Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
PLoS One ; 9(2): e87730, 2014.
Article in English | MEDLINE | ID: mdl-24498363

ABSTRACT

Taking advantage of the cluster effect observed in multivalent peptides, this work describes antifungal activity and possible mechanism of action of tetravalent peptide (B4010) which carries 4 copies of the sequence RGRKVVRR through a branched lysine core. B4010 displayed better antifungal properties than natamycin and amphotericin B. The peptide retained significant activity in the presence of monovalent/divalent cations, trypsin and serum and tear fluid. Moreover, B4010 is non-haemolytic and non-toxic to mice by intraperitoneal (200 mg/kg) or intravenous (100 mg/kg) routes. S. cerevisiae mutant strains with altered membrane sterol structures and composition showed hyper senstivity to B4010. The peptide had no affinity for cell wall polysaccharides and caused rapid dissipation of membrane potential and release of vital ions and ATP when treated with C. albicans. We demonstrate that additives which alter the membrane potential or membrane rigidity protect C. albicans from B4010-induced lethality. Calcein release assay and molecular dynamics simulations showed that the peptide preferentially binds to mixed bilayer containing ergosterol over phophotidylcholine-cholesterol bilayers. The studies further suggested that the first arginine is important for mediating peptide-bilayer interactions. Replacing the first arginine led to a 2-4 fold decrease in antifungal activities and reduced membrane disruption properties. The combined in silico and in vitro approach should facilitate rational design of new tetravalent antifungal peptides.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Saccharomyces cerevisiae/drug effects , Animals , Calorimetry , Candida albicans/growth & development , Candida albicans/metabolism , Cell Membrane/metabolism , Cells, Cultured , Circular Dichroism , Conjunctiva/cytology , Conjunctiva/drug effects , Conjunctiva/metabolism , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hemolysis/drug effects , Humans , Lipid Bilayers/metabolism , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Rabbits , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Wound Healing/drug effects
2.
J Med Chem ; 56(6): 2359-73, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23441632

ABSTRACT

This work describes how to tune the amphiphilic conformation of α-mangostin, a natural compound that contains a hydrophobic xanthone scaffold, to improve its antimicrobial activity and selectivity for Gram-positive bacteria. A series of xanthone derivatives was obtained by cationic modification of the free C3 and C6 hydroxyl groups of α-mangostin with amine groups of different pKa values. Modified structures using moieties with high pKa values, such as AM-0016 (3b), exhibited potent antimicrobial properties against Gram-positive bacteria. Compound 3b also killed bacteria rapidly without inducing drug resistance and was nontoxic when applied topically. Biophysical studies and molecular dynamics simulations revealed that 3b targets the bacterial inner membrane, forming an amphiphilic conformation at the hydrophobic-water interface. In contrast, moieties with low pKa values reduced the antimicrobial activity of the parent compound when conjugated to the xanthone scaffold. This strategy provides a new way to improve "hits" for the development of membrane-active antibiotics that target drug-resistant pathogens.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Drug Design , Hydrophobic and Hydrophilic Interactions , Xanthones/chemistry , Xanthones/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Bacteria/drug effects , Cell Membrane/metabolism , Chemistry Techniques, Synthetic , Microbial Sensitivity Tests , Molecular Conformation , Molecular Dynamics Simulation , Rabbits , Substrate Specificity , Xanthones/chemical synthesis , Xanthones/toxicity
3.
Biochim Biophys Acta ; 1828(2): 834-44, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22982495

ABSTRACT

The emergence of methicillin-resistant Staphylococcus aureus (MRSA) has created the need for better therapeutic options. In this study, five natural xanthones were extracted and purified from the fruit hull of Garcinia mangostana and their antimicrobial properties were investigated. α-Mangostin was identified as the most potent among them against Gram-positive pathogens (MIC=0.78-1.56 µg/mL) which included two MRSA isolates. α-Mangostin also exhibited rapid in vitro bactericidal activity (3-log reduction within 5 min). In a multistep (20 passage) resistance selection study using a MRSA isolated from the eye, no resistance against α-mangostin in the strains tested was observed. Biophysical studies using fluorescence probes for membrane potential and permeability, calcein encapsulated large unilamellar vesicles and scanning electron microscopy showed that α-mangostin rapidly disrupted the integrity of the cytoplasmic membrane leading to loss of intracellular components in a concentration-dependent manner. Molecular dynamic simulations revealed that isoprenyl groups were important to reduce the free energy for the burial of the hydrophobic phenyl ring of α-mangostin into the lipid bilayer of the membrane resulting in membrane breakdown and increased permeability. Thus, we suggest that direct interactions of α-mangostin with the bacterial membrane are responsible for the rapid concentration-dependent membrane disruption and bactericidal action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Xanthones/pharmacology , Animals , Biophysics/methods , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Ethidium/pharmacology , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Gram-Positive Bacteria/chemistry , Hemolysis , Hydrophobic and Hydrophilic Interactions , Kinetics , Membrane Potentials , Microbial Sensitivity Tests , Microscopy, Electron, Scanning/methods , Models, Chemical , Organic Chemicals/pharmacology , Permeability , Rabbits , Time Factors , Xanthones/chemistry
4.
J Biol Chem ; 287(32): 26606-17, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22700968

ABSTRACT

In recent years, interest has grown in the antimicrobial properties of certain natural and non-natural peptides. The strategy of inserting a covalent branch point in a peptide can improve its antimicrobial properties while retaining host biocompatibility. However, little is known regarding possible structural transitions as the peptide moves on the access path to the presumed target, the inner membrane. Establishing the nature of the interactions with the complex bacterial outer and inner membranes is important for effective peptide design. Structure-activity relationships of an amphiphilic, branched antimicrobial peptide (B2088) are examined using environment-sensitive fluorescent probes, electron microscopy, molecular dynamics simulations, and high resolution NMR in solution and in condensed states. The peptide is reconstituted in bacterial outer membrane lipopolysaccharide extract as well as in a variety of lipid media mimicking the inner membrane of Gram-negative pathogens. Progressive structure accretion is observed for the peptide in water, LPS, and lipid environments. Despite inducing rapid aggregation of bacteria-derived lipopolysaccharides, the peptide remains highly mobile in the aggregated lattice. At the inner membranes, the peptide undergoes further structural compaction mediated by interactions with negatively charged lipids, probably causing redistribution of membrane lipids, which in turn results in increased membrane permeability and bacterial lysis. These findings suggest that peptides possessing both enhanced mobility in the bacterial outer membrane and spatial structure facilitating its interactions with the membrane-water interface may provide excellent structural motifs to develop new antimicrobials that can overcome antibiotic-resistant Gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents/chemistry , Peptides/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Fluorescent Dyes , Hemolysis/drug effects , Microbial Sensitivity Tests , Microscopy, Electron, Transmission , Molecular Dynamics Simulation , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptides/pharmacology , Protein Conformation
5.
Biochemistry ; 48(30): 7229-39, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19580334

ABSTRACT

Defensins are small (3-5 kDa) cysteine-rich cationic proteins found in both vertebrates and invertebrates constituting the front line of host innate immunity. Despite intensive research, bactericidal and cytotoxic mechanisms of defensins are still largely unknown. Moreover, we recently demonstrated that small peptides derived from defensins are even more potent bactericidal agents with less toxicity toward host cells. In this paper, structures of three C-terminal (R36-K45) analogues of human beta-defensin-3 were studied by 1H NMR spectroscopy and extensive molecular dynamics simulations. Because of indications that these peptides might target the inner bacterial membrane, they were reconstituted in dodecylphosphocholine or dodecylphosphocholine/1-palmitoyl-2-oleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] mixed micelles, and lipid bicelles mimicking the phospholipid-constituted bilayer membrane of mammalian and bacterial cells. The results show that the binding affinity and partitioning into the lipid phase and the ability to dimerize and accrete well-defined structures upon interactions with lipid membranes contribute to compactization of positive charges within peptide oligomers. The peptide charge density, mediated by corresponding three-dimensional structures, was found to directly correlate with the antimicrobial activity. These novel observations may provide a new rationale for the design of improved antimicrobial agents.


Subject(s)
Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Peptides/chemistry , Protein Conformation , beta-Defensins/chemistry , Amino Acid Sequence , Animals , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Computer Simulation , Humans , Lipid Bilayers/chemistry , Micelles , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Pseudomonas aeruginosa/drug effects , beta-Defensins/genetics , beta-Defensins/metabolism , beta-Defensins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL