Structural Basis for Xenosiderophore Utilization by the Human Pathogen Staphylococcus aureus

Staphylococcus aureus produces a cocktail of metallophores (staphylopine, staphyloferrin A, and staphyloferrin B) to scavenge transition metals during infection of a host. In addition, S. aureus displays the extracellular surface lipoproteins FhuD1 and FhuD2 along with the ABC transporter complex Fh...

Full description

Saved in:
Bibliographic Details
Published inACS infectious diseases Vol. 3; no. 7; pp. 542 - 553
Main Authors Endicott, Nathaniel P, Lee, Eries, Wencewicz, Timothy A
Format Journal Article
LanguageEnglish
Published United States American Chemical Society 14.07.2017
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Staphylococcus aureus produces a cocktail of metallophores (staphylopine, staphyloferrin A, and staphyloferrin B) to scavenge transition metals during infection of a host. In addition, S. aureus displays the extracellular surface lipoproteins FhuD1 and FhuD2 along with the ABC transporter complex FhuCBG to facilitate the use of hydroxamate xenosiderophores such as desferrioxamine B (DFOB) for iron acquisition. DFOB is used as a chelation therapy to treat human iron overload diseases and has been linked to an increased risk of S. aureus infections. We used a panel of synthetic DFOB analogs and a FhuD2-selective trihydroxamate sideromycin to probe xenosiderophore utilization in S. aureus and establish structure–activity relationships for Fe­(III) binding, FhuD2 binding, S. aureus growth promotion, and competition for S. aureus cell entry. Fe­(III) binding assays and FhuD2 intrinsic fluorescence quenching experiments revealed that diverse chemical modifications of the terminal ends of linear ferrioxamine siderophores influences Fe­(III) affinity but not FhuD2 binding. Siderophore–sideromycin competition assays and xenosiderophore growth promotion assays revealed that S. aureus SG511 and ATCC 11632 can distinguish between competing siderophores based exclusively on net charge of the siderophore–Fe­(III) complex. Our work provides a roadmap for tuning hydroxamate xenosiderophore scaffolds to suppress (net negative charge) or enhance (net positive or neutral charge) uptake by S. aureus for applications in metal chelation therapy and siderophore-mediated antibiotic delivery, respectively.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2373-8227
2373-8227
DOI:10.1021/acsinfecdis.7b00036