Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-33106269

ABSTRACT

Lugdunin is the first reported nonribosomally synthesized antibiotic from human microbiomes. Its production by the commensal Staphylococcus lugdunensis eliminates the pathogen Staphylococcus aureus from human nasal microbiomes. The cycloheptapeptide lugdunin is the founding member of the new class of fibupeptide antibiotics, which have a novel mode of action and represent promising new antimicrobial agents. How S. lugdunensis releases and achieves producer self-resistance to lugdunin has remained unknown. We report that two ABC transporters encoded upstream of the lugdunin-biosynthetic operon have distinct yet overlapping roles in lugdunin secretion and self-resistance. While deletion of the lugEF transporter genes abrogated most of the lugdunin secretion, the lugGH transporter genes had a dominant role in resistance. Yet all four genes were required for full-level lugdunin resistance. The small accessory putative membrane protein LugI further contributed to lugdunin release and resistance levels conferred by the ABC transporters. Whereas LugIEFGH also conferred resistance to lugdunin congeners with inverse structures or with amino acid exchange at position 6, they neither affected the susceptibility to a lugdunin variant with an exchange at position 2 nor to other cyclic peptide antimicrobials such as daptomycin or gramicidin S. The obvious selectivity of the resistance mechanism raises hopes that it will not confer cross-resistance to other antimicrobials or to optimized lugdunin derivatives to be used for the prevention and treatment of S. aureus infections.


Subject(s)
Anti-Infective Agents , Staphylococcal Infections , Staphylococcus lugdunensis , ATP-Binding Cassette Transporters/genetics , Anti-Bacterial Agents/pharmacology , Humans , Microbial Sensitivity Tests , Peptides, Cyclic/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus , Thiazolidines
2.
ACS Chem Biol ; 14(9): 1972-1980, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31419109

ABSTRACT

Pyrones comprise a structurally diverse class of compounds. Although they are widespread in nature, their specific physiological functions remain unknown in most cases. We recently described that triketide pyrones mediate the sulfotransfer in caprazamycin biosynthesis. Herein, we report the identification of conexipyrones A-C, three previously unrecognized tetra-substituted α-pyrones, from the soil actinobacterium Conexibacter woesei. Insights into their biosynthesis via a type III polyketide synthase were obtained by feeding studies using isotope-enriched precursors. In vitro assays employing the genetically associated 3'-phosphoadenosine-5'-phosphosulfate (PAPS)-dependent sulfotransferase CwoeST revealed conexipyrones as the enzymes' genuine sulfate acceptor substrates. Furthermore, conexipyrones were determined to function as sulfate shuttles in a two-enzyme assay, because their sulfated derivatives were accepted as donor molecules by the PAPS-independent arylsulfate sulfotransferase (ASST) Cpz4 to yield sulfated caprazamycin intermediates.


Subject(s)
Actinobacteria/chemistry , Pyrones/metabolism , Sulfuric Acid Esters/metabolism , Arylsulfotransferase/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Polyketide Synthases/genetics , Pyrones/isolation & purification , Streptomyces coelicolor/genetics
3.
Nat Commun ; 10(1): 2730, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227691

ABSTRACT

Recently our groups discovered lugdunin, a new cyclic peptide antibiotic that inhibits Staphylococcus aureus epithelial colonization in humans and rodents. In this work, we analyzed its immuno-modulatory and antimicrobial potential as a single agent or in combination with other microbiota- or host-derived factors. We show that pretreatment of primary human keratinocytes or mouse skin with lugdunin in combination with microbiota-derived factors results in a significant reduction of S. aureus colonization. Moreover, lugdunin increases expression and release of LL-37 and CXCL8/MIP-2 in human keratinocytes and mouse skin, and results in the recruitment of monocytes and neutrophils in vivo, both by a TLR/MyD88-dependent mechanism. Interestingly, S. aureus elimination by lugdunin is additionally achieved by synergistic antimicrobial activity with LL-37 and dermcidin-derived peptides. In summary, our results indicate that lugdunin provides multi-level protection against S. aureus and may thus become a promising treatment option for S. aureus skin infections in the future.


Subject(s)
Anti-Bacterial Agents/pharmacology , Immunity, Innate/drug effects , Peptides, Cyclic/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Thiazolidines/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Microbiota/immunology , Peptides/immunology , Peptides, Cyclic/therapeutic use , Primary Cell Culture , Skin/drug effects , Skin/immunology , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Thiazolidines/therapeutic use , Cathelicidins
4.
Angew Chem Int Ed Engl ; 58(27): 9234-9238, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31059155

ABSTRACT

Lugdunin, a novel thiazolidine cyclopeptide, exhibits micromolar activity against methicillin-resistant Staphylococcus aureus (MRSA). For structure-activity relationship (SAR) studies, synthetic analogues obtained from alanine and stereo scanning as well as peptides with modified thiazolidine rings were tested for antimicrobial activity. The thiazolidine ring and the alternating d- and l-amino acid backbone are essential. Notably, the non-natural enantiomer displays equal activity, thus indicating the absence of a chiral target. The antibacterial activity strongly correlates with dissipation of the membrane potential in S. aureus. Lugdunin equalizes pH gradients in artificial membrane vesicles, thereby maintaining membrane integrity, which demonstrates that proton translocation is the mode of action (MoA). The incorporation of extra tryptophan or propargyl moieties further expands the diversity of this class of thiazolidine cyclopeptides.


Subject(s)
Anti-Infective Agents/chemical synthesis , Peptides, Cyclic/chemistry , Thiazolidines/chemistry , Alanine/chemistry , Amino Acid Sequence , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protons , Stereoisomerism , Structure-Activity Relationship , Thiazolidines/chemical synthesis , Thiazolidines/pharmacology
6.
Nature ; 535(7613): 511-6, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27466123

ABSTRACT

The vast majority of systemic bacterial infections are caused by facultative, often antibiotic-resistant, pathogens colonizing human body surfaces. Nasal carriage of Staphylococcus aureus predisposes to invasive infection, but the mechanisms that permit or interfere with pathogen colonization are largely unknown. Whereas soil microbes are known to compete by production of antibiotics, such processes have rarely been reported for human microbiota. We show that nasal Staphylococcus lugdunensis strains produce lugdunin, a novel thiazolidine-containing cyclic peptide antibiotic that prohibits colonization by S. aureus, and a rare example of a non-ribosomally synthesized bioactive compound from human-associated bacteria. Lugdunin is bactericidal against major pathogens, effective in animal models, and not prone to causing development of resistance in S. aureus. Notably, human nasal colonization by S. lugdunensis was associated with a significantly reduced S. aureus carriage rate, suggesting that lugdunin or lugdunin-producing commensal bacteria could be valuable for preventing staphylococcal infections. Moreover, human microbiota should be considered as a source for new antibiotics.


Subject(s)
Anti-Bacterial Agents/metabolism , Peptides, Cyclic/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Staphylococcus lugdunensis/metabolism , Symbiosis , Thiazolidines/metabolism , Animals , Anti-Bacterial Agents/biosynthesis , Carrier State/microbiology , Disease Models, Animal , Drug Resistance, Microbial , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbiota/physiology , Nose/microbiology , Sigmodontinae , Staphylococcal Infections/drug therapy , Staphylococcus aureus/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...