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.
Int J Mol Sci ; 21(18)2020 Sep 13.
Article in English | MEDLINE | ID: mdl-32933215

ABSTRACT

Amphibian skin is a promising natural resource for antimicrobial peptides (AMPs), key effectors of innate immunity with attractive therapeutic potential to fight antibiotic-resistant pathogens. Our previous studies showed that the skin of the Sahara Frog (Pelophylax saharicus) contains broad-spectrum AMPs of the temporin family, named temporins-SH. Here, we focused our study on temporin-SHe, a temporin-SHd paralog that we have previously identified in this frog but was never structurally and functionally characterized. We synthesized and determined the structure of temporin-SHe. This non-amphipathic α-helical peptide was demonstrated to strongly destabilize the lipid chain packing of anionic multilamellar vesicles mimicking bacterial membranes. Investigation of the antimicrobial activity revealed that temporin-SHe targets Gram-negative and Gram-positive bacteria, including clinical isolates of multi-resistant Staphylococcus aureus strains. Temporin-SHe exhibited also antiparasitic activity toward different Leishmania species responsible for visceral leishmaniasis, as well as cutaneous and mucocutaneous forms. Functional assays revealed that temporin-SHe exerts bactericidal effects with membrane depolarization and permeabilization, via a membranolytic mechanism observed by scanning electron microscopy. Temporin-SHe represents a new member of the very limited group of antiparasitic temporins/AMPs. Despite its cytotoxicity, it is nevertheless an interesting tool to study the AMP antiparasitic mechanism and design new antibacterial/antiparasitic agents.


Subject(s)
Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Anura/metabolism , Leishmania/metabolism , Africa, Northern , Amino Acid Sequence , Amphibian Proteins/metabolism , Amphibian Proteins/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antiparasitic Agents/metabolism , Antiparasitic Agents/pharmacology , Bacteria/drug effects , Cell Line, Tumor , Humans , Protein Conformation, alpha-Helical/physiology , Skin/metabolism , THP-1 Cells
2.
PLoS One ; 12(3): e0174024, 2017.
Article in English | MEDLINE | ID: mdl-28319176

ABSTRACT

Antimicrobial peptides (AMPs) are promising drugs to kill resistant pathogens. In contrast to bacteria, protozoan parasites, such as Leishmania, were little studied. Therefore, the antiparasitic mechanism of AMPs is still unclear. In this study, we sought to get further insight into this mechanism by focusing our attention on temporin-SHa (SHa), a small broad-spectrum AMP previously shown to be active against Leishmania infantum. To improve activity, we designed analogs of SHa and compared the antibacterial and antiparasitic mechanisms. [K3]SHa emerged as a highly potent compound active against a wide range of bacteria, yeasts/fungi, and trypanosomatids (Leishmania and Trypanosoma), with leishmanicidal intramacrophagic activity and efficiency toward antibiotic-resistant strains of S. aureus and antimony-resistant L. infantum. Multipassage resistance selection demonstrated that temporins-SH, particularly [K3]SHa, are not prone to induce resistance in Escherichia coli. Analysis of the mode of action revealed that bacterial and parasite killing occur through a similar membranolytic mechanism involving rapid membrane permeabilization and depolarization. This was confirmed by high-resolution imaging (atomic force microscopy and field emission gun-scanning electron microscopy). Multiple combined techniques (nuclear magnetic resonance, surface plasmon resonance, differential scanning calorimetry) allowed us to detail peptide-membrane interactions. [K3]SHa was shown to interact selectively with anionic model membranes with a 4-fold higher affinity (KD = 3 x 10-8 M) than SHa. The amphipathic α-helical peptide inserts in-plane in the hydrophobic lipid bilayer and disrupts the acyl chain packing via a detergent-like effect. Interestingly, cellular events, such as mitochondrial membrane depolarization or DNA fragmentation, were observed in L. infantum promastigotes after exposure to SHa and [K3]SHa at concentrations above IC50. Our results indicate that these temporins exert leishmanicidal activity via a primary membranolytic mechanism but can also trigger apoptotis-like death. The many assets demonstrated for [K3]SHa make this small analog an attractive template to develop new antibacterial/antiparasitic drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antiprotozoal Agents/pharmacology , Ampicillin/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacokinetics , Antimicrobial Cationic Peptides/toxicity , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/toxicity , Apoptosis/drug effects , Bacteria/drug effects , Cell Line , Cell Membrane Permeability/drug effects , DNA Fragmentation/drug effects , DNA, Protozoan/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Bacterial , Humans , Leishmania/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potentials/drug effects , Time Factors , Trypanosoma/drug effects , Unilamellar Liposomes/chemistry
3.
J Pept Sci ; 20(7): 563-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24919960

ABSTRACT

Development of resistant bacteria onto biomaterials is a major problem leading to nosocomial infections. Antimicrobial peptides are good candidates for the generation of antimicrobial surfaces because of their broad-spectrum activity and their original mechanism of action (i.e. rapid lysis of the bacterial membrane) making them less susceptible to the development of bacterial resistance. In this study, we report on the covalent immobilisation of temporin-SHa on a gold surface modified by a thiolated self-assembled monolayer. Temporin-SHa (FLSGIVGMLGKLF amide) is a small hydrophobic and low cationic antimicrobial peptide with potent and very broad-spectrum activity against Gram-positive and Gram-negative bacteria, yeasts and parasites. We have analysed the influence of the binding mode of temporin-SHa on the antibacterial efficiency by using a covalent binding either via the peptide NH2 groups (random grafting of α- and ε-NH2 to the surface) or via its C-terminal end (oriented grafting using the analogue temporin-SHa-COOH). The surface functionalization was characterised by IR spectroscopy (polarisation modulation reflection absorption IR spectroscopy) while antibacterial activity against Listeria ivanovii was assessed by microscopy techniques, such as atomic force microscopy and scanning electron microscopy equipped with a field emission gun. Our results revealed that temporin-SHa retains its antimicrobial activity after covalent grafting. A higher amount of bound temporin-SHa is observed for the C-terminally oriented grafting compared with the random grafting (NH2 groups). Temporin-SHa therefore represents an attractive candidate as antimicrobial coating agent.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Gold/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Listeria/drug effects , Microbial Sensitivity Tests , Microscopy, Atomic Force , Microscopy, Electron, Scanning , Surface Properties
4.
PLoS One ; 8(8): e70782, 2013.
Article in English | MEDLINE | ID: mdl-23967105

ABSTRACT

Transcriptomic and peptidomic analysis of skin secretions from the Painted-belly leaf frog Phyllomedusa sauvagii led to the identification of 5 novel phylloseptins (PLS-S2 to -S6) and also of phylloseptin-1 (PSN-1, here renamed PLS-S1), the only member of this family previously isolated in this frog. Synthesis and characterization of these phylloseptins revealed differences in their antimicrobial activities. PLS-S1, -S2, and -S4 (79-95% amino acid sequence identity; net charge  = +2) were highly potent and cidal against Gram-positive bacteria, including multidrug resistant S. aureus strains, and killed the promastigote stage of Leishmania infantum, L. braziliensis and L. major. By contrast, PLS-S3 (95% amino acid identity with PLS-S2; net charge  = +1) and -S5 (net charge  = +2) were found to be almost inactive against bacteria and protozoa. PLS-S6 was not studied as this peptide was closely related to PLS-S1. Differential scanning calorimetry on anionic and zwitterionic multilamellar vesicles combined with circular dichroism spectroscopy and membrane permeabilization assays on bacterial cells indicated that PLS-S1, -S2, and -S4 are structured in an amphipathic α-helix that disrupts the acyl chain packing of anionic lipid bilayers. As a result, regions of two coexisting phases could be formed, one phase rich in peptide and the other lipid-rich. After reaching a threshold peptide concentration, the disruption of lipid packing within the bilayer may lead to local cracks and disintegration of the microbial membrane. Differences in the net charge, α-helical folding propensity, and/or degree of amphipathicity between PLS-S1, -S2 and -S4, and between PLS-S3 and -S5 appear to be responsible for their marked differences in their antimicrobial activities. In addition to the detailed characterization of novel phylloseptins from P. sauvagii, our study provides additional data on the previously isolated PLS-S1 and on the mechanism of action of phylloseptins.


Subject(s)
Amphibian Proteins/metabolism , Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Anura/metabolism , Membranes/metabolism , Amino Acid Sequence , Amphibian Proteins/chemistry , Amphibian Proteins/genetics , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Base Sequence , Cell Membrane Permeability , Cloning, Molecular , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Inhibitory Concentration 50 , Kinetics , Leishmania infantum/drug effects , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Skin/metabolism , Staphylococcus aureus/drug effects
5.
Biochimie ; 95(2): 388-99, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23116712

ABSTRACT

Temporins are a family of short antimicrobial peptides (8-17 residues) that mostly show potent activity against Gram-positive bacteria. Herein, we demonstrate that temporin-SHd, a 17-residue peptide with a net charge of +2 (FLPAALAGIGGILGKLF(amide)), expressed a broad spectrum of antimicrobial activity. This peptide displayed potent antibacterial activities against Gram-negative and Gram-positive bacteria, including multi-drug resistant Staphylococcus aureus strains, as well as antiparasitic activity against promastigote and the intracellular stage (amastigote) of Leishmania infantum, at concentration not toxic for the macrophages. Temporin-SHd that is structured in a non-amphipathic α-helix in anionic membrane-mimetic environments, strongly and selectively perturbs anionic bilayer membranes by interacting with the polar head groups and acyl region of the phospholipids, with formation of regions of two coexisting phases: one phase rich in peptide and the other lipid-rich. The disruption of lipid packing within the bilayer may lead to the formation of transient pores and membrane permeation/disruption once a threshold peptide accumulation is reached. To our knowledge, Temporin-SHd represents the first known 17-residue long temporin expressing such broad spectrum of antimicrobial activity including members of the trypanosomatidae family. Additionally, since only a few shorter members (13 residues) of the temporin family are known to display antileishmanial activity (temporins-TA, -TB and -SHa), SHd is an interesting tool to analyze the antiparasitic mechanism of action of temporins.


Subject(s)
Amphibian Proteins/pharmacology , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Leishmania infantum/drug effects , Proteins/pharmacology , Ranidae/metabolism , Trypanosoma/drug effects , Amino Acid Sequence , Amphibian Proteins/isolation & purification , Animals , Anti-Infective Agents/isolation & purification , Antimicrobial Cationic Peptides/isolation & purification , Cell Line , Circular Dichroism , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/growth & development , Humans , Inhibitory Concentration 50 , Leishmania infantum/growth & development , Lipid Bilayers/chemistry , Macrophages/drug effects , Macrophages/parasitology , Molecular Sequence Data , Phospholipids/chemistry , Proteins/isolation & purification , Skin/metabolism , Solid-Phase Synthesis Techniques , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypanosoma/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL