Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Microbiol ; 25(4): 811-831, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36571575

RESUMO

Pseudomonas aeruginosa is a ubiquitous bacterium found in many natural and man-made environments. It is also a pathogen for plants, animals, and humans. As for almost all living organisms, iron is an essential nutrient for the growth of P. aeruginosa. The bacterium has evolved complex systems to access iron and maintain its homeostasis to survive in diverse natural and dynamic host environments. To access ferric iron, P. aeruginosa is able to produce two siderophores (pyoverdine and pyochelin), as well as use a variety of siderophores produced by other bacteria (mycobactins, enterobactin, ferrioxamine, ferrichrome, vibriobactin, aerobactin, rhizobactin and schizokinen). Furthermore, it can also use citrate, in addition to catecholamine neuromediators and plant-derived mono catechols, as siderophores. The P. aeruginosa genome also encodes three heme-uptake pathways (heme being an iron source) and one ferrous iron acquisition pathway. This review aims to summarize current knowledge concerning the molecular mechanisms involved in all the iron and heme acquisition strategies used by P. aeruginosa.


Assuntos
Ferro , Sideróforos , Humanos , Ferro/metabolismo , Sideróforos/metabolismo , Pseudomonas aeruginosa/metabolismo , Ferricromo/metabolismo , Transporte Biológico
2.
ACS Infect Dis ; 8(6): 1134-1146, 2022 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-35500104

RESUMO

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.


Assuntos
Pseudomonas aeruginosa , Sideróforos , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Benzamidas , Catecóis/metabolismo , Catecóis/farmacologia , Bactérias Gram-Negativas/metabolismo , Hidroxibenzoatos , Ferro/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Sideróforos/farmacologia , Zinco/metabolismo
3.
Environ Microbiol ; 24(2): 878-893, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33350053

RESUMO

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.


Assuntos
Pseudomonas aeruginosa , Sideróforos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Catecolaminas/metabolismo , Ferro/metabolismo , Neurotransmissores/metabolismo , Proteômica , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo
4.
Microorganisms ; 8(11)2020 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-33218210

RESUMO

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.

5.
J Vis Exp ; (162)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32925892

RESUMO

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.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Microscopia de Fluorescência/métodos , Mapeamento de Interação de Proteínas , Pseudomonas aeruginosa/metabolismo , Algoritmos , Sítios de Ligação , Cromossomos Bacterianos/genética , Corantes Fluorescentes/metabolismo , Genoma Bacteriano , Fótons , Plasmídeos/metabolismo , Pseudomonas aeruginosa/genética , Software
6.
ACS Chem Biol ; 15(10): 2741-2751, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32902248

RESUMO

Iron is a key nutrient for almost all living organisms. Paradoxically, it is poorly soluble and consequently poorly bioavailable. Bacteria have thus developed multiple strategies to access this metal. One of the most common consists of the use of siderophores, small compounds that chelate ferric iron with very high affinity. Many bacteria are able to produce their own siderophores or use those produced by other microorganisms (exosiderophores) in a piracy strategy. Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, and is also able to use a large panel of exosiderophores. We investigated the ability of P. aeruginosa to use nocardamine (NOCA) and ferrioxamine B (DFOB) as exosiderophores under iron-limited planktonic growth conditions. Proteomic and RT-qPCR approaches showed induction of the transcription and expression of the outer membrane transporter FoxA in the presence of NOCA or DFOB in the bacterial environment. Expression of the proteins of the heme- or pyoverdine- and pyochelin-dependent iron uptake pathways was not affected by the presence of these two tris-hydroxamate siderophores. 55Fe uptake assays using foxA mutants showed ferri-NOCA to be exclusively transported by FoxA, whereas ferri-DFOB was transported by FoxA and at least one other unidentified transporter. The crystal structure of FoxA complexed with NOCA-Fe revealed very similar siderophore binding sites between NOCA-Fe and DFOB-Fe. We discuss iron uptake by hydroxamate exosiderophores in P. aeruginosa cells in light of these results.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Ferro/metabolismo , Peptídeos Cíclicos/metabolismo , Pseudomonas aeruginosa/metabolismo , Receptores de Superfície Celular/metabolismo , Sideróforos/metabolismo , Cristalografia por Raios X , Desferroxamina/metabolismo , Compostos Férricos/metabolismo , Expressão Gênica/efeitos dos fármacos , Ligação Proteica , Transcrição Gênica/efeitos dos fármacos
7.
Mol Cell Proteomics ; 19(4): 589-607, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32024770

RESUMO

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.


Assuntos
Adaptação Fisiológica , Pseudomonas aeruginosa/fisiologia , Sideróforos/metabolismo , Células A549 , Adaptação Fisiológica/efeitos dos fármacos , Adaptação Fisiológica/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico/efeitos dos fármacos , Catecóis/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Sideróforos/química , Transcrição Gênica/efeitos dos fármacos , Fatores de Virulência/metabolismo
8.
Metallomics ; 11(11): 1937-1951, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31633703

RESUMO

Much data shows that biological metals other than Fe3+ can interfere with Fe3+ acquisition by siderophores in bacteria. Siderophores are small Fe3+ chelators produced by the microorganisms to obtain access to Fe3+. Here, we show that Co2+ is imported into Pseudomonas aeruginosa cells in a complex with the siderophore pyochelin (PCH) by the ferri-PCH outer membrane transporter FptA. Moreover, the presence of Co2+ in the bacterial environment strongly affects the production of PCH. Proteomic and transcriptomic approaches showed that a decrease of PCH production is associated with repression of the expression of the genes involved in PCH biosynthesis. We used various molecular biology approaches to show that this repression is not Fur-(ferric uptake transcriptional regulator) dependent but due to competition of PCH-Co with PCH-Fe for PchR (transcriptional activator), thus inhibiting the formation of PchR-PCH-Fe and consequently the expression of the PCH genes. We observed a similar mechanism of repression of PCH production, but to a lesser extent, by Ni2+, but not for Zn2+, Cu2+, or Mn2+. Here, we show, for the first time at a molecular level, how the presence of a contaminant metal can interfere with Fe3+ acquisition by the siderophores PCH and PVD.


Assuntos
Cobalto/metabolismo , Ferro/metabolismo , Sideróforos/metabolismo , Proteínas de Bactérias/metabolismo , Cobalto/farmacologia , Regulação para Baixo/genética , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Modelos Biológicos , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Óperon/genética , Fenóis/química , Fenóis/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Tiazóis/química , Tiazóis/metabolismo , Regulação para Cima/genética
9.
ACS Chem Biol ; 13(9): 2603-2614, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30086222

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Enterobactina/metabolismo , Esterases/metabolismo , Ferro/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Proteínas de Bactérias/química , Sítios de Ligação , Cristalografia por Raios X , Esterases/química , Humanos , Hidrólise , Modelos Moleculares , Conformação Proteica , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/enzimologia
10.
Environ Microbiol ; 18(3): 819-32, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26718479

RESUMO

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.


Assuntos
Antibacterianos/metabolismo , Catecóis/metabolismo , Enterobactina/metabolismo , Fenóis/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Tiazóis/metabolismo , Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Pseudomonas aeruginosa/genética
11.
Org Biomol Chem ; 13(46): 11401, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26555129

RESUMO

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.

12.
Org Biomol Chem ; 13(47): 11567-79, 2015 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-26509593

RESUMO

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.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Catecóis/química , Catecóis/farmacologia , Oxazolidinonas/química , Oxazolidinonas/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Antibacterianos/síntese química , Catecóis/síntese química , Humanos , Testes de Sensibilidade Microbiana , Oxazolidinonas/síntese química , Infecções por Pseudomonas/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...