Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Publication year range
1.
Ann Clin Microbiol Antimicrob ; 21(1): 38, 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36038932

ABSTRACT

BACKGROUND: Antibiotic-resistant Gram-negative bacteria are becoming a major public health threat such as the important opportunistic pathogen Pseudomonas aeruginosa (P. aeruginosa). The present study investigated enhancement of the linezolid spectrum, which is normally used to treat Gram-positive bacteria, at inhibiting P. aeruginosa growth. METHODS: The checkerboard test or time-kill assay were carried out to determine the antibacterial effects of linezolid in cooperation with polymyxin B octapeptide PBOP (LP) against P. aeruginosa based on in vitro model. The protective effect of LP against P. aeruginosa infection was assessed based on a Caenorhabditis elegans (C. elegans) model. RESULTS: The synergistic activity and antibacterial effects were significantly increased against P. aeruginosa by LP treatment, while linezolid and PBOP as monotherapies exhibited no remarkably bactericidal activity against the clinical strains. Additionally, LP treatment modified biofilm production, morphology, swimming motility of P. aeruginosa, and protected C. elegans from P. aeruginosa infection. CONCLUSIONS: This research demonstrates that LP combination has significant synergistic activity against P. aeruginosa, and PBOP is potential to be an activity enhancer. Notably, this strategy improved the antibacterial activity spectrum of linezolid and other anti-Gram-positive agents and represents an effective choice to surmount the antibiotic resistance of bacteria in the long term.


Subject(s)
Caenorhabditis elegans , Pseudomonas aeruginosa , Animals , Anti-Bacterial Agents/pharmacology , Drug Synergism , Humans , Linezolid/pharmacology , Microbial Sensitivity Tests , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology
2.
Bioorg Chem ; 80: 639-648, 2018 10.
Article in English | MEDLINE | ID: mdl-30053708

ABSTRACT

Continuous development of new antibacterial agents is necessary to counter the problem of antimicrobial resistance. Polymyxins are considered as drugs of last resort to combat multidrug-resistant Gram-negative pathogens. Structural optimization of polymyxins requires an in-depth understanding of its structure and how it relates to its antibacterial activity. Herein, the effect of hydrophobicity was explored by adding a secondary fatty acyl component of varying length onto the polymyxin structure at the amine side-chain of l-diaminobutyric acid at position 1, resulting to the development of dilipid polymyxins. The incorporation of an additional lipid was found to confer polymyxin activity against Gram-positive bacteria, to which polymyxins are inherently inactive against. The dilipid polymyxins showed selective antibacterial activity against Pseudomonas aeruginosa. Moreover, dilipid polymyxin 1 that consists of four carbon-long aliphatic lipids displayed the ability to enhance the antibacterial potency of other antibiotics in combination against P. aeruginosa, resembling the adjuvant activity of the well-known outer membrane permeabilizer polymyxin B nonapeptide (PMBN). Interestingly, our data revealed that dilipid polymyxin 1 and PMBN are substrates for the MexAB-OprM efflux system, and therefore are affected by efflux. In contrast, dilipid polymyxin analogs that consist of longer lipids and colistin were not affected by efflux, suggesting that the lipid component of polymyxin plays an important role in resisting active efflux. Our work described herein provides an understanding to the polymyxin structure that may be used to usher the development of enhanced polymyxin analogs.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Lipids/chemistry , Lipids/pharmacology , Polymyxins/chemistry , Polymyxins/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Bacterial Infections/drug therapy , Drug Design , Hemolysis/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Lipids/chemical synthesis , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Polymyxins/chemical synthesis , Swine
3.
Biochim Biophys Acta ; 1858(2): 333-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26607008

ABSTRACT

Resistance to all known antibiotics is a growing concern worldwide, and has renewed the interest in antimicrobial peptides, a structurally diverse class of amphipathic molecules that essentially act on the bacterial membrane. Propelled by the antimicrobial potential of this compound class, we have designed three new lipopeptides derived from polymyxin B, sp-34, sp-96 and sp-100, with potent antimicrobial activity against both Gram positive and Gram negative bacteria. The three peptides bind with high affinity to lipopolysaccharide as demonstrated by monolayer penetration and dansyl-displacement. The interaction with the cytoplasmic membrane has been elucidated by biophysical experiments with model membranes of POPG or POPE/POPG (6:4), mimicking the Gram positive and Gram negative bacterial membrane. Trp-based fluorescence experiments including steady-state, quenching, anisotropy and FRET, reveal selectivity for anionic phospholipids and deep insertion into the membrane. All three lipopeptides induce membrane fusion and leakage from anionic vesicles, a process that is favored by the presence of POPE. The molecules bind to zwitterionic POPC vesicles, a model of the eukaryotic membrane, but in a different way, with lower affinity, less penetration into the bilayer and no fusion or permeabilization of the membrane. Results in model membranes are consistent with flow cytometry experiments in Escherichia coli and Staphylococcus aureus using a membrane potential sensitive dye (bis-oxonol) and a nucleic acid dye (propidium iodide), suggesting that the mechanism of action is based on membrane binding and collapse of membrane integrity by depolarization and permeabilization.


Subject(s)
Anti-Bacterial Agents , Escherichia coli/growth & development , Polymyxin B , Staphylococcus aureus/growth & development , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Polymyxin B/analogs & derivatives , Polymyxin B/chemical synthesis , Polymyxin B/chemistry , Polymyxin B/pharmacology
4.
J Bacteriol ; 197(15): 2479-88, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25962916

ABSTRACT

UNLABELLED: The constitutively expressed AcrAB multidrug efflux system of Escherichia coli shows a high degree of homology with the normally silent AcrEF system. Exposure of a strain with acrAB deleted to antibiotic selection pressure frequently leads to the insertion sequence-mediated activation of the homologous AcrEF system. In this study, we used strains constitutively expressing either AcrAB or AcrEF from their normal chromosomal locations to resolve a controversy about whether phenylalanylarginine ß-naphthylamide (PAßN) inhibits the activities of AcrAB and AcrEF and/or acts synergistically with antibiotics by destabilizing the outer membrane permeability barrier. Real-time efflux assays allowed a clear distinction between the efflux pump-inhibiting activity of PAßN and the outer membrane-destabilizing action of polymyxin B nonapeptide (PMXBN). When added in equal amounts, PAßN, but not PMXBN, strongly inhibited the efflux activities of both AcrAB and AcrEF pumps. In contrast, when outer membrane destabilization was assessed by the nitrocefin hydrolysis assay, PMXBN exerted a much greater damaging effect than PAßN. Strong action of PAßN in inhibiting efflux activity compared to its weak action in destabilizing the outer membrane permeability barrier suggests that PAßN acts mainly by inhibiting efflux pumps. We concluded that at low concentrations, PAßN acts specifically as an inhibitor of both AcrAB and AcrEF efflux pumps; however, at high concentrations, PAßN in the efflux-proficient background not only inhibits efflux pump activity but also destabilizes the membrane. The effects of PAßN on membrane integrity are compounded in cells unable to extrude PAßN. IMPORTANCE: The increase in multidrug-resistant bacterial pathogens at an alarming rate has accelerated the need for implementation of better antimicrobial stewardship, discovery of new antibiotics, and deeper understanding of the mechanism of drug resistance. The work carried out in this study highlights the importance of employing real-time fluorescence-based assays in differentiating multidrug efflux-inhibitory and outer membrane-destabilizing activities of antibacterial compounds.


Subject(s)
Bacterial Outer Membrane Proteins/physiology , Cell Membrane/physiology , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Genes, MDR/physiology , Membrane Transport Proteins/physiology , Animals , Anti-Bacterial Agents/pharmacology , Dipeptides/pharmacology , Drug Resistance, Multiple, Bacterial , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Genes, MDR/genetics , Humans , Microbial Sensitivity Tests , Permeability , Phenotype , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Rabbits
5.
Antimicrob Agents Chemother ; 59(12): 7823-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392511

ABSTRACT

We investigated the antimicrobial activity of four polymyxin B components, B1, B2, B3, and isoleucine (Ile)-B1, individually and in combination. B3 was the most active agent against all organisms tested except Acinetobacter baumannii, for which Ile-B1 was most active. One combination met the criteria for synergy, B3 plus Ile-B1. No combinations exhibited antagonism. The dominant components of polymyxin B products (B1 and B2) were associated with the lowest probability of improved antibacterial activity when combined.


Subject(s)
Anti-Bacterial Agents/pharmacology , Caprylates/chemistry , Polymyxin B/pharmacology , Structure-Activity Relationship , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/growth & development , Drug Synergism , Escherichia coli/drug effects , Escherichia coli/growth & development , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Microbial Sensitivity Tests , Polymyxin B/analogs & derivatives , Polymyxin B/chemistry , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
6.
Antimicrob Agents Chemother ; 59(12): 7489-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392495

ABSTRACT

Polymyxins are cyclic lipopeptide antibiotics that serve as a last line of defense against Gram-negative bacterial superbugs. However, the extensive accumulation of polymyxins in renal tubular cells can lead to nephrotoxicity, which is the major dose-limiting factor in clinical use. In order to gain further insights into the mechanism of polymyxin-induced nephrotoxicity, we have rationally designed novel fluorescent polymyxin probes to examine the localization of polymyxins in rat renal tubular (NRK-52E) cells. Our design strategy focused on incorporating a dansyl fluorophore at the hydrophobic centers of the polymyxin core structure. To this end, four novel regioselectively labeled monodansylated polymyxin B probes (MIPS-9541, MIPS-9542, MIPS-9543, and MIPS-9544) were designed, synthesized, and screened for their antimicrobial activities and apoptotic effects against rat kidney proximal tubular cells. On the basis of the assessment of antimicrobial activities, cellular uptake, and apoptotic effects on renal tubular cells, incorporation of a dansyl fluorophore at either position 6 or 7 (MIPS-9543 and MIPS-9544, respectively) of the polymyxin core structure appears to be an appropriate strategy for generating representative fluorescent polymyxin probes to be utilized in intracellular imaging and mechanistic studies. Furthermore, confocal imaging experiments utilizing these probes showed evidence of partial colocalization of the polymyxins with both the endoplasmic reticulum and mitochondria in rat renal tubular cells. Our results highlight the value of these new fluorescent polymyxin probes and provide further insights into the mechanism of polymyxin-induced nephrotoxicity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dansyl Compounds/pharmacology , Epithelial Cells/drug effects , Fluorescent Dyes/pharmacology , Polymyxin B/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/growth & development , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Apoptosis/drug effects , Biological Transport , Cell Line , Dansyl Compounds/chemical synthesis , Dansyl Compounds/metabolism , Drug Design , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Hydrophobic and Hydrophilic Interactions , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/ultrastructure , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Microbial Sensitivity Tests , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Molecular Imaging/methods , Polymyxin B/analogs & derivatives , Polymyxin B/chemical synthesis , Polymyxin B/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Rats
7.
J Pept Sci ; 21(3): 231-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25640745

ABSTRACT

The antimicrobial activity of polymyxins against Gram-negative bacteria has been known for several decades, but the mechanism of action leading to cell death has not been fully explored. A key step after binding of the antibiotic to lipopolysaccharide (LPS) exposed at the cell surface is 'self-promoted uptake' across the outer membrane (OM), in which the antibiotic traverses the asymmetric LPS-phospholipid bilayer before reaching the periplasm and finally targeting and disrupting the bacterial phospholipid inner membrane. The work described here was prompted by the hypothesis that polymyxins might interact with proteins in the OM, as part of their self-promoted uptake and permeabilizing effects. One way to test this is through photolabeling experiments. We describe the design and synthesis of a photoprobe based upon polymyxin B, containing photoleucine and an N-acyl group with a terminal alkyne suitable for coupling to a biotin tag using click chemistry. The resulting photoprobe retains potent antimicrobial activity, and in initial photolabeling experiments with Escherichia coli ATCC25922 is shown to photolabel several OM proteins. This photoprobe might be a valuable tool in more detailed studies on the mechanism of action of this family of antibiotics.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Cell Membrane/drug effects , Escherichia coli/drug effects , Molecular Probes/chemical synthesis , Polymyxin B/chemical synthesis , Staining and Labeling/methods , Alkynes/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/chemistry , Biotin/chemistry , Cell Membrane/chemistry , Click Chemistry , Escherichia coli/chemistry , Escherichia coli/physiology , Light , Lipopolysaccharides/chemistry , Microbial Sensitivity Tests , Molecular Probe Techniques , Molecular Probes/pharmacology , Phospholipids/chemistry , Photochemical Processes , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Solid-Phase Synthesis Techniques/methods
8.
BMC Microbiol ; 14: 65, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24629000

ABSTRACT

BACKGROUND: Nitric oxide (NO) is produced as part of the host immune response to bacterial infections, including urinary tract infections. The enzyme flavohemoglobin, coded by the hmp gene, is involved in protecting bacterial cells from the toxic effects of NO and represents a potentially interesting target for development of novel treatment concepts against resistant uropathogenic bacteria. The aim of the present study was to investigate if the in vitro antibacterial effects of NO can be enhanced by pharmacological modulation of the enzyme flavohemoglobin. RESULTS: Four clinical isolates of multidrug-resistant extended-spectrum ß-lactamase (ESBL)-producing uropathogenic E. coli were included in the study. It was shown that the NO-donor substance DETA/NO, but not inactivated DETA/NO, caused an initial growth inhibition with regrowth noted after 8 h of exposure. An hmp-deficient strain showed a prolonged growth inhibition in response to DETA/NO compared to the wild type. The imidazole antibiotic miconazole, that has been shown to inhibit bacterial flavohemoglobin activity, prolonged the DETA/NO-evoked growth inhibition. When miconazole was combined with polymyxin B nonapeptide (PMBN), in order to increase the bacterial wall permeability, DETA/NO caused a prolonged bacteriostatic response that lasted for up to 24 h. CONCLUSION: An NO-donor in combination with miconazole and PMBN showed enhanced antimicrobial effects and proved effective against multidrug-resistant ESBL-producing uropathogenic E. coli.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/metabolism , Hemeproteins/metabolism , Miconazole/pharmacology , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide/pharmacology , Polymyxin B/analogs & derivatives , Uropathogenic Escherichia coli/drug effects , Drug Synergism , Humans , Microbial Sensitivity Tests , Nanoparticles/metabolism , Polymyxin B/pharmacology , Uropathogenic Escherichia coli/enzymology , Uropathogenic Escherichia coli/growth & development , beta-Lactamases/metabolism
9.
Bioconjug Chem ; 25(4): 750-60, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24635310

ABSTRACT

The dry antibiotic development pipeline coupled with the emergence of multidrug resistant Gram-negative 'superbugs' has driven the revival of the polymyxin lipopeptide antibiotics. Polymyxin resistance implies a total lack of antibiotics for the treatment of life-threatening infections. The lack of molecular imaging probes that possess native polymyxin-like antibacterial activity is a barrier to understanding the resistance mechanisms and the development of a new generation of polymyxin lipopeptides. Here we report the regioselective modification of the polymyxin B core scaffold at the N-terminus with the dansyl fluorophore to generate an active probe that mimics polymyxin B pharmacologically. Time-lapse laser scanning confocal microscopy imaging of the penetration of probe (1) into Gram-negative bacterial cells revealed that the probe initially accumulates in the outer membrane and subsequently penetrates into the inner membrane and finally the cytoplasm. The implementation of this polymyxin-mimetic probe will advance the development of platforms for the discovery of novel polymyxin lipopeptides with efficacy against polymyxin-resistant strains.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Drug Design , Gram-Negative Bacteria/metabolism , Molecular Imaging , Polymyxin B/analogs & derivatives , Polymyxin B/metabolism , Acinetobacter baumannii/cytology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/growth & development , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Klebsiella pneumoniae/cytology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Microbial Sensitivity Tests , Microscopy, Electron , Models, Molecular , Molecular Conformation , Polymyxin B/chemistry , Polymyxin B/pharmacology , Pseudomonas aeruginosa/cytology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development
10.
Microbiol Spectr ; 12(4): e0368723, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38391225

ABSTRACT

Antibiotic-resistant Gram-negative bacteria remain a globally leading cause of bacterial infection-associated mortality, and it is imperative to identify novel therapeutic strategies. Recently, the advantage of using antibacterials selective against Gram-negative bacteria has been demonstrated with polymyxins that specifically target the lipopolysaccharides of Gram-negative bacteria. However, the severe cytotoxicity of polymyxins limits their clinical use. Here, we demonstrate that polymyxin B nonapeptide (PMBN), a polymyxin B derivative without the terminal amino acyl residue, can significantly enhance the effectiveness of commonly used antibiotics against only Gram-negative bacteria and their persister cells. We show that although PMBN itself does not exhibit antibacterial activity or cytotoxicity well above the 100-fold minimum inhibitory concentration of polymyxin B, PMBN can increase the potency of co-treated antibiotics. We also demonstrate that using PMBN in combination with other antibiotics significantly reduces the frequency of resistant mutant formation. Together, this work provides evidence of the utilities of PMBN as a novel potentiator for antibiotics against Gram-negative bacteria and insights for the eradication of bacterial persister cells during antibiotic treatment. IMPORTANCE: The significance of our study lies in addressing the problem of antibiotic-resistant Gram-negative bacteria, which continue to be a global cause of mortality associated with bacterial infections. Therefore, identifying innovative therapeutic approaches is an urgent need. Recent research has highlighted the potential of selective antibacterials like polymyxins, which specifically target the lipopolysaccharides of Gram-negative bacteria. However, the clinical use of polymyxins is limited by their severe cytotoxicity. This study unveils the effectiveness of polymyxin B nonapeptide (PMBN) in significantly enhancing the eradication of persister cells in Gram-negative bacteria. Although PMBN itself does not exhibit antibacterial activity or cytotoxicity, it remarkably reduces persister cells during the treatment of antibiotics. Moreover, combining PMBN with other antibiotics reduces the emergence of resistant mutants. Our research emphasizes the utility of PMBN as a novel potentiator to decrease persister cells during antibiotic treatments for Gram-negative bacteria.


Subject(s)
Bacterial Infections , Gram-Negative Bacterial Infections , Polymyxin B/analogs & derivatives , Humans , Polymyxin B/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Polymyxins/chemistry , Polymyxins/pharmacology , Gram-Negative Bacteria , Lipopolysaccharides , Microbial Sensitivity Tests
11.
Sci Rep ; 12(1): 11629, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35804085

ABSTRACT

The study of clinically relevant bacterial pathogens relies on molecular and genetic approaches. However, the generally low transformation frequency among natural isolates poses technical hurdles to widely applying common methods in molecular biology, including transformation of large constructs, chromosomal genetic manipulation, and dense mutant library construction. Here we demonstrate that culturing clinical isolates in the presence of polymyxin B nonapeptide (PMBN) improves their transformation frequency via electroporation by up to 100-fold in a dose-dependent and reversible manner. The effect was observed for PMBN-binding uropathogenic Escherichia coli (UPEC) and Salmonella enterica strains but not naturally polymyxin resistant Proteus mirabilis. Using our PMBN electroporation method we show efficient delivery of large plasmid constructs into UPEC, which otherwise failed using a conventional electroporation protocol. Moreover, we show a fivefold increase in the yield of engineered mutant colonies obtained in S. enterica with the widely used lambda-Red recombineering method, when cells are cultured in the presence of PMBN. Lastly, we demonstrate that PMBN treatment can enhance the delivery of DNA-transposase complexes into UPEC and increase transposon mutant yield by eightfold when constructing Transposon Insertion Sequencing (TIS) libraries. Therefore, PMBN can be used as a powerful electropermeabilisation adjuvant to aid the delivery of DNA and DNA-protein complexes into clinically important bacteria.


Subject(s)
Escherichia coli , Polymyxin B , Electroporation , Polymyxin B/analogs & derivatives , Polymyxin B/chemistry , Polymyxin B/pharmacology , Polymyxins
12.
Chem Pharm Bull (Tokyo) ; 59(5): 597-602, 2011.
Article in English | MEDLINE | ID: mdl-21532197

ABSTRACT

Polymyxin B (PMB) is a cationic cyclic decapeptide antibiotic with a fatty acyl (FA) modification at the α-amino group of Dab¹ (Dab: L-α,γ-diaminobutyric acid). In this study, which is part of a series of PMB structure-activity relationship investigations focused on identifying clinically useful peptide antibiotics, we synthesized ten des-FA PMB derivatives whose N-terminal moieties were changed to basic or hydrophilic amino acids. The antimicrobial and lipopolysaccharide (LPS) binding activities of these synthetic analogs were tested. The analogs showed more potent antimicrobial activity against Pseudomonas aeruginosa (P. aeruginosa) compared with the PMB nonapeptide. In particular, [Ser²-Dap³]-PMB(2-10), Guanyl-[Thr²-Dab³]-PMB(2-10), Guanyl-[Dab¹-Thr²-Dab³]-PMB(1-10), and N(α,γ)-diguanyl-[Dap³]-PMB(3-10) had antimicrobial activity equivalent to PMB. In LPS binding assays, the displacement curves shifted in a manner proportional to the number of positive charges available to bind to Escherichia coli (E. coli) and P. aeruginosa. Furthermore, peptides with basic side chains were comparable to PMB in binding activity assays against E. coli and P. aeruginosa. The acute toxicities of the peptides were evaluated by intravenously administering the peptides to mice through the tail vein. The toxicities of [Ser²-Dap³]-PMB(2-10), [Dap³]-PMB(3-10), and [Ser³]-PMB(3-10) were lower that of PMB (LD50, 4.8 µmol/kg).


Subject(s)
Anti-Infective Agents/pharmacology , Fatty Acids, Unsaturated/pharmacology , Polymyxin B/pharmacology , Pseudomonas aeruginosa/drug effects , Animals , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Anti-Infective Agents/toxicity , Binding Sites , Escherichia coli/drug effects , Fatty Acids, Unsaturated/chemical synthesis , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/toxicity , Lipopolysaccharides/metabolism , Male , Mice , Mice, Inbred Strains , Microbial Sensitivity Tests , Polymyxin B/analogs & derivatives , Polymyxin B/chemical synthesis , Polymyxin B/chemistry , Polymyxin B/toxicity , Structure-Activity Relationship
13.
Antimicrob Agents Chemother ; 54(8): 3341-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20479195

ABSTRACT

Polymyxins are cationic lipopeptides (five cationic charges) and the last resort for the treatment of serious Gram-negative infections caused by multiresistant strains. NAB741 has a cyclic peptide portion identical to that of polymyxin B but carries in the linear peptide portion a threonyl-D-serinyl residue (no cationic charges) instead of the diaminobutyryl-threonyl-diaminobutyryl residue (two cationic charges). At the N terminus of the peptide, NAB741 carries an acetyl group instead of a mixture of methyl octanoyl and methyl heptanoyl residues. NAB741 sensitized Escherichia coli, Klebsiella pneumoniae, Enterobacter cloacae, and Acinetobacter baumannii to antibiotics against which the intact outer membrane is an effective permeability barrier. When tested by using Etest strips on plates containing increasing concentrations of NAB741, the fractional inhibition concentration index (FICI) of the combination of NAB741 with rifampin ranged from

Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Cell Membrane Permeability , Gram-Negative Bacteria/drug effects , Polymyxin B/analogs & derivatives , Polymyxin B/metabolism , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Cricetinae , Cricetulus , Drug Synergism , Fibroblasts/drug effects , Gram-Negative Bacteria/classification , Guinea Pigs , Lung/cytology , Lung/drug effects , Male , Microbial Sensitivity Tests , Polymyxin B/chemistry , Polymyxin B/pharmacology , Polymyxin B/toxicity , Rats , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 20(5): 1771-5, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20138759

ABSTRACT

A series of cationic cyclic heptapeptides based on polymyxin B have been synthesized for use as permeabilizers of the outer membrane of Gram-negative bacteria. Only analogs with the Dab(2)-d-Phe(3)-Leu(4)-Xxx(5) sequence (Xxx = Dab or Orn) showed a synergistic bactericidal effect when combined with conventional antibiotics, indicating that the Dab(2) residue plays a critical role in permeation of the outer membrane of Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents/chemistry , Cell Membrane Permeability/drug effects , Escherichia coli/drug effects , Peptides, Cyclic/chemistry , Polymyxin B/analogs & derivatives , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Circular Dichroism , Microbial Sensitivity Tests , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Polymyxin B/chemistry , Polymyxin B/pharmacology , Structure-Activity Relationship
15.
Int J Antimicrob Agents ; 56(1): 106011, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32417206

ABSTRACT

INTRODUCTION: This study examined ceftazidime-avibactam activity against carbapenem-resistant Enterobacterales (CRE) clinical isolates and resistance mechanisms among non-metallo ß-lactamase (MBL) producers displaying ceftazidime-avibactam MIC values at 4 mg/L. METHODS: CRE isolates (286 of 8161 Enterobacterales) collected in Asia-Pacific, Europe and Latin America during 2016 were screened for carbapenemase genes. Selected isolates were susceptibility tested for ceftazidime-avibactam in the presence or absence of phenylalanine-arginyl ß-naphthylamide (PAßN) and polymyxin B nonapeptide (PMBN). Genome sequences were investigated for the integrity of outer membrane protein (OMP) genes and multilocus sequence typing. qRT-PCR assays were conducted to determine expression of acrA, ampC, and OMP genes. RESULTS: Ceftazidime-avibactam inhibited 99.2% of the Enterobacterales, 22 (78.7%) of the 286 CRE and 226 (100%) non-MBL producers. Among carbapenemase producers (85.3%; 244 of 286), the most common gene was blaKPC (76 blaKPC-3 and 46 blaKPC-2), followed by blaOXA-48-like (60 isolates) and blaNDM (37). Ceftazidime-avibactam MIC values at 4 mg/L were noted among 14 Klebsiella pneumoniae (13 carrying blaKPC and 1 blaCTX-M-15) mostly from Italy and Brazil and 1 Klebsiella aerogenes overexpressing ampC. PAßN did not significantly decrease ceftazidime-avibactam results, but adding PMBN did significantly decrease the MIC results for the combination. All K. pneumoniae isolates had a premature stop codon at OmpK35 and most isolates had L3 alterations of OmpK36, low expression of this gene, or OmpC disruption (K. aerogenes). Nine K. pneumoniae isolates belonged to clonal complex 258 and displayed intrahospital clonality. CONCLUSION: Ceftazidime-avibactam is an important addition to the armamentarium against multidrug-resistant organisms, and elevated MIC results for this combination seem to be associated with L3 OmpK36 alterations and ß-lactamases able to hydrolyze ceftazidime.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Bacterial Proteins/genetics , Carbapenem-Resistant Enterobacteriaceae/genetics , Ceftazidime/pharmacology , Klebsiella pneumoniae/genetics , Porins/genetics , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/drug effects , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Drug Combinations , Drug Resistance, Multiple, Bacterial/genetics , Enterobacter aerogenes/drug effects , Enterobacter aerogenes/genetics , Enterobacter aerogenes/isolation & purification , Humans , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , beta-Lactamases/metabolism
16.
J Microbiol ; 57(5): 405-412, 2019 May.
Article in English | MEDLINE | ID: mdl-30796747

ABSTRACT

Xanthorrhizol (XTZ), isolated from Curcuma xanthorrhiza, has potent antifungal and antibacterial activity. It shows very strong activity against Gram-positive bacteria, such as Streptococcus mutans and Staphylococcus aureus, but is generally not active against Gram-negative bacteria. In this study, we explored the possibility of using a combination strategy for expanding the antimicrobial spectrum of XTZ against Gram-negative bacteria. To take advantage of XTZ being a food-grade material, 10 food-grade or generally recognized as safe (GRAS) antimicrobial compounds with low toxicities were selected for combination therapy. In addition, polymyxin B nonapeptide (PMBN), which is less toxic than polymyxin B, was also selected as an outer membrane permeabilizer. The antibacterial activity of various double or triple combinations with or without XTZ were assayed in vitro against four Gram-negative bacterial species (Escherichia coli, Salmonella enterica serovar Typhi, Salmonella enterica serovar Typhimurium, and Vibrio cholerae), with synergistic combinations exhibiting clear activity subjected to further screening. The combinations with the greatest synergism were XTZ + PMBN + nisin, XTZ + PMBN + carvacrol, and XTZ + PMBN + thymol. These combinations also showed potent antimicrobial activity against Shigella spp., Yersinia enterocolitica, and Acinetobacter baumannii. In time-kill assays, the three combinations achieved complete killing of E. coli within 2 h, and S. Typhi and V. cholera within 15 min. This is the first report on expanding the activity spectrum of XTZ against Gram-negative bacteria through combination with PMBN and food-grade or GRAS substances, with the resulting findings being particularly useful for increasing the industrial and medical applications of XTZ.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Monoterpenes/pharmacology , Nisin/pharmacology , Phenols/pharmacology , Polymyxin B/analogs & derivatives , Acinetobacter baumannii/drug effects , Curcuma/metabolism , Cymenes , Drug Combinations , Drug Synergism , Escherichia coli/drug effects , Microbial Sensitivity Tests , Polymyxin B/pharmacology , Salmonella typhi/drug effects , Salmonella typhimurium/drug effects , Vibrio cholerae/drug effects , Yersinia enterocolitica/drug effects
17.
Antimicrob Agents Chemother ; 52(9): 3229-36, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18591267

ABSTRACT

The lack of novel antibiotics against gram-negative bacteria has reinstated polymyxins as the drugs of last resort to treat serious infections caused by extremely multiresistant gram-negative organisms. However, polymyxins are nephrotoxic, and this feature may complicate therapy or even require its discontinuation. Like that of aminoglycosides, the nephrotoxicity of polymyxins might be related to the highly cationic nature of the molecule. Colistin and polymyxin B carry five positive charges. Here we show that novel polymyxin derivatives carrying only three positive charges are effective antibacterial agents. NAB739 has a cyclic peptide portion identical to that of polymyxin B, but in the linear portion of the peptide, it carries the threonyl-D-serinyl residue (no cationic charges) instead of the diaminobutyryl-threonyl-diaminobutyryl residue (two cationic charges). The MICs of NAB739 for 17 strains of Escherichia coli were identical, or very close, to those of polymyxin B. Furthermore, NAB739 was effective against other polymyxin-susceptible strains of Enterobacteriaceae and against Acinetobacter baumannii. At subinhibitory concentrations, it dramatically sensitized A. baumannii to low concentrations of antibiotics such as rifampin, clarithromycin, vancomycin, fusidic acid, and meropenem. NAB739 methanesulfonate was a prodrug analogous to colistin methanesulfonate. NAB740 was the most active derivative against Pseudomonas aeruginosa. NAB7061 (linear portion of the peptide, threonyl-aminobutyryl) lacked direct antibacterial activity but sensitized the targets to hydrophobic antibiotics by factors up to 2,000. The affinities of the NAB compounds for isolated rat kidney brush border membrane were significantly lower than that of polymyxin B.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Cricetinae , Fibroblasts/drug effects , Gram-Negative Bacteria/classification , Kidney Cortex , Lung/cytology , Lung/drug effects , Microbial Sensitivity Tests , Microvilli/metabolism , Polymyxin B/metabolism , Polymyxin B/toxicity , Rats , Structure-Activity Relationship
18.
Int J Antimicrob Agents ; 31(3): 224-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18083010

ABSTRACT

The role of membrane permeabilisation and disruption in the mechanism of action of some polymyxin analogues against Gram-negative organisms is contentious. The effects of polymyxin B (PMB) and its analogue polymyxin B nonapeptide (PMBN) on Escherichia coli envelopes should correlate, but previous work by other workers suggests that PMBN has a different mode of action. This study has reassessed the biochemical techniques used previously and has shown that, in contrast to previous studies, PMBN (a well-characterised antibacterial synergist) readily releases periplasmic proteins and lipopolysaccharide from treated E. coli at subinhibitory concentrations in normal physiological buffer conditions. We conclude that, when tested with appropriate methodology, PMBN closely correlates with the early effects of PMB on the cell envelope of E. coli and this study shows that it is now consistent with the accepted interactions of membrane-active agents against Gram-negative cells.


Subject(s)
Cell Membrane Permeability/drug effects , Cell Membrane/drug effects , Escherichia coli/drug effects , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , Cephalosporins/metabolism , Escherichia coli Proteins/metabolism , Lipopolysaccharides/metabolism , Nitrophenols/metabolism , Periplasmic Proteins/metabolism , Polysaccharides, Bacterial/metabolism , Time Factors
19.
Bioorg Med Chem Lett ; 18(20): 5512-7, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18805690

ABSTRACT

Tetrapeptides derived from glycine and beta-alanine were hooked at the C-3beta position of the modified cholic acid to realize novel linear tetrapeptide-linked cholic acid derivatives. All the synthesized compounds were tested against a wide variety of microorganisms (gram-negative bacteria, gram-positive bacteria and fungi) and their cytotoxicity was evaluated against human embryonic kidney (HEK293) and human mammary adenocarcinoma (MCF-7) cell lines. While relatively inactive by themselves, these compounds interact synergistically with antibiotics such as fluconazole and erythromycin to inhibit growth of fungi and bacteria, respectively, at 1-24 microg/mL. The synergistic effect shown by our novel compounds is due to their inherent amphiphilicity. The fractional inhibitory concentrations reported are comparable to those reported for Polymyxin B derivatives.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Cholic Acid/chemistry , Peptides/chemistry , Anti-Bacterial Agents/pharmacology , Cell Line , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Drug Design , Erythromycin/pharmacology , Fluconazole/pharmacology , Glycine/chemistry , Humans , Models, Chemical , Molecular Conformation , Polymyxin B/analogs & derivatives , Polymyxin B/pharmacology , beta-Alanine/chemistry
20.
Int J Antimicrob Agents ; 52(3): 365-371, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29807163

ABSTRACT

A number of companies manufacture polymyxin B using United States Pharmacopeia (USP) metrics, rather than chemical composition, to report biological activity. Given that polymyxin B contains several different components, it is unknown whether pharmacokinetic and pharmacodynamic variability exists between the different brands and whether USP metrics capture this variability. Here we investigated the composition of polymyxin B obtained from four manufacturers (Sigma-Aldrich, AK Scientific, USP and MP Biomedicals) and evaluated their rate and extent of killing against multidrug-resistant Acinetobacter baumannii and Klebsiella pneumoniae using in vitro static time-kill experiments. Ultraviolet (UV) fingerprinting and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis revealed similarities and differences between component distributions. The significant differences between products, based on UV fingerprinting and LC-MS/MS, did not translate into pharmacodynamic differences at the three concentrations evaluated. The aggregate polymyxin B concentration, rather than that of the individual components, influences overall bacterial killing.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Klebsiella pneumoniae/drug effects , Polymyxin B , Acinetobacter baumannii/genetics , Chromatography, Liquid , Drug Resistance, Multiple, Bacterial/genetics , Humans , Klebsiella pneumoniae/genetics , Microbial Sensitivity Tests , Polymyxin B/analogs & derivatives , Polymyxin B/chemistry , Polymyxin B/pharmacology , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL