Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34725157

ABSTRACT

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/physiology , Fimbriae, Bacterial/physiology , Neisseria meningitidis/physiology , Animals , Antibodies/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Cell Line , Drug Evaluation, Preclinical , Female , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/ultrastructure , Humans , Meningococcal Infections/drug therapy , Mice, SCID
2.
Structure ; 25(9): 1423-1435.e4, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28877506

ABSTRACT

We report here cryoelectron microscopy reconstructions of type IV pili (T4P) from two important human pathogens, Pseudomonas aeruginosa and Neisseria gonorrhoeae, at ∼ 8 and 5 Å resolution, respectively. The two structures reveal distinct arrangements of the pilin globular domains on the pilus surfaces, which impart different helical parameters, but similar packing of the conserved N-terminal α helices, α1, in the filament core. In contrast to the continuous α helix seen in the X-ray crystal structures of the P. aeruginosa and N. gonorrhoeae pilin subunits, α1 in the pilus filaments has a melted segment located between conserved helix-breaking residues Gly14 and Pro22, as seen for the Neisseria meningitidis T4P. Using mutagenesis we show that Pro22 is critical for pilus assembly, as are Thr2 and Glu5, which are positioned to interact in the hydrophobic filament core. These structures provide a framework for understanding T4P assembly, function, and biophysical properties.


Subject(s)
Cryoelectron Microscopy/methods , Fimbriae Proteins/chemistry , Neisseria gonorrhoeae/ultrastructure , Pseudomonas aeruginosa/metabolism , Amino Acid Sequence , Fimbriae Proteins/genetics , Fimbriae, Bacterial/ultrastructure , Models, Molecular , Mutation , Neisseria gonorrhoeae/genetics , Protein Structure, Secondary , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/ultrastructure
3.
Article in English | MEDLINE | ID: mdl-25705612

ABSTRACT

Francisella tularensis, the agent of the zoonotic disease tularemia, is a highly infectious bacterium for a large number of animal species and can be transmitted to humans by various means. The bacterium is able to infect a variety of cell types but replicates in mammalian hosts mainly in the cytosol of infected macrophages. In order to resist the stressful and nutrient-restricted intracellular environments, it encounters during its systemic dissemination, Francisella has developed dedicated stress resistance mechanisms and adapted its metabolic and nutritional needs. Recent data form our laboratory and from several other groups have shown that Francisella simultaneously relies on multiple host amino acid sources during its intracellular life cycle. This review will summarize how intracellular Francisella use different amino acid sources, and their role in phagosomal escape and/or cytosolic multiplication and systemic dissemination. We will first summarize the data that we have obtained on two amino acid transporters involved in Francisella phagosomal escape and cytosolic multiplication i.e., the glutamate transporter GadC and the asparagine transporter AnsP, respectively. The specific contribution of glutamate and asparagine to the physiology of the bacterium will be evoked. Then, we will discuss how Francisella has adapted to obtain and utilize host amino acid resources, and notably the contribution of host transporters and autophagy process in the establishment of a nutrient-replete intracellular niche.


Subject(s)
Amino Acids/metabolism , Francisella tularensis/metabolism , Macrophages/microbiology , Tularemia/microbiology , Animals , Francisella , Francisella tularensis/genetics , Humans , Macrophages/metabolism , Tularemia/metabolism
4.
Mol Cell Proteomics ; 14(4): 870-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25616868

ABSTRACT

Upon entry into mammalian host cells, the pathogenic bacterium Francisella must import host cell arginine to multiply actively in the host cytoplasm. We identified and functionally characterized an arginine transporter (hereafter designated ArgP) whose inactivation considerably delayed bacterial phagosomal escape and intracellular multiplication. Intramacrophagic growth of the ΔargP mutant was fully restored upon supplementation of the growth medium with excess arginine, in both F. tularensis subsp. novicida and F. tularensis subsp. holarctica LVS, demonstrating the importance of arginine acquisition in these two subspecies. High-resolution mass spectrometry revealed that arginine limitation reduced the amount of most of the ribosomal proteins in the ΔargP mutant. In response to stresses such as nutritional limitation, repression of ribosomal protein synthesis has been observed in all kingdoms of life. Arginine availability may thus contribute to the sensing of the intracellular stage of the pathogen and to trigger phagosomal egress. All MS data have been deposited in the ProteomeXchange database with identifier PXD001584 (http://proteomecentral.proteomexchange.org/dataset/PXD001584).


Subject(s)
Arginine/metabolism , Francisella/metabolism , Host-Pathogen Interactions , Phagosomes/microbiology , Ribosomal Proteins/metabolism , Animals , Autophagy , Bacterial Proteins/metabolism , Bacterial Vaccines/immunology , Cluster Analysis , Cytosol/metabolism , Female , Francisella/pathogenicity , Macrophages/metabolism , Macrophages/microbiology , Macrophages/ultrastructure , Membrane Transport Proteins/metabolism , Mice, Inbred BALB C , Microbial Viability , Models, Biological , Mutation/genetics , Phagosomes/metabolism , Phagosomes/ultrastructure , Protein Transport , Proteome/metabolism , Stress, Physiological , Subcellular Fractions/metabolism , Virulence
5.
Infect Immun ; 83(1): 173-83, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332124

ABSTRACT

Intracellular bacterial pathogens have adapted their metabolism to optimally utilize the nutrients available in infected host cells. We recently reported the identification of an asparagine transporter required specifically for cytosolic multiplication of Francisella. In the present work, we characterized a new member of the major super family (MSF) of transporters, involved in isoleucine uptake. We show that this transporter (here designated IleP) plays a critical role in intracellular metabolic adaptation of Francisella. Inactivation of IleP severely impaired intracellular F. tularensis subsp. novicida multiplication in all cell types tested and reduced bacterial virulence in the mouse model. To further establish the importance of the ileP gene in F. tularensis pathogenesis, we constructed a chromosomal deletion mutant of ileP (ΔFTL_1803) in the F. tularensis subsp. holarctica live vaccine strain (LVS). Inactivation of IleP in the F. tularensis LVS provoked comparable intracellular growth defects, confirming the critical role of this transporter in isoleucine uptake. The data presented establish, for the first time, the importance of isoleucine utilization for efficient phagosomal escape and cytosolic multiplication of Francisella and suggest that virulent F. tularensis subspecies have lost their branched-chain amino acid biosynthetic pathways and rely exclusively on dedicated uptake systems. This loss of function is likely to reflect an evolution toward a predominantly intracellular life style of the pathogen. Amino acid transporters should be thus considered major players in the adaptation of intracellular pathogens.


Subject(s)
Adaptation, Physiological , Francisella tularensis/physiology , Isoleucine/metabolism , Membrane Transport Proteins/metabolism , Animals , Cytosol/microbiology , Disease Models, Animal , Female , Francisella tularensis/genetics , Francisella tularensis/growth & development , Francisella tularensis/metabolism , Gene Deletion , Membrane Transport Proteins/genetics , Mice, Inbred BALB C , Phagosomes/microbiology , Tularemia/microbiology , Tularemia/pathology
6.
PLoS Pathog ; 10(1): e1003893, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24453979

ABSTRACT

Intracellular bacterial pathogens have developed a variety of strategies to avoid degradation by the host innate immune defense mechanisms triggered upon phagocytocis. Upon infection of mammalian host cells, the intracellular pathogen Francisella replicates exclusively in the cytosolic compartment. Hence, its ability to escape rapidly from the phagosomal compartment is critical for its pathogenicity. Here, we show for the first time that a glutamate transporter of Francisella (here designated GadC) is critical for oxidative stress defense in the phagosome, thus impairing intra-macrophage multiplication and virulence in the mouse model. The gadC mutant failed to efficiently neutralize the production of reactive oxygen species. Remarkably, virulence of the gadC mutant was partially restored in mice defective in NADPH oxidase activity. The data presented highlight links between glutamate uptake, oxidative stress defense, the tricarboxylic acid cycle and phagosomal escape. This is the first report establishing the role of an amino acid transporter in the early stage of the Francisella intracellular lifecycle.


Subject(s)
Citric Acid Cycle , Francisella tularensis/metabolism , Glutamic Acid/metabolism , Macrophages/microbiology , Phagosomes/metabolism , Tularemia/metabolism , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Female , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Glutamic Acid/genetics , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mutation , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phagosomes/genetics , Phagosomes/microbiology , Phagosomes/pathology , Tularemia/genetics
7.
Cell Microbiol ; 16(3): 434-49, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134488

ABSTRACT

In order to develop a successful infectious cycle, intracellular bacterial pathogens must be able to adapt their metabolism to optimally utilize the nutrients available in the cellular compartments and tissues where they reside. Francisella tularensis, the agent of the zoonotic disease tularaemia, is a highly infectious bacterium for a large number of animal species. This bacterium replicates in its mammalian hosts mainly in the cytosol of infected macrophages. We report here the identification of a novel amino acid transporter of the major facilitator superfamily of secondary transporters that is required for bacterial intracellular multiplication and systemic dissemination. We show that inactivation of this transporter does not affect phagosomal escape but prevents multiplication in the cytosol of all cell types tested. Remarkably, the intracellular growth defect of the mutant was fully and specifically reversed by addition of asparagine or asparagine-containing dipeptides as well as by simultaneous addition of aspartic acid and ammonium. Importantly, bacterial virulence was also restored in vivo, in the mouse model, by asparagine supplementation. This work unravels thus, for the first time, the importance of asparagine for cytosolicmultiplication of Francisella. Amino acid transporters are likely to constitute underappreciated players in bacterial intracellular parasitism.


Subject(s)
Amino Acid Transport Systems/genetics , Asparagine/metabolism , Bacterial Proteins/genetics , Francisella tularensis/growth & development , Ammonium Compounds/pharmacology , Animals , Asparagine/pharmacology , Aspartic Acid/metabolism , Aspartic Acid/pharmacology , Bacterial Proteins/pharmacokinetics , Cell Line, Tumor , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Hep G2 Cells , Humans , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Phagosomes/microbiology , Tularemia/microbiology
8.
Mol Cell Proteomics ; 12(8): 2278-92, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23669032

ABSTRACT

Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularemia. In vivo, this facultative intracellular bacterium survives and replicates mainly in the cytoplasm of infected cells. We have recently identified a genetic locus, designated moxR that is important for stress resistance and intramacrophage survival of F. tularensis. In the present work, we used tandem affinity purification coupled to mass spectrometry to identify in vivo interacting partners of three proteins encoded by this locus: the MoxR-like ATPase (FTL_0200), and two proteins containing motifs predicted to be involved in protein-protein interactions, bearing von Willebrand A (FTL_0201) and tetratricopeptide (FTL_0205) motifs. The three proteins were designated here for simplification, MoxR, VWA1, and TPR1, respectively. MoxR interacted with 31 proteins, including various enzymes. VWA1 interacted with fewer proteins, but these included the E2 component of 2-oxoglutarate dehydrogenase and TPR1. The protein TPR1 interacted with one hundred proteins, including the E1 and E2 subunits of both oxoglutarate and pyruvate dehydrogenase enzyme complexes, and their common E3 subunit. Remarkably, chromosomal deletion of either moxR or tpr1 impaired pyruvate dehydrogenase and oxoglutarate dehydrogenase activities, supporting the hypothesis of a functional role for the interaction of MoxR and TPR1 with these complexes. Altogether, this work highlights possible links between stress resistance and metabolism in F. tularensis virulence.


Subject(s)
Bacterial Proteins/metabolism , Citric Acid Cycle/physiology , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Stress, Physiological , Amino Acid Sequence , Animals , DNA, Bacterial/genetics , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Sequence Analysis, DNA , Tularemia
9.
Future Microbiol ; 7(11): 1255-68, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23075445

ABSTRACT

Francisella tularensis, the etiological agent of tularemia, is a member of the γ-proteobacteria class of Gram-negative bacteria. This highly virulent bacterium can infect a large range of mammalian species and has been recognized as a human pathogen for a century. F. tularensis is able to survive in vitro in a variety of cell types. In vivo, the bacterium replicates mainly in infected macrophages, using the cytoplasmic compartment as a replicative niche. To successfully adapt to this stressful environment, F. tularensis must simultaneously: produce and regulate the expression of a series of dedicated virulence factors; adapt its metabolic needs to the nutritional context of the host cytosol; and control the innate immune cytosolic surveillance pathways to avoid premature cell death. We will focus here on the secretion or release of bacterial proteins in the host, as well as on the envelope proteins, involved in bacterial survival inside macrophages.


Subject(s)
Francisella tularensis/physiology , Macrophages/microbiology , Tularemia/microbiology , Bacterial Proteins/metabolism , Cytosol/microbiology , Francisella tularensis/growth & development , Francisella tularensis/metabolism , Humans , Macrophages/immunology , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...