Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892200

ABSTRACT

The pyoverdine siderophore is produced by Pseudomonas aeruginosa to access iron. Its synthesis involves the complex coordination of four nonribosomal peptide synthetases (NRPSs), which are responsible for assembling the pyoverdine peptide backbone. The precise cellular organization of these NRPSs and their mechanisms of interaction remain unclear. Here, we used a combination of several single-molecule microscopy techniques to elucidate the spatial arrangement of NRPSs within pyoverdine-producing cells. Our findings reveal that PvdL differs from the three other NRPSs in terms of localization and mobility patterns. PvdL is predominantly located in the inner membrane, while the others also explore the cytoplasmic compartment. Leveraging the power of multicolor single-molecule localization, we further reveal co-localization between PvdL and the other NRPSs, suggesting a pivotal role for PvdL in orchestrating the intricate biosynthetic pathway. Our observations strongly indicates that PvdL serves as a central orchestrator in the assembly of NRPSs involved in pyoverdine biosynthesis, assuming a critical regulatory function.


Subject(s)
Oligopeptides , Peptide Synthases , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/enzymology , Oligopeptides/biosynthesis , Oligopeptides/metabolism , Peptide Synthases/metabolism , Peptide Synthases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Siderophores/biosynthesis , Siderophores/metabolism
2.
FEBS Lett ; 597(23): 2963-2974, 2023 12.
Article in English | MEDLINE | ID: mdl-37758521

ABSTRACT

Iron is an essential nutrient for the survival and virulence of Pseudomonas aeruginosa. The pathogen expresses at least 15 different iron-uptake pathways, the majority involving small iron chelators called siderophores. P. aeruginosa produces two siderophores, but can also use many produced by other microorganisms. This implies that the bacterium expresses appropriate TonB-dependent transporters (TBDTs) at the outer membrane to import the ferric form of each of the siderophores used. Here, we show that the two α-carboxylate-type siderophores rhizoferrin-Fe and staphyloferrin A-Fe are transported into P. aeruginosa cells by the TBDT ActA. Among the mixed α-carboxylate/hydroxamate-type siderophores, we found aerobactin-Fe to be transported by ChtA and schizokinen-Fe and arthrobactin-Fe by ChtA and another unidentified TBDT. Our findings enhance the understanding of the adaptability of P. aeruginosa and hold significant implications for developing novel strategies to combat antibiotic resistance.


Subject(s)
Pseudomonas aeruginosa , Siderophores , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Iron/metabolism
3.
J Med Chem ; 66(1): 553-576, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36548006

ABSTRACT

Rising infection rates with multidrug-resistant pathogens calls for antibiotics with novel modes of action. Herein, we identify the inner membrane protein TonB, a motor of active uptake in Gram-negative bacteria, as a novel target in antimicrobial therapy. The interaction of the TonB box of outer membrane transporters with TonB is crucial for the internalization of essential metabolites. We designed TonB box peptides and coupled them with synthetic siderophores in order to facilitate their uptake into bacteria in up to 32 synthetic steps. Three conjugates repressed the growth of Pseudomonas aeruginosa cells unable to produce their own siderophores, with minimal inhibitory concentrations between 0.1 and 0.5 µM. The transporters mediating uptake of these compounds were identified as PfeA and PirA. The study illustrates a variant of cellular suicide where a transporter imports its own inhibitor and demonstrates that artificial siderophores can import cargo with molecular weights up to 4 kDa.


Subject(s)
Anti-Bacterial Agents , Siderophores , Siderophores/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Pseudomonas aeruginosa/metabolism , Membrane Proteins/metabolism , Biological Transport , Membrane Transport Proteins/metabolism
4.
ACS Infect Dis ; 8(9): 1894-1904, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35881068

ABSTRACT

Enterobactin (ENT) is a tris-catechol siderophore used to acquire iron by multiple bacterial species. These ENT-dependent iron uptake systems have often been considered as potential gates in the bacterial envelope through which one can shuttle antibiotics (Trojan horse strategy). In practice, siderophore analogues containing catechol moieties have shown promise as vectors to which antibiotics may be attached. Bis- and tris-catechol vectors (BCVs and TCVs, respectively) were shown using structural biology and molecular modeling to mimic ENT binding to the outer membrane transporter PfeA in Pseudomonas aeruginosa. TCV but not BCV appears to cross the outer membrane via PfeA when linked to an antibiotic (linezolid). TCV is therefore a promising vector for Trojan horse strategies against P. aeruginosa, confirming the ENT-dependent iron uptake system as a gate to transport antibiotics into P. aeruginosa cells.


Subject(s)
Enterobactin , Oxazolidinones , Anti-Bacterial Agents/chemistry , Catechols/chemistry , Catechols/metabolism , Enterobactin/metabolism , Iron/metabolism , Membrane Transport Proteins/metabolism , Oxazolidinones/chemistry , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism
5.
mBio ; 13(4): e0149822, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35770947

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen responsible for acute and chronic infections in immunocompromised hosts. This organism is known to compete efficiently against coinfecting microorganisms, due in part to the secretion of antimicrobial molecules and the synthesis of siderophore molecules with high affinity for iron. P. aeruginosa possess a large repertoire of TonB-dependent transporters for the uptake of its own, as well as xenosiderophores released from other bacteria or fungi. Here, we show that P. aeruginosa is also capable of utilizing plant-derived polyphenols as an iron source. We found that exclusively plant-derived phenols containing a catechol group (i.e., chlorogenic acid, caffeic acid, quercetin, luteolin) induce the expression of the TonB-dependent transporters PiuA or PirA. This induction requires the two-component system PirR-PirS. Chlorogenic acid in its Fe(III)-loaded form was actively transported by PiuA and PirA and supported growth under iron-limiting conditions. Coincidentally, PiuA and PirA are also the main TonB transporters for the recently approved siderophore-drug conjugate cefiderocol. Surprisingly, quercetin supplementation increased the susceptibility of P. aeruginosa to siderophore-drug conjugates, due to induction of piuA and pirA expression mediated by the PirR-PirS two-component system. These findings suggest a potential novel therapeutic application for these biologically active dietary polyphenols. IMPORTANCE Iron is an essential element for living organisms. Most bacteria synthesize species-specific iron chelators, called siderophores, able to capture iron from their host or the environment. Pseudomonas aeruginosa, an opportunistic pathogen, produces two endogenous siderophores but is able to acquire iron also via xenosiderophores, produced by other bacteria or fungi, using a set of conserved TonB transporters. Here, we show that P. aeruginosa is also able to use plant metabolites, like quercetin and chlorogenic acid, as siderophores. These metabolites possess an iron-chelating catechol group and are recognized and transported by the TonB transporters PirA and PiuA. Since these transporters also promote the specific uptake of siderophore-drug conjugates, P. aeruginosa exposed to these plant catechols becomes hypersusceptible to this novel class of antibiotics. This unexpected finding suggests a potential therapeutic application for quercetin and chlorogenic acid, which were mainly investigated for their antioxidant and anti-inflammatory properties.


Subject(s)
Pseudomonas aeruginosa , Siderophores , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Catechols/metabolism , Catechols/pharmacology , Chlorogenic Acid/metabolism , Chlorogenic Acid/pharmacology , Ferric Compounds/metabolism , Iron/metabolism , Iron Chelating Agents/metabolism , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Quercetin/metabolism , Siderophores/metabolism
6.
ACS Infect Dis ; 8(6): 1134-1146, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35500104

ABSTRACT

The development of new antibiotics against Gram-negative bacteria has to deal with the low permeability of the outer membrane. This obstacle can be overcome by utilizing siderophore-dependent iron uptake pathways as entrance routes for antibiotic uptake. Iron-chelating siderophores are actively imported by bacteria, and their conjugation to antibiotics allows smuggling the latter into bacterial cells. Synthetic siderophore mimetics based on MECAM (1,3,5-N,N',N″-tris-(2,3-dihydroxybenzoyl)-triaminomethylbenzene) and DOTAM (1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane) cores, both chelating iron via catechol groups, have been recently applied as versatile carriers of functional cargo. In the present study, we show that MECAM and the MECAM-ampicillin conjugate 3 transport iron into Pseudomonas aeruginosa cells via the catechol-type outer membrane transporters PfeA and PirA and DOTAM solely via PirA. Differential proteomics and quantitative real-time polymerase chain reaction (qRT-PCR) showed that MECAM import induced the expression of pfeA, whereas 3 led to an increase in the expression of pfeA and ampc, a gene conferring ampicillin resistance. The presence of DOTAM did not induce the expression of pirA but upregulated the expression of two zinc transporters (cntO and PA0781), pointing out that bacteria become zinc starved in the presence of this compound. Iron uptake experiments with radioactive 55Fe demonstrated that import of this nutrient by MECAM and DOTAM was as efficient as with the natural siderophore enterobactin. The study provides a functional validation for DOTAM- and MECAM-based artificial siderophore mimetics as vehicles for the delivery of cargo into Gram-negative bacteria.


Subject(s)
Pseudomonas aeruginosa , Siderophores , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Benzamides , Catechols/metabolism , Catechols/pharmacology , Gram-Negative Bacteria/metabolism , Hydroxybenzoates , Iron/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Siderophores/pharmacology , Zinc/metabolism
7.
Environ Microbiol ; 24(2): 878-893, 2022 02.
Article in English | MEDLINE | ID: mdl-33350053

ABSTRACT

Iron is an essential nutrient for bacterial growth and the cause of a fierce battle between the pathogen and host during infection. Bacteria have developed several strategies to access iron from the host, the most common being the production of siderophores, small iron-chelating molecules secreted into the bacterial environment. The opportunist pathogen Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, and is also able to use a wide panoply of xenosiderophores, siderophores produced by other microorganisms. Here, we demonstrate that catecholamine neurotransmitters (dopamine, l-DOPA, epinephrine and norepinephrine) are able to chelate iron and efficiently bring iron into P. aeruginosa cells via TonB-dependent transporters (TBDTs). Bacterial growth assays under strong iron-restricted conditions and with numerous mutants showed that the TBDTs involved are PiuA and PirA. PiuA exhibited more pronounced specificity for dopamine uptake than for norepinephrine, epinephrine and l-DOPA, whereas PirA specificity appeared to be higher for l-DOPA and norepinephrine. Proteomic and qRT-PCR approaches showed pirA transcription and expression to be induced in the presence of all four catecholamines. Finally, the oxidative properties of catecholamines enable them to reduce iron, and we observed ferrous iron uptake via the FeoABC system in the presence of l-DOPA.


Subject(s)
Pseudomonas aeruginosa , Siderophores , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catecholamines/metabolism , Iron/metabolism , Neurotransmitter Agents/metabolism , Proteomics , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism
8.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33802163

ABSTRACT

Bacteria access iron, a key nutrient, by producing siderophores or using siderophores produced by other microorganisms. The pathogen Pseudomonas aeruginosa produces two siderophores but is also able to pirate enterobactin (ENT), the siderophore produced by Escherichia coli. ENT-Fe complexes are imported across the outer membrane of P. aeruginosa by the two outer membrane transporters PfeA and PirA. Iron is released from ENT in the P. aeruginosa periplasm by hydrolysis of ENT by the esterase PfeE. We show here that pfeE gene deletion renders P. aeruginosa unable to grow in the presence of ENT because it is unable to access iron via this siderophore. Two-species co-cultures under iron-restricted conditions show that P. aeruginosa strongly represses the growth of E. coli as long it is able to produce its own siderophores. Both strains are present in similar proportions in the culture as long as the siderophore-deficient P. aeruginosa strain is able to use ENT produced by E. coli to access iron. If pfeE is deleted, E. coli has the upper hand in the culture and P. aeruginosa growth is repressed. Overall, these data show that PfeE is the Achilles' heel of P. aeruginosa in communities with bacteria producing ENT.


Subject(s)
Carrier Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Esterases/metabolism , Iron/metabolism , Pseudomonas aeruginosa/metabolism , Carrier Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Esterases/genetics , Pseudomonas aeruginosa/genetics
9.
Microorganisms ; 8(11)2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33218210

ABSTRACT

Iron acquisition pathways have often been considered to be gateways for the uptake of antibiotics into bacteria. Bacteria excrete chelators, called siderophores, to access iron. Antibiotic molecules can be covalently attached to siderophores for their transport into pathogens during the iron-uptake process. P. aeruginosa produces two siderophores and is also able to use many siderophores produced by other bacteria. We investigated the phenotypic plasticity of iron-uptake pathway expression in an epithelial cell infection assay in the presence of two different siderophore-antibiotic conjugates, one with a hydroxamate siderophore and the second with a tris-catechol. Proteomic and RT-qPCR approaches showed that P. aeruginosa was able to sense the presence of both compounds in its environment and adapt the expression of its iron uptake pathways to access iron via them. Moreover, the catechol-type siderophore-antibiotic was clearly more efficient in inducing the expression of its corresponding transporter than the hydroxamate compound when both were simultaneously present. In parallel, the expression of the proteins of the two iron uptake pathways using siderophores produced by P. aeruginosa was significantly repressed in the presence of both conjugates. Altogether, the data indicate that catechol-type siderophores are more promising vectors for antibiotic vectorization using a Trojan-horse strategy.

10.
J Vis Exp ; (162)2020 08 25.
Article in English | MEDLINE | ID: mdl-32925892

ABSTRACT

Protein-protein interactions (PPIs) control various key processes in cells. Fluorescence lifetime imaging microscopy (FLIM) combined with Förster resonance energy transfer (FRET) provide accurate information about PPIs in live cells. FLIM-FRET relies on measuring the fluorescence lifetime decay of a FRET donor at each pixel of the FLIM image, providing quantitative and accurate information about PPIs and their spatial cellular organizations. We propose here a detailed protocol for FLIM-FRET measurements that we applied to monitor PPIs in live Pseudomonas aeruginosa in the particular case of two interacting proteins expressed with highly different copy numbers to demonstrate the quality and robustness of the technique at revealing critical features of PPIs. This protocol describes in detail all the necessary steps for PPI characterization - starting from bacterial mutant constructions up to the final analysis using recently developed tools providing advanced visualization possibilities for a straightforward interpretation of complex FLIM-FRET data.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Microscopy, Fluorescence/methods , Protein Interaction Mapping , Pseudomonas aeruginosa/metabolism , Algorithms , Binding Sites , Chromosomes, Bacterial/genetics , Fluorescent Dyes/metabolism , Genome, Bacterial , Photons , Plasmids/metabolism , Pseudomonas aeruginosa/genetics , Software
11.
Mol Cell Proteomics ; 19(4): 589-607, 2020 04.
Article in English | MEDLINE | ID: mdl-32024770

ABSTRACT

Bacteria secrete siderophores to access iron, a key nutrient poorly bioavailable and the source of strong competition between microorganisms in most biotopes. Many bacteria also use siderophores produced by other microorganisms (exosiderophores) in a piracy strategy. Pseudomonas aeruginosa, an opportunistic pathogen, produces two siderophores, pyoverdine and pyochelin, and is also able to use a panel of exosiderophores. We first investigated expression of the various iron-uptake pathways of P. aeruginosa in three different growth media using proteomic and RT-qPCR approaches and observed three different phenotypic patterns, indicating complex phenotypic plasticity in the expression of the various iron-uptake pathways. We then investigated the phenotypic plasticity of iron-uptake pathway expression in the presence of various exosiderophores (present individually or as a mixture) under planktonic growth conditions, as well as in an epithelial cell infection assay. In all growth conditions tested, catechol-type exosiderophores were clearly more efficient in inducing the expression of their corresponding transporters than the others, showing that bacteria opt for the use of catechol siderophores to access iron when they are present in the environment. In parallel, expression of the proteins of the pyochelin pathway was significantly repressed under most conditions tested, as well as that of proteins of the pyoverdine pathway, but to a lesser extent. There was no effect on the expression of the heme and ferrous uptake pathways. Overall, these data provide precise insights on how P. aeruginosa adjusts the expression of its various iron-uptake pathways (phenotypic plasticity and switching) to match varying levels of iron and competition.


Subject(s)
Adaptation, Physiological , Pseudomonas aeruginosa/physiology , Siderophores/metabolism , A549 Cells , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Bacterial Proteins/metabolism , Biological Transport/drug effects , Catechols/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial/drug effects , Humans , Iron/metabolism , Iron Chelating Agents/pharmacology , Pseudomonas aeruginosa/cytology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Siderophores/chemistry , Transcription, Genetic/drug effects , Virulence Factors/metabolism
12.
Q Rev Biophys ; 53: e1, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31915092

ABSTRACT

The bio-synthesis of pyoverdine (PVD) in Pseudomonas aeruginosa involves multiple enzymatic steps including the action of non-ribosomal peptide synthetases (NRPSs). One hallmark of NRPS is their ability to make usage of non-proteinogenic amino-acids synthesized by co-expressed accessory enzymes. It is generally proposed that different enzymes of a secondary metabolic pathway assemble into large supra-molecular complexes. However, evidence for the assembly of sequential enzymes in the cellular context is sparse. Here, we used in cellulo single-molecule tracking and Förster resonance energy transfer measured by fluorescence lifetime microscopy (FRET-FLIM) to explore the spatial partitioning of the ornithine hydroxylase PvdA and its interactions with NRPS. We found PvdA was mostly diffusing bound to large complexes in the cytoplasm with a small exchangeable trapped fraction. FRET-FLIM clearly showed that PvdA is physically interacting with PvdJ, PvdI, PvdL, and PvdD, the four NRPS involved in the PVD pathway in Pseudomonas aeruginosa PAO1. The binding modes of PvdA were strikingly different according to the NRPS it is interacting with, suggesting that PvdA binding sites have co-evolved with the enzymatic active sites of NRPS. Our data provide evidence for strongly organized multi-enzymatic complexes responsible for the bio-synthesis of PVD and illustrate how binding sites have evolved to finely control the co-localization of sequential enzymes and promote metabolic pathway efficiency.


Subject(s)
Mixed Function Oxygenases/metabolism , Oligopeptides/chemistry , Pseudomonas aeruginosa/metabolism , Single Molecule Imaging/methods , Cluster Analysis , Cytoplasm/metabolism , Escherichia coli/metabolism , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence/methods , Mutation , Oligopeptides/metabolism , Protein Binding
13.
Nat Commun ; 10(1): 3673, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413254

ABSTRACT

Bacteria use small molecules called siderophores to scavenge iron. Siderophore-Fe3+ complexes are recognised by outer-membrane transporters and imported into the periplasm in a process dependent on the inner-membrane protein TonB. The siderophore enterobactin is secreted by members of the family Enterobacteriaceae, but many other bacteria including Pseudomonas species can use it. Here, we show that the Pseudomonas transporter PfeA recognises enterobactin using extracellular loops distant from the pore. The relevance of this site is supported by in vivo and in vitro analyses. We suggest there is a second binding site deeper inside the structure and propose that correlated changes in hydrogen bonds link binding-induced structural re-arrangements to the structural adjustment of the periplasmic TonB-binding motif.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Enterobactin/metabolism , Iron/metabolism , Pseudomonas aeruginosa/metabolism , Receptors, Cell Surface/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins , Binding Sites , Carrier Proteins/chemistry , Crystallization , Crystallography, X-Ray , Enterobactin/chemistry , Escherichia coli , In Vitro Techniques , Iron Radioisotopes , Membrane Proteins , Receptors, Cell Surface/chemistry , Siderophores/chemistry , Siderophores/metabolism
14.
ACS Chem Biol ; 13(9): 2603-2614, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30086222

ABSTRACT

Enterobactin (ENT) is a siderophore (iron-chelating compound) produced by Escherichia coli to gain access to iron, an indispensable nutrient for bacterial growth. ENT is used as an exosiderophore by Pseudomonas aeruginosa with transport of ferri-ENT across the outer membrane by the PfeA transporter. Next to the pfeA gene on the chromosome is localized a gene encoding for an esterase, PfeE, whose transcription is regulated, as for pfeA, by the presence of ENT in bacterial environment. Purified PfeE hydrolyzed ferri-ENT into three molecules of 2,3-DHBS (2,3-dihydroxybenzoylserine) still complexed with ferric iron, and complete dissociation of iron from ENT chelating groups was only possible in the presence of both PfeE and an iron reducer, such as DTT. The crystal structure of PfeE and an inactive PfeE mutant complexed with ferri-ENT or a nonhydrolyzable ferri-catechol complex allowed identification of the enzyme binding site and the catalytic triad. Finally, cell fractionation and fluorescence microscopy showed periplasmic localization of PfeE in P. aeruginosa cells. Thus, the molecular mechanism of iron dissociation from ENT in P. aeruginosa differs from that previously described in E. coli. In P. aeruginosa, siderophore hydrolysis occurs in the periplasm, with ENT never reaching the bacterial cytoplasm. In E. coli, ferri-ENT crosses the inner membrane via the ABC transporter FepBCD and ferri-ENT is hydrolyzed by the esterase Fes only once it is in the cytoplasm.


Subject(s)
Bacterial Proteins/metabolism , Enterobactin/metabolism , Esterases/metabolism , Iron/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Bacterial Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Esterases/chemistry , Humans , Hydrolysis , Models, Molecular , Protein Conformation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/cytology , Pseudomonas aeruginosa/enzymology
15.
ACS Chem Biol ; 12(4): 1056-1065, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28192658

ABSTRACT

Siderophores are iron chelators produced by bacteria to access iron, an essential nutriment. Pyoverdine (PVDI), the major siderophore produced by Pseudomonas aeruginosa PAO1, consists of a fluorescent chromophore linked to an octapeptide. The ferric form of PVDI is transported from the extracellular environment into the periplasm by the outer membrane transporter, FpvA. Iron is then released from the siderophore in the periplasm by a mechanism that does not involve chemical modification of the chelator but an iron reduction step. Here, we followed the kinetics of iron release from PVDI, in vitro and in living cells, by monitoring its fluorescence (as apo PVDI is fluorescent, whereas PVDI-Fe(III) is not). Deletion of the inner membrane proteins fpvG (PA2403) and fpvH (PA2404) affected 55Fe uptake via PVDI and completely abolished PVDI-Fe dissociation, indicating that these two proteins are involved in iron acquisition via this siderophore. PVDI-Fe dissociation studies, using an in vitro assay, showed that iron release from this siderophore requires the presence of an iron reducer (DTT) and an iron chelator (ferrozine). In this assay, DTT could be replaced by the inner membrane protein, FpvG, and ferrozine by the periplasmic protein, FpvC, suggesting that FpvG acts as a reductase and FpvC as an Fe2+ chelator in the process of PVDI-Fe dissociation in the periplasm of P. aeruginosa cells. This mechanism of iron release from PVDI is atypical among Gram-negative bacteria but seems to be conserved among Pseudomonads.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Oligopeptides/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Bacterial Proteins/genetics , Gene Deletion , Genes, Bacterial , Pseudomonas aeruginosa/genetics , Spectrometry, Fluorescence
16.
Environ Microbiol ; 18(3): 819-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718479

ABSTRACT

Previous studies have suggested that antibiotic vectorization by siderophores (iron chelators produced by bacteria) considerably increases the efficacy of such drugs. The siderophore serves as a vector: when the pathogen tries to take up iron via the siderophore, it also takes up the antibiotic. Catecholates are among the most common iron-chelating compounds used in synthetic siderophore-antibiotic conjugates. Using reverse transcription polymerase chain reaction and proteomic approaches, we showed that the presence of catecholate compounds in the medium of Pseudomonas aeruginosa led to strong activation of the transcription and expression of the outer membrane transporter PfeA, the ferri-enterobactin importer. Iron-55 uptake assays on bacteria with and without PfeA expression confirmed that catechol compounds imported iron into P. aeruginosa cells via PfeA. Uptake rates were between 0.3 × 10(3) and 2 × 10(3) Fe atoms/bacterium/min according to the used catechol siderophore in iron-restricted medium, and remained as high as 0.8 × 10(3) Fe atoms/bacterium/min for enterobactin, even in iron-rich medium. Reverse transcription polymerase chain reaction and proteomic approaches showed that in parallel to this switching on of PfeA expression, a repression of the expression of pyochelin (PCH) pathway genes (PCH being one of the two siderophores produced by P. aeruginosa for iron acquisition) was observed.


Subject(s)
Anti-Bacterial Agents/metabolism , Catechols/metabolism , Enterobactin/metabolism , Phenols/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Thiazoles/metabolism , Iron/metabolism , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/genetics
17.
Org Biomol Chem ; 13(46): 11401, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26555129

ABSTRACT

Correction for 'Synthesis and antibiotic activity of oxazolidinone-catechol conjugates against Pseudomonas aeruginosa' by Aurélie Paulen, et al., Org. Biomol. Chem., 2016, DOI: 10.1039/c5ob01859e.

18.
Org Biomol Chem ; 13(47): 11567-79, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26509593

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative pathogenic bacterium responsible for severe infections in which resistance to most of the approved families of antibiotics is increasing. Oxazolidinone antibiotics are active against many Gram-positive bacteria, but are only weakly active against Gram-negative pathogens. We describe the synthesis of conjugates between a catechol moiety and oxazolidinone antibiotics. These conjugates were significantly more active against P. aeruginosa (218-1024 µM) than linezolid (MIC > 1024 µM), the reference molecule from the oxazolidinone family. Antibiotic activity was slightly higher in medium depleted of iron, suggesting the involvement of a bacterial iron uptake system in this biological activity. The bacterial iron uptake pathway involved in the transport is still to be addressed, but the present data excluded a contribution of the enterobactin transporter PfeA.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Catechols/chemistry , Catechols/pharmacology , Oxazolidinones/chemistry , Oxazolidinones/pharmacology , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemical synthesis , Catechols/chemical synthesis , Humans , Microbial Sensitivity Tests , Oxazolidinones/chemical synthesis , Pseudomonas Infections/drug therapy
19.
J Inorg Biochem ; 148: 27-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25697961

ABSTRACT

Pyoverdine I (PVDI) and pyochelin (PCH) are the two major siderophores produced by Pseudomonas aeruginosa PAO1 to import iron. The biochemistry of the biosynthesis of these two siderophores has been described in detail in the literature over recent years. PVDI assembly requires the coordinated action of seven cytoplasmic enzymes and is followed by a periplasmic maturation before secretion of the siderophore into the extracellular medium by the efflux system PvdRT-OpmQ. PCH biosynthesis also involves seven cytoplasmic enzymes but no periplasmic maturation. Recent findings indicate that the cytoplasmic enzymes involved in each of these two siderophore biosynthesis pathways can form siderophore-specific multi-enzymatic complexes called siderosomes associated with the inner leaflet of the cytoplasmic membrane. This organization may optimize the transfer of the siderophore precursors between the various participating enzymes and avoid the diffusion of siderophore precursors, able to chelate metals, throughout the cytoplasm. Here, we describe these recently published findings and discuss the existence of these siderosomes in P. aeruginosa.


Subject(s)
Biosynthetic Pathways , Oligopeptides/biosynthesis , Pseudomonas aeruginosa/metabolism , Siderophores/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytoplasm/enzymology , Cytoplasm/metabolism , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Molecular Structure , Oligopeptides/chemistry , Pseudomonas aeruginosa/genetics , Siderophores/chemistry
20.
Environ Microbiol ; 17(1): 171-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24947078

ABSTRACT

Pyochelin (PCH) is a siderophore produced and secreted by Pseudomonas aeruginosa for iron capture. Using (55) Fe uptake and binding assays, we showed that PCH-Fe uptake in P. aeruginosa involves, in addition to the highly studied outer membrane transporter FptA, the inner membrane permease FptX, which recognizes PCH-(55) Fe with an affinity of 0.6 ± 0.2 nM and transports the ferri-siderophore complex from the periplasm into the cytoplasm: fptX deletion inhibited (55) Fe accumulation in the bacterial cytoplasm. Chromosomal replacement was used to generate P. aeruginosa strains producing fluorescent fusions with FptX, PchR (an AraC regulator), PchA (the first enzyme involved in the PCH biosynthesis) and PchE (a non-ribosomic peptide-synthetase involved in a further step). Fluorescence imaging and cellular fractionation showed a uniform repartition of FptX in the inner membrane. PchA and PchE were found in the cytoplasm, associated to the inner membrane all over the bacteria and also concentrated at the bacterial poles. PchE clustering at the bacterial poles was dependent on PchA expression, but on the opposite PchA clustering and membrane association was PchE-independent. PchA and PchE cellular organization suggests the existence of a siderosome for PCH biosynthesis as previously proposed for pyoverdine biosynthesis (another siderophore produced by P. aeruginosa).


Subject(s)
Iron/metabolism , Phenols/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Thiazoles/metabolism , Bacterial Proteins/metabolism , Biological Transport , Membrane Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL