An amphipathic peptide with antibiotic activity against multidrug-resistant Gram-negative bacteria

Autor: Mark E. Cooper, Alysha G. Elliott, Amy K. Cain, Ingrid A. Edwards, Mehdi Mobli, Carina Vingsbo Lundberg, Johnny X. Huang, Sergio Lociuro, Søren Neve, Magnus Strandh, Mark A. T. Blaskovich, Jason A. Steen, Kaela M. Porter, Mark S. Butler, Lars Barquist, Christine J. Boinett, Johannes Zuegg
Rok vydání: 2020
Předmět:
Male
0301 basic medicine
Carbapenem
Cell Membrane Permeability
Antibiotics
General Physics and Astronomy
Peptide
Drug resistance
Mice
Drug Resistance
Multiple
Bacterial

lcsh:Science
chemistry.chemical_classification
Multidisciplinary
biology
Drug discovery
Helminth Proteins
Haemolysis
Anti-Bacterial Agents
3. Good health
Urinary Tract Infections
Female
medicine.drug
Membrane permeability
medicine.drug_class
Science
030106 microbiology
Microbial Sensitivity Tests
Peritonitis
Article
General Biochemistry
Genetics and Molecular Biology

Microbiology
03 medical and health sciences
Gram-Negative Bacteria
medicine
Animals
Humans
Colistin
Pneumonia
General Chemistry
Phospholipid transport
biology.organism_classification
Disease Models
Animal

030104 developmental biology
Carbapenems
chemistry
lcsh:Q
Bacteria
Antimicrobial Cationic Peptides
Zdroj: Nature Communications, Vol 11, Iss 1, Pp 1-13 (2020)
Nature Communications
Nature communications
United Kingdom
England
ISSN: 2041-1723
Popis: Peptide antibiotics are an abundant and synthetically tractable source of molecular diversity, but they are often cationic and can be cytotoxic, nephrotoxic and/or ototoxic, which has limited their clinical development. Here we report structure-guided optimization of an amphipathic peptide, arenicin-3, originally isolated from the marine lugworm Arenicola marina. The peptide induces bacterial membrane permeability and ATP release, with serial passaging resulting in a mutation in mlaC, a phospholipid transport gene. Structure-based design led to AA139, an antibiotic with broad-spectrum in vitro activity against multidrug-resistant and extensively drug-resistant bacteria, including ESBL, carbapenem- and colistin-resistant clinical isolates. The antibiotic induces a 3–4 log reduction in bacterial burden in mouse models of peritonitis, pneumonia and urinary tract infection. Cytotoxicity and haemolysis of the progenitor peptide is ameliorated with AA139, and the ‘no observable adverse effect level’ (NOAEL) dose in mice is ~10-fold greater than the dose generally required for efficacy in the infection models.
Peptide antibiotics often display a very narrow therapeutic index. Here, the authors present an optimized peptide antibiotic with broad-spectrum in vitro activities, in vivo efficacy in multiple disease models against multidrug-resistant Gram-negative infections, and reduced toxicity.
Databáze: OpenAIRE