribuição Aleatória
19.
Colloids Surf B Biointerfaces ; 182: 110374, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31330430

RESUMO

This work presents the outcomes of a comparative study of molecular interactions of polymyxin B (PMB) and F12 and F13 formulations in the mole ratios of 1:2 and 1:3 of PMB:sodium deoxycholate sulfate (SDCS), respectively, and a commercial PMB formulation (CPMB) with lipopolysaccharides (LPS). Several spectroscopic and interfacial studies were performed to obtain LPS-peptide interactions at a molecular level. The fluorescence titrimetry method revealed that the F12 formulation (325 nM) exhibited a lower number of binding sites to the LPS compared to CPMB and F13 as well as PMB alone (537 nM). Similarly, in the presence of LPS, the F12 formulation (88 nm) exhibited smaller particle sizes in the dynamic light scattering study compared to PMB (116 nm), CPMB, and the F13 formulation. An interfacial study and circular dichroism spectroscopy revealed PMB and CPMB insertion into the LPS micelles to destabilize and disrupt the LPS membrane, whereas the F12 and F13 formulations may induce pseudo-aggregation. The NMR and IR studies showed that the presence of SDCS, the hydrophobicity of PMB increased by hydrogen bonding and electrostatic interactions and formed stabilized PMB-SDCS micelles. The PMB-SDCS formulation is likely to release PMB for easy penetration into the lipid membrane and cause disruption of the complex LPS micelles. Furthermore, the PMB-SDCS formulations neutralized and detoxified the LPS micelles with minimal toxicity to normal kidney tubular cells as well as an immortalised kidney cell line. The antimicrobial properties of PMBloaded SDCS nanomicelles were effective against a resistant strain of Pseudomonas aeruginosa.


Assuntos
Ácido Desoxicólico/química , Lipopolissacarídeos/química , Polimixina B/química , Sulfatos/química , Antibacterianos/química , Antibacterianos/farmacologia , Sítios de Ligação , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Desoxicólico/metabolismo , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Escherichia coli/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Polimixina B/metabolismo , Polimixina B/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Eletricidade Estática , Sulfatos/metabolismo
20.
J Microbiol ; 57(8): 704-710, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31187416

RESUMO

KatA is the major catalase required for hydrogen peroxide (H2O2) resistance and acute virulence in Pseudomonas aeruginosa PA14, whose transcription is governed by its dual promoters (katAp1 and katAp2). Here, we observed that KatA was not required for acute virulence in another wild type P. aeruginosa strain, PAO1, but that PAO1 exhibited higher KatA expression than PA14 did. This was in a good agreement with the observation that PAO1 was more resistant than PA14 to H2O2 as well as to the antibiotic peptide, polymyxin B (PMB), supposed to involve reactive oxygen species (ROS) for its antibacterial activity. The higher KatA expression in PAO1 than in PA14 was attributed to both katAp1 and katAp2 transcripts, as assessed by S1 nuclease mapping. In addition, it was confirmed that the PMB resistance is attributed to both katAp1 and katAp2 in a complementary manner in PA14 and PAO1, by exploiting the promoter mutants for each -10 box (p1m, p2m, and p1p2m). These results provide an evidence that the two widely used P. aeruginosa strains display different virulence mechanisms associated with OxyR and Anr, which need to be further characterized for better understanding of the critical virulence pathways that may differ in various P. aeruginosa strains.


Assuntos
Proteínas de Bactérias/genética , Catalase/genética , Regulação Bacteriana da Expressão Gênica , Peróxido de Hidrogênio/metabolismo , Polimixina B/metabolismo , Pseudomonas aeruginosa/enzimologia , Antibacterianos/metabolismo , Regiões Promotoras Genéticas , Pseudomonas aeruginosa/patogenicidade , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA