Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
J Pept Sci ; 20(7): 517-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24863176

ABSTRACT

Bicelles are model membrane systems that can be macroscopically oriented in a magnetic field at physiological temperature. The macroscopic orientation of bicelles allows to detect, by means of magnetic resonance spectroscopies, small changes in the order of the bilayer caused by solutes interacting with the membrane. These changes would be hardly detectable in isotropic systems such as vesicles or micelles. The aim of this work is to show that bicelles represent a convenient tool to investigate the behavior of antimicrobial peptides (AMPs) interacting with membranes, using electron paramagnetic resonance (EPR) spectroscopy. We performed the EPR experiments on spin-labeled bicelles using various AMPs of different length, charge, and amphipathicity: alamethicin, trichogin GA IV, magainin 2, HP(2-20), and HPA3. We evaluated the changes in the order parameter of the spin-labeled lipids as a function of the peptide-to-lipid ratio. We show that bicelles labeled at position 5 of the lipid chains are very sensitive to the perturbation induced by the AMPs even at low peptide concentrations. Our study indicates that peptides that are known to disrupt the membrane by different mechanisms (i.e., alamethicin vs magainin 2) show very distinct trends of the order parameter as a function of peptide concentration. Therefore, spin-labeled bicelles proved to be a good system to evaluate the membrane disruption mechanism of new AMPs.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Dimyristoylphosphatidylcholine/chemistry , Lipid Bilayers/chemistry , Lipopeptides/chemistry , Phospholipid Ethers/chemistry , Alamethicin/chemistry , Hydrophobic and Hydrophilic Interactions , Micelles
2.
Biomaterials ; 35(3): 1025-39, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24176194

ABSTRACT

In response to the growing problem of multidrug-resistant pathogenic microbes, much attention is being paid to naturally occurring and synthetic antimicrobial peptides (AMPs) and the effects of their structural modification. Among these modifications, amino acid substitution is a simple approach to enhancing biological activity and reducing cytotoxicity. An earlier study indicated that HPA3, an analog of HP (2-20) derived from the N-terminus of Helicobacter pylori ribosomal protein L1, forms large pores and shows considerable cytotoxicity. However, HPA3P, in which a proline (Pro) is substituted for glutamic acid (Glu) at position 9 of HPA3, shows markedly less cytotoxicity. This may be attributable to the presence of a Pro-kink into middle of the HPA3P structure within the membrane environment. Unfortunately, HPA3P is not an effective antibacterial agent in vivo. We therefore designed a helix-PXXP-helix structure (HPA3P2), in which Pro was substituted for the Glu and phenylalanine (Phe) at positions 9 and 12 of HPA3, yielding a molecule with a flexible central hinge. As compared to HPA3P, HPA3P3 exhibited dramatically increased antibacterial activity in vivo. ICR mice infected with clinically isolated multidrug-resistant Pseudomonas aeruginosa showed 100% survival when administered one 0.5-mg/kg dose of HPA3P2 or three 0.1-mg/kg doses of HPA3P2. Moreover, in a mouse model of septic shock induced by P. aeruginosa LPS, HPA3P2 reduced production of pro-inflammatory mediators and correspondingly reduced lung (alveolar) and liver tissue damage. The changes in HPA3 behavior with the introduction of Pro likely reflects alterations of the mechanism of action: i) HPA3 forms pores in the bacterial cell membranes, ii) HPA3P permeates the cell membranes and binds to intracellular RNA and DNA, and iii) HPA3P2 acts on the outer cellular membrane component LPS. Collectively, these results suggest HPA3P2 has the potential to be an effective antibiotic for use against multidrug-resistant bacterial strains.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , Bacterial Proteins/therapeutic use , Helicobacter pylori/chemistry , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Ribosomal Proteins/therapeutic use , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/toxicity , Bacterial Proteins/chemistry , Bacterial Proteins/toxicity , Cell Line , Cell Survival/drug effects , Drug Resistance, Multiple, Bacterial , Hemolysis/drug effects , Humans , Mice , Mice, Inbred ICR , Protein Structure, Secondary , Ribosomal Proteins/chemistry , Ribosomal Proteins/toxicity
3.
J Pept Sci ; 19(12): 758-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24243598

ABSTRACT

Antimicrobial peptides (AMPs) are promising compounds for developing new antibiotic drugs against drug-resistant bacteria. Many of them kill bacteria by perturbing their membranes but exhibit no significant toxicity towards eukaryotic cells. The identification of the features responsible for this selectivity is essential for their pharmacological development. AMPs exhibit few conserved features, but a statistical analysis of an AMP sequence database indicated that many α-helical AMPs surprisingly have a helix-breaking Pro residue in the middle of their sequence. To discriminate among the different possible hypotheses for the functional role of this feature, we designed an analogue of the antimicrobial peptide P5, in which the central Pro was deleted (analogue P5Del). Pro removal resulted in a dramatic increase of toxicity. This was explained by the observation that P5Del binds both charged and neutral membranes, whereas P5 has no appreciable affinity towards neutral bilayers. CD and simulative data provided a rationalization of this behavior. In solution P5, due to the presence of Pro, attains compact conformations, in which its apolar residues are partially shielded from the solvent, whereas P5Del is more helical. These structural differences reduce the hydrophobic driving force for association of P5 to neutral membranes, whereas its binding to anionic bilayers can still take place because of electrostatic attraction. After membrane binding, the Pro residue does not preclude the attainment of a membrane-active amphiphilic helical conformation. These findings shed light on the role of Pro residues in the selectivity of AMPs and provide hints for the design of new, highly selective compounds.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Proline/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hemolysis/drug effects , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Structure, Secondary
4.
PLoS One ; 8(7): e67597, 2013.
Article in English | MEDLINE | ID: mdl-23935838

ABSTRACT

HP (2-20) is a 19-aa, amphipathic, α-helical peptide with antimicrobial properties that was derived from the N-terminus of Helicobacter pylori ribosomal protein L1. We previously showed that increasing the net hydrophobicity of HP (2-20) by substituting Trp for Gln(17) and Asp(19) (Anal 3) increased the peptide's antimicrobial activity. In hydrophobic medium, Anal 3 forms an amphipathic structure consisting of an N-terminal random coil region (residues 2-5) and an extended helical region (residues 6-20). To investigate the structure-activity relationship of Anal 3, we substituted Pro for Glu(9) (Anal 3-Pro) and then examined the new peptide's three-dimensional structure, antimicrobial activity and mechanism of action. Anal 3-Pro had an α-helical structure in the presence of trifluoroethanol (TFE) and sodium dodecyl sulfate (SDS). NMR spectroscopic analysis of Anal 3-Pro's tertiary structure in SDS micelles confirmed that the kink potential introduced by Pro(10) was responsible for the helix distortion. We also found that Anal 3-Pro exhibited about 4 times greater antimicrobial activity than Anal 3. Fluorescence activated flow cytometry and confocal fluorescence microscopy showed that incorporating a Pro-hinge into Anal 3 markedly reduced its membrane permeability so that it accumulated in the cytoplasm without remaining in the cell membrane. To investigate the translocation mechanism, we assessed its ability to release of FITC-dextran. The result showed Anal 3-Pro created a pore <1.8 nm in diameter, which is similar to buforin II. Notably, scanning electron microscopic observation of Candida albicans revealed that Anal 3-Pro and buforin II exert similar effects on cell membranes, whereas magainin 2 exerts a different, more damaging, effect. In addition, Anal 3-Pro assumed a helix-hinge-helix structure in the presence of biological membranes and formed micropores in both bacterial and fungal membranes, through which it entered the cytoplasm and tightly bound to DNA. These results indicate that the bending region of Anal 3- Pro peptide is prerequisite for effective antibiotic activity and may facilitate easy penetration of the lipid bilayers of the cell membrane.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Proline/chemistry , Amino Acid Sequence , Bacteria/cytology , Bacteria/drug effects , Candida albicans/cytology , Candida albicans/drug effects , Cell Membrane Permeability/drug effects , Chitin/metabolism , Cholesterol/metabolism , Circular Dichroism , Electrophoretic Mobility Shift Assay , Flow Cytometry , Hemolysis/drug effects , Humans , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Microscopy, Confocal , Molecular Sequence Data , Peptidoglycan/metabolism , Phosphatidylcholines/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Unilamellar Liposomes/metabolism
5.
Biochim Biophys Acta ; 1828(2): 443-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22982494

ABSTRACT

In an earlier study, we determined that HP(2-20) (residues 2-20 of parental HP derived from the N-terminus of the Helicobacter pylori ribosomal protein L1) and its analog, HPA3NT3, had potent antimicrobial effects. However, HPA3NT3 also showed undesirable cytotoxicity against HaCaT cells. In the present study, we designed peptide analogs including HPA3NT3-F1A (-F1A), HPA3NT3-F8A (-F8A), HPA3NT3-F1AF8A (-F1AF8A), HPA3NT3-A1 (-A1) and HPA3NT3-A2 (-A2) in an effort to investigate the effects of amino acid substitutions in reducing their hydrophobicity or increasing their cationicity, and any resulting effects on their selectivity in their interactions with human cells and pathogens, as well as their mechanism of antimicrobial action. With the exception of HPA3NT3-A1, all of these peptides showed potent antimicrobial activity. Moreover, substitution of Ala for Phe at positions 1 and/or 8 of the HPA3NT3 peptides (-F1A, -F8A and -F1AF8A) dramatically reduced their cytotoxicity. Thus the cytotoxicity of HPA3NT3 appears to be related to its Phe residues (positions 1 and 8), which strongly interact with sphingomyelin in the mammalian cell membrane. HPA3NT3 exerted its bactericidal effects through membrane permeabilization mediated by pore formation. In contrast, fluorescent dye leakage and nucleic acid gel retardation assays showed that -A2 acted by penetrating into the cytoplasm, where it bound to nucleic acids and inhibited protein synthesis. Notably, Staphylococcus aureus did not develop resistance to -A2 as it did with rifampin. These results suggest that the -A2 peptide could potentially serve as an effective antibiotic agent against multidrug-resistant bacterial strains.


Subject(s)
Anti-Infective Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Peptides/chemistry , Amino Acids/chemistry , Antifungal Agents/pharmacology , Cations , Cell Line, Tumor , Cell Membrane Permeability , Drug Resistance, Multiple, Fungal , Fluorescent Dyes/pharmacology , Humans , Kinetics , Liposomes/chemistry , Microbial Sensitivity Tests , Microscopy, Confocal/methods , Microscopy, Electron, Transmission/methods , Models, Chemical , Organic Chemicals/pharmacology , Rifampin/chemistry , Sphingomyelins/chemistry , Staphylococcus aureus/metabolism , Tryptophan/chemistry
6.
PLoS One ; 7(11): e47642, 2012.
Article in English | MEDLINE | ID: mdl-23144825

ABSTRACT

An antibacterial protein (about 12 kDa) was isolated from human amniotic fluid through dialysis, ultrafiltration and C18 reversed-phase HPLC steps. Automated Edman degradation showed that the N-terminal sequence of the antibacterial protein was NH(2)-Ile-Gln-Arg-Thr-Pro-Lys-Ile-Gln-Val-Tyr-Ser-Arg-His-Pro-Ala-Glu-Asn-Gly-. The N-terminal sequence of the antibacterial protein was found to be identical to that of ß(2)-microglobulin, a component of MHC class I molecules, which are present on all nucleated cells. Matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) revealed that the molecular mass of the antibacterial protein was 11,631 Da. This antibacterial protein, ß(2)M, possessed potent antibacterial activity against pathogenic bacteria. Specially, antibacterial activity was observed in potassium buffer, and potassium ion was found to be critical for the antibacterial activity. Interestingly, the antibacterial action of ß(2)M was associated with dissipation of the transmembrane potential, but the protein did not cause damage to the membrane that would result in SYTOX green uptake. In addition, stimulation of WISH amniotic epithelial cells with the bacterial endotoxin lipopolysaccharide (LPS) induced dose-dependent upregulation of ß(2)M mRNA expression. These results suggest that ß(2)M contributes to a self-defense response when amniotic cells are exposed to pathogens.


Subject(s)
Amniotic Fluid/metabolism , Antimicrobial Cationic Peptides/metabolism , beta 2-Microglobulin/metabolism , Amino Acid Sequence , Amniotic Fluid/cytology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Cells, Cultured , Chromatography, High Pressure Liquid , Drug Resistance, Bacterial , Epithelial Cells/immunology , Epithelial Cells/metabolism , Escherichia coli/drug effects , Escherichia coli/physiology , Female , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Lipopolysaccharides/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/physiology , Membrane Potentials/drug effects , Microbial Sensitivity Tests , Molecular Sequence Data , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, Protein , beta 2-Microglobulin/genetics , beta 2-Microglobulin/isolation & purification , beta 2-Microglobulin/pharmacology
7.
Langmuir ; 28(49): 17079-91, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23140333

ABSTRACT

Metal ions binding exert a crucial influence upon the aggregation properties and stability of peptides, and the propensity of folding in various substates. Herein, we demonstrate the use of the α-HL protein as a powerful nanoscopic tool to probe Cu(2+)-triggered physicochemical changes of a 20 aminoacids long, antimicrobial-derived chimera peptide with a His residue as metal-binding site, and simultaneously dissect the kinetics of the free- and Cu(2+)-bound peptide interaction to the α-HL pore. Combining single-molecule electrophysiology on reconstituted lipid membranes and fluorescence spectroscopy, we show that the association rate constant between the α-HL pore and a Cu(2+)-free peptide is higher than that of a Cu(2+)-complexed peptide. We posit that mainly due to conformational changes induced by the bound Cu(2+) on the peptide, the resulting complex encounters a higher energy barrier toward its association with the protein pore, stemming most likely from an extra entropy cost needed to fit the Cu(2+)-complexed peptide within the α-HL lumen region. The lower dissociation rate constant of the Cu(2+)-complexed peptide from α-HL pore, as compared to that of Cu(2+)-free peptide, supports the existence of a deeper free energy well for the protein interaction with a Cu(2+)-complexed peptide, which may be indicative of specific Cu(2+)-mediated contributions to the binding of the Cu(2+)-complexed peptide within the pore lumen.


Subject(s)
Bacterial Toxins/chemistry , Copper/chemistry , Hemolysin Proteins/chemistry , Histidine/chemistry , Lipid Bilayers/chemistry , Peptides/chemistry , Recombinant Fusion Proteins/chemistry , Antimicrobial Cationic Peptides/chemistry , Binding Sites , Kinetics , Magainins/chemistry , Membrane Potentials , Nanopores , Peptides/chemical synthesis , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemical synthesis , Spectrometry, Fluorescence , Thermodynamics
8.
Mol Biosyst ; 8(11): 2860-3, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22945295

ABSTRACT

Herein we explored the role of topological distribution of aromatic amino acids in peptide-membrane interfacial interactions. The membrane activity of closely related peptides and their binding energy is sensitive to the positioning of minimum two tryptophans, and by the degree of flanking at the membrane interface mediated by aromatic amino acids.


Subject(s)
Anti-Infective Agents/chemistry , Cell Membrane/metabolism , Peptides/chemistry , Tryptophan/chemistry , Protein Binding , Protein Conformation
9.
J Agric Food Chem ; 60(27): 6875-81, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22632466

ABSTRACT

Abalone is a valuable seafood in the aquaculture industry worldwide as it is rich in protein. However, to date, research on the functional proteins of abalone is lacking. Herein, we report two peptides with antibacterial activity from Nordotis discus discus . The purification of peptides was performed by solvent extraction, ultrafiltration, and reverse-phase high performance liquid chromatography. The N-terminal amino acid sequences of the isolated antibacterial peptides, named as Nod1 and Nod2, were identified by Edman degradation and did not show any similarity to other proteins and peptides in databases based on results of BLAST homology analysis. Molecular masses of Nod1 and Nod2 were 6145.06 and 6360.07 Da, respectively, as determined by mass spectrometric analysis. The two peptides displayed pH-dependent antibacterial activity against various bacteria that was more potent at pH 5.4 than pH 7.4, but they did not inhibit fungal growth at either pH levels. Their antibacterial activity was due to membranolytic action, which was assayed by SYTOX-green uptake. In addition, both peptides were virtually noncytolytic for human erythrocytes and mammalian cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biological Factors/pharmacology , Gastropoda/chemistry , Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Bacteria/drug effects , Biological Factors/chemistry , Biological Factors/isolation & purification , Erythrocytes/drug effects , Humans , Molecular Weight , Peptide Mapping , Peptides/chemistry , Peptides/isolation & purification
10.
Amino Acids ; 43(4): 1761-77, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22484376

ABSTRACT

The lipopeptaibol trichogin GA IV is a natural, non-ribosomally synthesized, antimicrobial peptide remarkably resistant to the action of hydrolytic enzymes. This feature may be connected to the multiple presence in its sequence of the non-coded residue α-aminoisobutyric acid (Aib), which is known to be responsible for the adoption of particularly stable helical structures already at the level of short peptides. To investigate the role of Aib residues on the 3D-structure and bioactivity of trichogin GA IV, we synthesized and fully characterized four analogs where one or two Aib residues are replaced by L-Leu. Our extensive conformational studies (including an X-ray diffraction analysis) and biological assays performed on these analogs showed that the Aib to L-Leu replacements do not affect the resistance to proteolysis, but modulate the bioactivity of trichogin GA IV in a 3D-structure related manner.


Subject(s)
Aminoisobutyric Acids/chemistry , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Lipopeptides/chemical synthesis , Lipopeptides/pharmacology , Amino Acid Substitution , Candida albicans/drug effects , Candida albicans/growth & development , Circular Dichroism , Escherichia coli/drug effects , Escherichia coli/growth & development , Kinetics , Leucine/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Spectroscopy, Fourier Transform Infrared , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/growth & development , Structure-Activity Relationship , Thermodynamics , Trichosporon/drug effects , Trichosporon/growth & development , X-Ray Diffraction
11.
Protein Pept Lett ; 19(6): 652-6, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21838696

ABSTRACT

In a previous study, we reported that truncation of HP (2-20) (derived from the N-terminal region of Helicobacter pylori Ribosomal Protein L1 (RPL1)) at the N- (residues 2-3) and C-terminal (residues 17-20) truncated fragments to give HP (4-16) induces increased antibiotic activity against several bacterial strains without hemolysis. In this study, to develop novel short antibiotic peptides useful as therapeutic drugs, an analogue was designed to possess increased hydrophobicity by Trp substitution in position 2 region of HP (4-16). Synthetic HP (4-16)-W showed an enhanced antimicrobial and antitumor activity. The antimicrobial activity of this peptide and others was measured by their growth inhibitory effect upon S. aureus, B. subtilis, S. epidermidis, E. coli, S. typimurium, P. aeruginosa, C. albicans, T. beigelii and S. cerevisiae. None of the peptides exhibited hemolytic activity against human erythrocyte cells except melittin as a positive control. Its antibiotic activity suggests that HP (4-16)-W is an excellent candidate as a lead compound for the development of novel antibiotic agents.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Ribosomal Proteins/chemistry , Ribosomal Proteins/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Bacteria/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Discovery , Erythrocytes , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Hemolysis/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Melitten/pharmacology , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Engineering/methods , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Tryptophan/genetics , Tryptophan/metabolism , Yeasts/drug effects
12.
Org Biomol Chem ; 10(6): 1285-99, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22179201

ABSTRACT

Trichogin GA IV, isolated from the fungus Trichoderma longibrachiatum, is the prototype of lipopeptaibols, the sub-class of short-length peptaibiotics exhibiting membrane-modifying properties. This peptaibol is predominantly folded in a mixed 3(10)-/α- helical conformation with a clear, albeit modest, amphiphilic character, which is likely to be responsible for its capability to perturb bacterial membranes and to induce cell death. In previous papers, we reported on the interesting biological properties of trichogin GA IV, namely its good activity against Gram positive bacteria, in particular methicillin-resistant S. aureus strains, its stability towards proteolytic degradation, and its low hemolytic activity. Aiming at broadening the antimicrobial activity spectrum by increasing the peptide helical amphiphilicity, in this work we synthesized, by solution and solid-phase methodologies, purified and fully characterized a set of trichogin GA IV analogs in which the four Gly residues at positions 2, 5, 6, 9, lying in the poorly hydrophilic face of the helical structure, are substituted by one (position 2, 5, 6 or 9), two (positions 5 and 6), three (positions 2, 5, and 9), and four (positions 2, 5, 6, and 9) Lys residues. The conformational preferences of the Lys-containing analogs were assessed by FT-IR absorption, CD and 2D-NMR techniques in aqueous, organic, and membrane-mimetic environments. Interestingly, it turns out that the presence of charged residues induces a transition of the helical conformation adopted by the peptaibols (from 3(10)- to α-helix) as a function of pH in a reversible process. The role played in the analogs by the markedly increased amphiphilicity was further tested by fluorescence leakage experiments in model membranes, protease resistance, antibacterial and antifungal activities, cytotoxicity, and hemolysis. Taken together, our biological results provide evidence that some of the least substituted among these analogs are good candidates for the development of new membrane-active antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Lipopeptides/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Fungi/drug effects , Humans , Lipopeptides/chemistry , Lipopeptides/pharmacology , Microbial Sensitivity Tests , Structure-Activity Relationship
13.
Int J Mol Sci ; 12(9): 5971-92, 2011.
Article in English | MEDLINE | ID: mdl-22016639

ABSTRACT

Over the last decade, decreasing effectiveness of conventional antimicrobial-drugs has caused serious problems due to the rapid emergence of multidrug-resistant pathogens. Furthermore, biofilms, which are microbial communities that cause serious chronic infections and dental plaque, form environments that enhance antimicrobial resistance. As a result, there is a continuous search to overcome or control such problems, which has resulted in antimicrobial peptides being considered as an alternative to conventional drugs. Antimicrobial peptides are ancient host defense effector molecules in living organisms. These peptides have been identified in diverse organisms and synthetically developed by using peptidomimic techniques. This review was conducted to demonstrate the mode of action by which antimicrobial peptides combat multidrug-resistant bacteria and prevent biofilm formation and to introduce clinical uses of these compounds for chronic disease, medical devices, and oral health. In addition, combinations of antimicrobial peptides and conventional drugs were considered due to their synergetic effects and low cost for therapeutic treatment.


Subject(s)
Antimicrobial Cationic Peptides/therapeutic use , Bacteria/drug effects , Bacterial Infections/drug therapy , Biofilms/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Antimicrobial Cationic Peptides/pharmacology , Bacterial Infections/microbiology , Bacterial Physiological Phenomena/drug effects , Humans , Microbial Sensitivity Tests , Models, Biological
14.
J Pept Sci ; 17(12): 812-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956793

ABSTRACT

pVEC is a cell-penetrating peptide derived from the murine vascular endothelial-cadherin protein. To evaluate the potential of pVEC as antimicrobial peptide (AMP), we synthesized pVEC and its analogs with Trp and Arg/Lys substitution, and their antimicrobial and lipopolysaccharide (LPS)-neutralizing activities were investigated. pVEC and its analogs displayed a potent antimicrobial activity (minimal inhibitory concentration: 4-16 µM) against Gram-positive and Gram-negative bacteria but no or less hemolytic activity (less than 10% hemolysis) even at a concentration of 200 µM. These peptides induced a near-complete membrane depolarization (more than 80%) at 4 µM against Staphylococcus aureus and a significant dye leakage (35-70%) from bacterial membrane-mimicking liposome at a concentration as low as 1 µM. The fluorescence profiles of pVEC and its analogs in dye leakage from liposome and membrane depolarization were similar to those of a frog-derived AMP, magainin 2. These results suggest that pVEC and its analogs kill bacteria by forming a pore or ion channel in the cytoplasmic membrane. pVEC and its analogs significantly inhibited nitric oxide production or tumor necrosis factor-α release in LPS-stimulated mouse macrophage RAW264.7 cells at 10 to 50 µM, in which RAW264.7 were not damaged. Taken together, our results suggest that pVEC and its analogs with potent antimicrobial and LPS-neutralizing activities can serve as AMPs for the treatment of microbial infection and sepsis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell-Penetrating Peptides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemical synthesis , Cell Line , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemical synthesis , Hemolytic Agents , Macrophages/drug effects , Macrophages/metabolism , Mice , Microbial Sensitivity Tests , Molecular Sequence Data , Nitric Oxide/metabolism , Peptides/chemical synthesis , Permeability , Staphylococcus aureus/drug effects , Tumor Necrosis Factor-alpha/metabolism
15.
J Pept Sci ; 17(5): 329-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21462284

ABSTRACT

A class of cationic antimicrobial peptides involved in host defense consists of sequences rich in Lys and Trp. Small peptides, (WK)(3) and (KW)(3) , were designed by the combination of alternating Lys (K) and Trp (W) amino acids, and then their antimicrobial and hemolytic activities were determined. It was noticed that the reversed sequence of (KW)(3) showed more activity against all strains than did (WK)(3) . The non-hemolytic behavior of (WK)(3) is identical to that of the reversed analog of (KW)(3) . CD spectra revealed that these peptides had an unfolded structure in buffer and EYPC:CH (10:1, w/w), but adopted folded conformation in the presence of EYPE:EYPG (7:3, w/w). The reversed-(KW)(3) peptide caused a higher extent of calcein release from EYPE:EYPG (7:3, w/w), though the activity was higher than that of the (WK)(3) . The interaction of the peptides with model lipid vesicles was examined using Trp fluorescence. The reversed-(KW)(3) showed higher interaction with EYPE:EYPG (7:3, w/w) membrane than did (WK)(3) . Both the peptides show less affinities while binding to EYPC:CH (10:1, w/w). This clearly indicated that the reversal of sequence factors is relevant to increased antimicrobial activity and lipid membrane permeability.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Amino Acid Sequence , Anti-Infective Agents/adverse effects , Anti-Infective Agents/chemical synthesis , Antimicrobial Cationic Peptides/adverse effects , Antimicrobial Cationic Peptides/chemical synthesis , Bacteria/drug effects , Circular Dichroism , Fluoresceins/chemistry , Hemolysis/drug effects , Humans , Lysine/chemistry , Microbial Sensitivity Tests , Tryptophan/chemistry
16.
J Pept Sci ; 17(8): 585-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21495119

ABSTRACT

The medium-length peptaibiotics are characterized by a primary structure of 14-16 amino acid residues. Despite the interesting antibiotic and antifungal properties exhibited by these membrane-active peptides, their exact mechanism of action is still unknown. Here, we present our results on heptaibin, a 14-amino acid peptaibiotic found to exhibit antimicrobial activity against Staphylococcus aureus. We carried out the very challenging synthesis of heptaibin on solid phase and a detailed conformational analysis in solution. The peptaibiotic is folded in a mixed 310-/α-helix conformation which exhibits a remarkable amphiphilic character. We also find that it is highly stable toward degradation by proteolytic enzymes and nonhemolytic. Finally, fluorescence leakage experiments using small unilamellar vesicles of three different compositions revealed that heptaibin, although uncharged, is a selective compound for permeabilization of model membranes mimicking the overall negatively charged surface of Gram-positive bacteria. This latter finding is in agreement with the originally published antimicrobial activity data.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptide Hydrolases/metabolism , Anti-Bacterial Agents/chemistry , Cell Membrane/drug effects , Cell Membrane Permeability/drug effects , Enzyme Stability/drug effects , Erythrocytes/drug effects , HEK293 Cells , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Oligopeptides/chemistry , Staphylococcus aureus/drug effects , Structure-Activity Relationship
17.
18.
J Pept Sci ; 17(5): 353-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21465628

ABSTRACT

Recently, the rapid emergence of microbial pathogens which are resistant to currently available antibiotics has triggered considerable interest searching for naturally occuring antimicrobial peptides (AMPs). Because AMPs from food organisms are comparatively nontoxic, a number of them are used as sources, purified in new antibiotics. Herein, an antibacterial peptide (heat-stable KPS-1) was isolated from Korean pen shell (Atrina pectinata) by the following procedures: solvent-extraction, heating, ultrafiltration, and RP-HPLC. The molecular weight of KPS-1 (4549.1 Da) was revealed by MALDI-TOF/MS analysis. Interestingly, KPS-1 inhibited in vitro growth of Gram-negative bacteria, including Escherichia coli, E. coli O157, Pseudomonas aeruginosa, Enterobacter sakazakii, and Salmonella typhimurium, at pH 5.2, rather than at pH 7.2. Its minimal inhibitory concentrations (MICs) were ranged from 20 to 80 µg/ml; however, it was not effective against human red blood cells at a concentration of 500 µg/ml. This suggests that this peptide is useful as a clinical agent for some human organs in an acidic environment.


Subject(s)
Anti-Infective Agents/pharmacology , Peptides/pharmacology , Anti-Infective Agents/chemistry , Cronobacter sakazakii/drug effects , Electrophoresis, Polyacrylamide Gel , Hemolysis/drug effects , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Peptides/chemistry , Pseudomonas aeruginosa/drug effects , Salmonella typhimurium/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Ultrafiltration
19.
J Pept Sci ; 17(5): 335-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21294230

ABSTRACT

Cell-penetrating peptides (CPPs) are cationic oligopeptides able to translocate across biological membranes without perturbing them, while antimicrobial peptides (AMPs) kill bacteria mainly by disrupting their membranes. The two peptide classes share several characteristics (charge, amphipathicity, helicity, and length), and therefore the molecular properties discriminating between the two different bioactivities are not clear. Pep-1-K (KKTWWKTWWTKWSQPKKKRKV) is a new AMP derived from the widely studied CPP Pep-1 (KETWWETWWTEWSQPKKKRKV), or 'Chariot', known for its ability to carry large cargoes across biological membranes. Pep-1-K was obtained from Pep-1 by substituting the three Glu residues with Lys, to increase its cationic character. Previous studies showed that these modifications endow Pep-1-K with a potent antimicrobial activity, with MICs in the low micromolar range. Here, we characterized the interaction of Pep-1 and Pep-1-K with model membranes to understand the reason for the antimicrobial activity of Pep-1-K. The data show that this peptide causes vesicle aggregation, perturbs membrane order, and induces the leakage of ions, but not of larger solutes, while these effects were not observed for Pep-1. These differences are likely due, at least in part, to the higher affinity of Pep-1-K toward anionic bilayers, which mimick the composition of bacterial membranes.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Cell Membrane/chemistry , Cell-Penetrating Peptides/chemistry , Lipid Bilayers/chemistry , Liposomes/chemistry , Microscopy, Confocal , Spectrometry, Fluorescence
20.
Eur Biophys J ; 40(4): 503-14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21222117

ABSTRACT

The effect of acyl chain structure and bilayer phase state on binding and penetration by the peptide HPA3 was studied using dual polarisation interferometry. This peptide is an analogue of Hp(2-20) derived from the N-terminus of Helicobacter pylori ribosomal protein L1 (RpL1) which has been shown to have antimicrobial and cell-penetrating properties. The binding of HPA3 to zwitterionic 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) or 1-palmitolyl-2-oleyl-sn-glycero-3-phosphocholine (POPC) and negatively charged membranes composed of DMPC and 1,2-dimyristoyl-sn-glycero-3-(phosphor-rac-(1-glycerol)) (DMPG) or POPC and 1-palmitolyl-2-oleyl-sn-glycero-3-(phosphor-rac-(1-glycerol)) (POPG) was determined using dual polarisation interferometry (DPI). Mass and birefringence were measured in real time, enabling the creation of birefringence-mass plots for detailed analysis of the changes in lipid bilayer order during the peptide-binding process. HPA3 bound to all four lipids and the binding progressed as a single phase for the saturated gel phase bilayers DMPC and DMPC-DMPG. However, the binding process involved two or more phases, with penetration of the unsaturated fluid phase POPC and POPC-POPG bilayers. Structural changes in the saturated bilayer were partially reversible whereas binding to the unsaturated bilayer resulted in irreversible changes in membrane structure. These results demonstrate that more disordered unsaturated bilayers are more susceptible to further disorganisation and have a lower capacity to recover from peptide-induced structural changes than saturated ordered bilayers. In addition, this study further establishes DPI as powerful tool for analysis of multiphase peptide-insertion processes associated with complex structural changes in the liquid-crystalline membrane.


Subject(s)
Acyl Carrier Protein/pharmacology , Anti-Infective Agents/pharmacology , Lipid Bilayers/metabolism , Peptide Fragments/pharmacology , Ribosomal Proteins/pharmacology , Acyl Carrier Protein/chemistry , Anti-Infective Agents/chemistry , Binding Sites/drug effects , Circular Dichroism/methods , Dimyristoylphosphatidylcholine/chemistry , Dimyristoylphosphatidylcholine/metabolism , Interferometry/methods , Lipid Bilayers/chemistry , Peptide Fragments/chemistry , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylglycerols/chemistry , Phosphatidylglycerols/metabolism , Ribosomal Proteins/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...