Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Science ; 382(6676): eadj3502, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38096285

ABSTRACT

The human gut microbiome plays an important role in resisting colonization of the host by pathogens, but we lack the ability to predict which communities will be protective. We studied how human gut bacteria influence colonization of two major bacterial pathogens, both in vitro and in gnotobiotic mice. Whereas single species alone had negligible effects, colonization resistance greatly increased with community diversity. Moreover, this community-level resistance rested critically upon certain species being present. We explained these ecological patterns through the collective ability of resistant communities to consume nutrients that overlap with those used by the pathogen. Furthermore, we applied our findings to successfully predict communities that resist a novel target strain. Our work provides a reason why microbiome diversity is beneficial and suggests a route for the rational design of pathogen-resistant communities.


Subject(s)
Gastrointestinal Microbiome , Host-Pathogen Interactions , Klebsiella Infections , Klebsiella pneumoniae , Salmonella Infections , Salmonella typhimurium , Animals , Humans , Mice , Nutrients/metabolism , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Symbiosis , Germ-Free Life , Klebsiella Infections/microbiology , Salmonella Infections/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism
2.
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
3.
Microlife ; 2: uqab001, 2021.
Article in English | MEDLINE | ID: mdl-34250489

ABSTRACT

Nutritional immunity is a powerful strategy at the core of the battlefield between host survival and pathogen proliferation. A host can prevent pathogens from accessing biological metals such as Mg, Fe, Zn, Mn, Cu, Co or Ni, or actively intoxicate them with metal overload. While the importance of metal homeostasis for the enteric pathogen Salmonella enterica Typhimurium was demonstrated many decades ago, inconsistent results across various mouse models, diverse Salmonella genotypes, and differing infection routes challenge aspects of our understanding of this phenomenon. With expanding access to CRISPR-Cas9 for host genome manipulation, it is now pertinent to re-visit past results in the context of specific mouse models, identify gaps and incongruities in current knowledge landscape of Salmonella homeostasis, and recommend a straight path forward towards a more universal understanding of this historic host-microbe relationship.

4.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34326266

ABSTRACT

Gram-negative bacterial pathogens have an outer membrane that restricts entry of molecules into the cell. Water-filled protein channels in the outer membrane, so-called porins, facilitate nutrient uptake and are thought to enable antibiotic entry. Here, we determined the role of porins in a major pathogen, Pseudomonas aeruginosa, by constructing a strain lacking all 40 identifiable porins and 15 strains carrying only a single unique type of porin and characterizing these strains with NMR metabolomics and antimicrobial susceptibility assays. In contrast to common assumptions, all porins were dispensable for Pseudomonas growth in rich medium and consumption of diverse hydrophilic nutrients. However, preferred nutrients with two or more carboxylate groups such as succinate and citrate permeated poorly in the absence of porins. Porins provided efficient translocation pathways for these nutrients with broad and overlapping substrate selectivity while efficiently excluding all tested antibiotics except carbapenems, which partially entered through OprD. Porin-independent permeation of antibiotics through the outer-membrane lipid bilayer was hampered by carboxylate groups, consistent with our nutrient data. Together, these results challenge common assumptions about the role of porins by demonstrating porin-independent permeation of the outer-membrane lipid bilayer as a major pathway for nutrient and drug entry into the bacterial cell.


Subject(s)
Anti-Bacterial Agents/metabolism , Cell Membrane/physiology , Nutrients/metabolism , Porins/metabolism , Pseudomonas aeruginosa/physiology , Bacterial Outer Membrane Proteins/metabolism , Biological Transport/physiology , Cell Membrane Permeability
5.
mBio ; 12(3): e0086921, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34061589

ABSTRACT

Bacterial outer membrane vesicles (OMVs) enriched with bioactive proteins, toxins, and virulence factors play a critical role in host-pathogen and microbial interactions. The two-component system PhoP-PhoQ (PhoPQ) of Salmonella enterica orchestrates the remodeling of outer membrane lipopolysaccharide (LPS) molecules and concomitantly upregulates OMV production. In this study, we document a novel use of nanoparticle tracking analysis to determine bacterial OMV size and number. Among the PhoPQ-activated genes tested, pagC expression had the most significant effect on the upregulation of OMV production. We provide the first evidence that PhoPQ-mediated upregulation of OMV production contributes to bacterial survival by interfering with complement activation. OMVs protected bacteria in a dose-dependent manner, and bacteria were highly susceptible to complement-mediated killing in their absence. OMVs from bacteria expressing PagC bound to complement component C3b in a dose-dependent manner and inactivated it by recruiting complement inhibitor Factor H. As we also found that Factor H binds to PagC, we propose that PagC interferes with complement-mediated killing of Salmonella in the following two steps: first by engaging Factor H, and second, through the production of PagC-enriched OMVs that divert and inactivate the complement away from the bacteria. Since PhoPQ activation occurs intracellularly, the resultant increase in PagC expression and OMV production is suggested to contribute to the local and systemic spread of Salmonella released from dying host cells that supports the infection of new cells. IMPORTANCE Bacterial outer membrane vesicles (OMVs) mediate critical bacterium-bacterium and host-microbial interactions that influence pathogenesis through multiple mechanisms, including the elicitation of inflammatory responses, delivery of virulence factors, and enhancement of biofilm formation. As such, there is a growing interest in understanding the underlying mechanisms of OMV production. Recent studies have revealed that OMV biogenesis is a finely tuned physiological process that requires structural organization and selective sorting of outer membrane components into the vesicles. In Salmonella, outer membrane remodeling and OMV production are tightly regulated by its PhoPQ system. In this study, we demonstrate that PhoPQ-regulated OMV production plays a significant role in defense against host innate immune attack. PhoPQ-activated PagC expression recruits the complement inhibitor Factor H and degrades the active C3 component of complement. Our results provide valuable insight into the combination of tools and environmental signals that Salmonella employs to evade complement-mediated lysis, thereby suggesting a strong evolutionary adaptation of this facultative intracellular pathogen to protect itself during its extracellular stage in the host.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement System Proteins/immunology , Host Microbial Interactions/immunology , Immunity, Innate , Salmonella typhimurium/immunology , Secretory Vesicles/immunology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins , Immune Evasion , Salmonella typhimurium/pathogenicity
6.
Metallomics ; 12(12): 2108-2120, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33355556

ABSTRACT

Siderophores are iron chelators produced by bacteria to access iron, an essential nutrient. The pathogen Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, the former with a high affinity for iron and the latter with a lower affinity. Furthermore, the production of both siderophores involves a positive auto-regulatory loop: the presence of the ferri-siderophore complex is essential for their large production. Since pyochelin has a lower affinity for iron it was hard to consider the role of pyochelin in drastic competitive environments where the host or the environmental microbiota produce strong iron chelators and may inhibit iron chelation by pyochelin. We showed here that the pyochelin pathway overcomes this difficulty through a more complex regulating mechanism for pyochelin production than previously described. Indeed, in the absence of pyoverdine, and thus higher difficulty to access iron, the bacteria are able to produce pyochelin independently of the presence of ferri-pyochelin. The regulation of the pyochelin pathway appeared to be more complex than expected with a more intricate tuning between repression and activation. Consequently, when the bacteria cannot produce pyoverdine they are able to produce pyochelin even in the presence of strong iron chelators. Such results support a more complex and varied role for this siderophore than previously described, and complexify the battle for iron during P. aeruginosa infection.


Subject(s)
Phenols/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Thiazoles/metabolism , Humans , Iron/metabolism , Oligopeptides/metabolism , Pseudomonas Infections/microbiology
7.
BMC Microbiol ; 20(1): 129, 2020 05 24.
Article in English | MEDLINE | ID: mdl-32448155

ABSTRACT

BACKGROUND: Gene editing is key for elucidating gene function. Traditional methods, such as consecutive single-crossovers, have been widely used to modify bacterial genomes. However, cumbersome cloning and limited efficiency of negative selection often make this method slower than other methods such as recombineering. RESULTS: Here, we established a time-effective variant of consecutive single-crossovers. This method exploits rapid plasmid construction using Gibson assembly, a convenient E. coli donor strain, and efficient dual-negative selection for improved suicide vector resolution. We used this method to generate in-frame deletions, insertions and point mutations in Salmonella enterica with limited hands-on time. Adapted versions enabled efficient gene editing also in Pseudomonas aeruginosa and multi-drug resistant (MDR) Escherichia coli clinical isolates. CONCLUSIONS: Our method is time-effective and allows facile manipulation of multiple bacterial species including MDR clinical isolates. We anticipate that this method might be broadly applicable to additional bacterial species, including those for which recombineering has been difficult to implement.


Subject(s)
Escherichia coli/genetics , Gene Editing/methods , Plasmids/genetics , Pseudomonas aeruginosa/genetics , Salmonella enterica/genetics , CRISPR-Cas Systems , Conjugation, Genetic , Drug Resistance, Multiple, Bacterial , Genes, Transgenic, Suicide , Genome, Bacterial , Mutation
8.
Science ; 366(6468): 995-999, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31753999

ABSTRACT

The pleiotropic host resistance factor SLC11A1 (NRAMP1) defends against diverse intracellular pathogens in mammals by yet-unknown mechanisms. We compared Salmonella infection of coisogenic mice with different SLC11A1 alleles. SLC11A1 reduced Salmonella replication and triggered up-regulation of uptake systems for divalent metal cations but no other stress responses. SLC11A1 modestly diminished iron availability and acutely restricted Salmonella access to magnesium. Growth of Salmonella cells in the presence of SLC11A1 was highly heterogeneous and inversely correlated with expression of the crucial magnesium transporter gene mgtB We observed superimposable single-cell patterns in mice lacking SLC11A1 when we restricted Salmonella access to magnesium by impairing its uptake. Together, these findings identify deprivation of the main group metal magnesium as the main resistance mechanism of SLC11A1 against Salmonella.


Subject(s)
Cation Transport Proteins/metabolism , Magnesium/metabolism , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/growth & development , Adenosine Triphosphatases/metabolism , Animals , Bacterial Proteins/metabolism , Cation Transport Proteins/genetics , Disease Resistance/genetics , Genetic Fitness , Iron/metabolism , Mice , Mice, Inbred C57BL , Mutation , Proteome , Salmonella Infections, Animal/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Single-Cell Analysis , Spleen/microbiology
9.
Metallomics ; 11(11): 1937-1951, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31633703

ABSTRACT

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.


Subject(s)
Cobalt/metabolism , Iron/metabolism , Siderophores/metabolism , Bacterial Proteins/metabolism , Cobalt/pharmacology , Down-Regulation/genetics , Gene Expression Regulation, Bacterial/drug effects , Models, Biological , Oligopeptides/chemistry , Oligopeptides/metabolism , Operon/genetics , Phenols/chemistry , Phenols/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Up-Regulation/genetics
10.
EBioMedicine ; 41: 479-487, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30852163

ABSTRACT

BACKGROUND: Efflux pumps mediate antimicrobial resistance in several WHO critical priority bacterial pathogens. However, most available data come from laboratory strains. The quantitative relevance of efflux in more relevant clinical isolates remains largely unknown. METHODS: We developed a versatile method for genetic engineering in multi-drug resistant (MDR) bacteria, and used this method to delete tolC and specific antibiotic-resistance genes in 18 representative MDR clinical E. coli isolates. We determined efflux activity and minimal inhibitory concentrations for a diverse set of clinically relevant antibiotics in these mutants. We also deleted oprM in MDR P. aeruginosa strains and determined the impact on antibiotic susceptibility. FINDINGS: tolC deletion abolished detectable efflux activity in 15 out of 18 tested E. coli strains, and modulated antibiotic susceptibility in many strains. However, all mutant strains retained MDR status, primarily because of other, antibiotic-specific resistance genes. Deletion of oprM altered antibiotic susceptibility in a fraction of clinical P. aeruginosa isolates. INTERPRETATION: Efflux modulates antibiotic resistance in clinical MDR isolates of E. coli and P. aeruginosa. However, when other antimicrobial-resistance mechanisms are present, inhibition of MDR efflux pumps alone is often not sufficient to restore full susceptibility even for antibiotics with a dramatic impact of efflux in laboratory strains. We propose that development of novel antibiotics should include target validation in clinical MDR isolates. FUND: Innovative Medicines Initiative of European Union and EFPIA, Schweizerischer Nationalfonds, Swiss National Research Program 72, EU Marie Sklodowska-Curie program. The funders played no role in design, data collection, data analysis, interpretation, writing of the report, and in the decision to submit the paper for publication.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Bacteremia/pathology , Bacterial Outer Membrane Proteins/genetics , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Escherichia coli Proteins/genetics , Gene Deletion , Humans , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Whole Genome Sequencing
11.
Curr Opin Microbiol ; 39: 57-63, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28988065

ABSTRACT

Infected host tissues have complex anatomy, diverse cell types, and dynamic inflammation. Traditional infection biology approaches largely ignore this complex host environment and its impact on pathogens, but recent single-cell technologies unravel extensively heterogeneous host-pathogen interactions in vivo. Salmonella are major model pathogens in this field due to the availability of excellent mouse disease models and facile molecular biology. The results show how Salmonella stochastically vary their virulence, exploit differential nutrient availability, experience and respond to widely varying stresses, and have disparate fates ranging from vigorous proliferation to eradication within the same host tissue. Specific Salmonella subsets drive disease progression, while others persist during antimicrobial chemotherapy. Further elucidation of the underlying mechanisms could provide a basis for improved infection control.


Subject(s)
Host-Pathogen Interactions , Salmonella Infections , Salmonella , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Mice
12.
Environ Microbiol ; 18(10): 3227-3246, 2016 10.
Article in English | MEDLINE | ID: mdl-27632589

ABSTRACT

Biological metal ions, including Co, Cu, Fe, Mg, Mn, Mo, Ni and Zn ions, are necessary for the survival and the growth of all microorganisms. Their biological functions are linked to their particular chemical properties: they play a role in structuring macromolecules and/or act as co-factors catalyzing diverse biochemical reactions. These metal ions are also essential for microbial pathogens during infection: they are involved in bacterial metabolism and various virulence factor functions. Therefore, during infection, bacteria need to acquire biological metal ions from the host such that there is competition for these ions between the bacterium and the host. Evidence is increasingly emerging of "nutritional immunity" against pathogens in the hosts; this includes strategies making access to metals difficult for infecting bacteria. It is clear that biological metals play key roles during infection and in the battle between the pathogens and the host. Here, we summarize current knowledge about the strategies used by Pseudomonas aeruginosa to access the various biological metals it requires. P. aeruginosa is a medically significant Gram-negative bacterial opportunistic pathogen that can cause severe chronic lung infections in cystic fibrosis patients and that is responsible for nosocomial infections worldwide.


Subject(s)
Biological Transport, Active/physiology , Ion Transport/physiology , Ions/metabolism , Metals/metabolism , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/metabolism , Respiratory Tract Infections/pathology , Cross Infection , Cystic Fibrosis/immunology , Humans , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Respiratory Tract Infections/microbiology , Virulence Factors/metabolism
13.
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
14.
Environ Microbiol ; 18(10): 3258-3267, 2016 10.
Article in English | MEDLINE | ID: mdl-26147433

ABSTRACT

In this paper, we describe the total metal composition (metallome) of Pseudomonas aeruginosa. Inductively coupled plasma atomic emission spectroscopy analyses showed that P. aeruginosa cells concentrate each metal of the metallome from the extracellular media with different efficiencies. Growth in nutrient-restricted media did not substantially affect the overall profile of the metallome; however, the uptake of some metals was strongly stimulated, showing the high potential of some metal acquisition pathways to adapt to changing growth conditions. We also investigated the role of the two major siderophores produced by P. aeruginosa, pyoverdine and pyochelin, in iron uptake and more generally in metallome homeostasis. In addition to their role in iron acquisition, siderophore production also significantly prevented the accumulation of toxic metals in P. aeruginosa cells, thus preserving the equilibrium of the metallome in a polluted environment.


Subject(s)
Metals/metabolism , Oligopeptides/metabolism , Phenols/metabolism , Pseudomonas aeruginosa/metabolism , Siderophores/metabolism , Thiazoles/metabolism , Biological Transport, Active , Metals/pharmacology , Spectrophotometry, Atomic
15.
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
16.
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
17.
EMBO J ; 31(9): 2134-43, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22473210

ABSTRACT

The opening of ligand-gated ion channels in response to agonist binding is a fundamental process in biology. In ATP-gated P2X receptors, little is known about the molecular events that couple ATP binding to channel opening. In this paper, we identify structural changes of the ATP site accompanying the P2X2 receptor activation by engineering extracellular zinc bridges at putative mobile regions as revealed by normal mode analysis. We provide evidence that tightening of the ATP sites shaped like open 'jaws' induces opening of the P2X ion channel. We show that ATP binding favours jaw tightening, whereas binding of a competitive antagonist prevents gating induced by this movement. Our data reveal the inherent dynamic of the binding jaw, and provide new structural insights into the mechanism of P2X receptor activation.


Subject(s)
Adenosine Triphosphate/physiology , Receptors, Purinergic P2X2/physiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Binding Sites , HEK293 Cells , Humans , Protein Binding , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...