Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Cell ; 143(7): 1149-60, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21183077

ABSTRACT

Following pilus-mediated adhesion to human brain endothelial cells, meningococcus (N. meningitidis), the bacterium causing cerebrospinal meningitis, initiates signaling cascades, which eventually result in the opening of intercellular junctions, allowing meningeal colonization. The signaling receptor activated by the pathogen remained unknown. We report that N. meningitidis specifically stimulates a biased ß2-adrenoceptor/ß-arrestin signaling pathway in endothelial cells, which ultimately traps ß-arrestin-interacting partners, such as the Src tyrosine kinase and junctional proteins, under bacterial colonies. Cytoskeletal reorganization mediated by ß-arrestin-activated Src stabilizes bacterial adhesion to endothelial cells, whereas ß-arrestin-dependent delocalization of junctional proteins results in anatomical gaps used by bacteria to penetrate into tissues. Activation of ß-adrenoceptor endocytosis with specific agonists prevents signaling events downstream of N. meningitidis adhesion and inhibits bacterial crossing of the endothelial barrier. The identification of the mechanism used for hijacking host cell signaling machineries opens perspectives for treatment and prevention of meningococcal infection.


Subject(s)
Arrestins/metabolism , Brain/microbiology , Endothelial Cells/microbiology , Meningococcal Infections/metabolism , Neisseria meningitidis/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Bacterial Adhesion , Blood-Brain Barrier , Cell Line , Humans , Meningococcal Infections/microbiology , beta-Arrestins
2.
Appl Environ Microbiol ; 90(9): e0088024, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39140741

ABSTRACT

The efficient natural transformation of Neisseria meningitidis allows the rapid construction of bacterial mutants in which the genes of interest are interrupted or replaced by antibiotic-resistance cassettes. However, this proved to be a double-edged sword, i.e., although facilitating the genetic characterization of this important human pathogen, it has limited the development of strategies for constructing markerless mutants without antibiotic-resistance markers. In addition, efficient tools for complementation or labeling are also lacking in N. meningitidis. In this study, we significantly expand the meningococcal genetic toolbox by developing new and efficient tools for the construction of markerless mutants (using a dual counterselection strategy), genetic complementation (using integrative vectors), and cell labeling (using a self-labeling protein tag). This expanded toolbox paves the way for more in-depth genetic characterization of N. meningitidis and might also be useful in other Neisseria species.IMPORTANCENeisseria meningitidis and Neisseria gonorrhoeae are two important human pathogens. Research focusing on these bacteria requires genetic engineering, which is facilitated by their natural ability to undergo transformation. However, the ease of mutant engineering has led the Neisseria community to neglect the development of more sophisticated tools for gene editing, particularly for N. meningitidis. In this study, we have significantly expanded the meningococcal genetic toolbox by developing novel and efficient tools for markerless mutant construction, genetic complementation, and cell tagging. This expanded toolbox paves the way for more in-depth genetic characterization of N. meningitidis and might also be useful in other Neisseria species.


Subject(s)
Gene Editing , Neisseria meningitidis , Neisseria meningitidis/genetics , Gene Editing/methods , Genetic Complementation Test
3.
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
4.
PLoS Pathog ; 17(2): e1009299, 2021 02.
Article in English | MEDLINE | ID: mdl-33592056

ABSTRACT

Neisseria meningitidis (the meningococcus) remains a major cause of bacterial meningitis and fatal sepsis. This commensal bacterium of the human nasopharynx can cause invasive diseases when it leaves its niche and reaches the bloodstream. Blood-borne meningococci have the ability to adhere to human endothelial cells and rapidly colonize microvessels. This crucial step enables dissemination into tissues and promotes deregulated inflammation and coagulation, leading to extensive necrotic purpura in the most severe cases. Adhesion to blood vessels relies on type IV pili (TFP). These long filamentous structures are highly dynamic as they can rapidly elongate and retract by the antagonistic action of two ATPases, PilF and PilT. However, the consequences of TFP dynamics on the pathophysiology and the outcome of meningococcal sepsis in vivo have been poorly studied. Here, we show that human graft microvessels are replicative niches for meningococci, that seed the bloodstream and promote sustained bacteremia and lethality in a humanized mouse model. Intriguingly, although pilus-retraction deficient N. meningitidis strain (ΔpilT) efficiently colonizes human graft tissue, this mutant did not promote sustained bacteremia nor induce mouse lethality. This effect was not due to a decreased inflammatory response, nor defects in bacterial clearance by the innate immune system. Rather, TFP-retraction was necessary to promote the release of TFP-dependent contacts between bacteria and, in turn, the detachment from colonized microvessels. The resulting sustained bacteremia was directly correlated with lethality. Altogether, these results demonstrate that pilus retraction plays a key role in the occurrence and outcome of meningococcal sepsis by supporting sustained bacteremia. These findings open new perspectives on the role of circulating bacteria in the pathological alterations leading to lethal sepsis.


Subject(s)
Bacteremia/microbiology , Disease Models, Animal , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/physiology , Meningococcal Infections/microbiology , Neisseria meningitidis/pathogenicity , Sepsis/microbiology , Animals , Bacteremia/metabolism , Bacteremia/pathology , Bacterial Adhesion , Endothelial Cells , Female , Fimbriae Proteins/genetics , Humans , Meningococcal Infections/metabolism , Meningococcal Infections/pathology , Mice , Mice, SCID , Sepsis/metabolism , Sepsis/pathology , Skin Transplantation
5.
PLoS Pathog ; 17(8): e1009326, 2021 08.
Article in English | MEDLINE | ID: mdl-34339477

ABSTRACT

Metabolic pathways are now considered as intrinsic virulence attributes of pathogenic bacteria and thus represent potential targets for antibacterial strategies. Here we focused on the role of the pentose phosphate pathway (PPP) and its connections with other metabolic pathways in the pathophysiology of Francisella novicida. The involvement of the PPP in the intracellular life cycle of Francisella was first demonstrated by studying PPP inactivating mutants. Indeed, we observed that inactivation of the tktA, rpiA or rpe genes severely impaired intramacrophage multiplication during the first 24 hours. However, time-lapse video microscopy demonstrated that rpiA and rpe mutants were able to resume late intracellular multiplication. To better understand the links between PPP and other metabolic networks in the bacterium, we also performed an extensive proteo-metabolomic analysis of these mutants. We show that the PPP constitutes a major bacterial metabolic hub with multiple connections to glycolysis, the tricarboxylic acid cycle and other pathways, such as fatty acid degradation and sulfur metabolism. Altogether our study highlights how PPP plays a key role in the pathogenesis and growth of Francisella in its intracellular niche.


Subject(s)
Bacterial Proteins/metabolism , Drosophila melanogaster/metabolism , Francisella/pathogenicity , Gram-Negative Bacterial Infections/microbiology , Metabolome , Pentose Phosphate Pathway , Proteome , Animals , Bacterial Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/microbiology , Francisella/metabolism , Gene Expression Regulation, Bacterial , Glycolysis , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mutation
6.
Proc Natl Acad Sci U S A ; 117(5): 2606-2612, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31964828

ABSTRACT

Bacterial infections are frequently based on the binding of lectin-like adhesins to specific glycan determinants exposed on host cell receptors. These interactions confer species-specific recognition and tropism for particular host tissues and represent attractive antibacterial targets. However, the wide structural diversity of carbohydrates hampers the characterization of specific glycan determinants. Here, we characterized the receptor recognition of type IV pili (Tfp), a key adhesive factor present in numerous bacterial pathogens, using Neisseria meningitidis as a model organism. We found that meningococcal Tfp specifically recognize a triantennary sialylated poly-N-acetyllactosamine-containing N-glycan exposed on the human receptor CD147/Basigin, while fucosylated derivatives of this N-glycan impaired bacterial adhesion. Corroborating the inhibitory role of fucosylation on receptor recognition, adhesion of the meningococcus on nonhuman cells expressing human CD147 required prior defucosylation. These findings reveal the molecular basis of the selective receptor recognition by meningococcal Tfp and thereby, identify a potential antibacterial target.


Subject(s)
Adhesins, Bacterial/metabolism , Fimbriae Proteins/metabolism , Meningococcal Infections/metabolism , Neisseria meningitidis/metabolism , Receptors, Cell Surface/metabolism , Adhesins, Bacterial/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Glycosylation , Humans , Meningococcal Infections/genetics , Meningococcal Infections/microbiology , Neisseria meningitidis/genetics , Polysaccharides/metabolism , Receptors, Cell Surface/genetics
7.
J Infect Dis ; 226(7): 1276-1285, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35524969

ABSTRACT

BACKGROUND: Staphylococcus aureus dominates the lung microbiota of children with cystic fibrosis (CF) and persistent clones are able to establish chronic infection for years, having a direct deleterious impact on lung function. However, in this context, the exact contribution of S. aureus to the decline in respiratory function in children with CF is not elucidated. METHODS: To investigate the contribution of persistent S. aureus clones in CF disease, we undertook the analysis of sequential isogenic isolates recovered from 15 young CF patients. RESULTS: Using an air-liquid infection model, we observed a strong correlation between S. aureus adaption in the lung (late isolates), low toxicity, and proinflammatory cytokine secretion. Conversely, early isolates appeared to be highly cytotoxic but did not promote cytokine secretion. We found that cytokine secretion was dependent on staphylococcal protein A (Spa), which was selectively expressed in late compared to early isolates as a consequence of dysfunctional agr quorum-sensing system. Finally, we demonstrated the involvement of TNF-α receptor 1 signaling in the inflammatory response of airway epithelial cells to these lung-adapted S. aureus isolates. CONCLUSIONS: Our results suggest an unexpected direct role of bacterial lung adaptation in the progression of chronic lung disease by promoting a proinflammatory response through acquired agr dysfunction.


Subject(s)
Cystic Fibrosis , Staphylococcal Infections , Child , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Humans , Lung/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Protein A , Staphylococcus aureus/physiology , Tumor Necrosis Factor-alpha
8.
J Clin Microbiol ; 59(3)2021 02 18.
Article in English | MEDLINE | ID: mdl-33328176

ABSTRACT

Staphylococcus epidermidis is a pathogen emerging worldwide as a leading cause of health care-associated infections. A standardized high-resolution typing method to document transmission and dissemination of multidrug-resistant S. epidermidis strains is needed. Our aim was to provide a core genome multilocus sequence typing (cgMLST) scheme for S. epidermidis to improve the international surveillance of S. epidermidis We defined a cgMLST scheme based on 699 core genes and used it to investigate the population structure of the species and the genetic relatedness of isolates recovered from infants hospitalized in several wards of a French hospital. Our results show the long-lasting endemic persistence of S. epidermidis clones within and across wards of hospitals and demonstrate the ability of our cgMLST approach to identify and track these clones. We made the scheme publicly available through the Institut Pasteur BIGSdb server (http://bigsdb.pasteur.fr/epidermidis/). This tool should enable international harmonization of the epidemiological surveillance of multidrug-resistant S. epidermidis clones. By comparing gene distribution among infection and commensal isolates, we also confirmed the association of the mecA locus with infection isolates and of the fdh gene with commensal isolates. (This study has been registered at ClinicalTrials.gov under registration no. NCT03374371.).


Subject(s)
Staphylococcal Infections , Staphylococcus epidermidis , Clone Cells , Genome, Bacterial/genetics , Hospitals , Humans , Multilocus Sequence Typing , Staphylococcal Infections/epidemiology , Staphylococcus epidermidis/genetics
9.
Cell Microbiol ; 22(1): e13132, 2020 01.
Article in English | MEDLINE | ID: mdl-31658405

ABSTRACT

The skull, spine, meninges, and cellular barriers at the blood-brain and the blood-cerebrospinal fluid interfaces well protect the brain and meningeal spaces against microbial invasion. However, once in the bloodstream, a range of pathogenic bacteria is able to reach the brain and cause meningitis. Despite advances in antibacterial therapy, bacterial meningitis remains one of the most important infectious diseases worldwide. The most common causative bacteria in children and adults are Streptococcus pneumoniae and Neisseria meningitidis associated with high morbidity and mortality, while among neonates, most cases of bacterial meningitis are due to group B Streptococcus and Escherichia coli. Here we summarise our current knowledge on the strategies used by these bacterial pathogens to survive in the bloodstream, to colonise the brain vasculature and to cross the blood-brain barrier.


Subject(s)
Bacteria/pathogenicity , Blood-Brain Barrier/microbiology , Animals , Biological Transport , Brain/microbiology , Endothelial Cells/microbiology , Humans , Inflammation , Neisseria meningitidis/pathogenicity , Neisseria meningitidis/physiology , Streptococcus pneumoniae/pathogenicity , Streptococcus pneumoniae/physiology , Virulence Factors
10.
Cell Microbiol ; 22(4): e13185, 2020 04.
Article in English | MEDLINE | ID: mdl-32185901

ABSTRACT

Neisseria meningitidis (meningococcus) is a Gram-negative bacterium responsible for two devastating forms of invasive diseases: purpura fulminans and meningitis. Interaction with both peripheral and cerebral microvascular endothelial cells is at the heart of meningococcal pathogenesis. During the last two decades, an essential role for meningococcal type IV pili in vascular colonisation and disease progression has been unravelled. This review summarises 20 years of research on meningococcal type IV pilus-dependent virulence mechanisms, up to the identification of promising anti-virulence compounds that target type IV pili.


Subject(s)
Bacterial Adhesion , Fimbriae, Bacterial/classification , Fimbriae, Bacterial/metabolism , Meningococcal Infections/microbiology , Neisseria meningitidis/pathogenicity , Animals , Endothelial Cells/microbiology , Humans , Mice , Virulence
11.
Mol Cell Proteomics ; 18(12): 2418-2432, 2019 12.
Article in English | MEDLINE | ID: mdl-31578219

ABSTRACT

The bacterial pathogen Francisella tularensis possesses a noncanonical type VI secretion system (T6SS) that is required for phagosomal escape in infected macrophages. KCl stimulation has been previously used to trigger assembly and secretion of the T6SS in culture. By differential proteomics, we found here that the amounts of the T6SS proteins remained unchanged upon KCl stimulation, suggesting involvement of post-translational modifications in T6SS assembly. A phosphoproteomic analysis indeed identified a unique phosphorylation site on IglB, a key component of the T6SS sheath. Substitutions of Y139 with alanine or phosphomimetics prevented T6SS formation and abolished phagosomal escape whereas substitution with phenylalanine delayed but did not abolish phagosomal escape in J774-1 macrophages. Altogether our data demonstrated that the Y139 site of IglB plays a critical role in T6SS biogenesis, suggesting that sheath phosphorylation could participate to T6SS dynamics.Data are available via ProteomeXchange with identifier PXD013619; and on MS-Viewer, key lkaqkllxwx.


Subject(s)
Francisella tularensis/metabolism , Type VI Secretion Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Electronic Data Processing , Francisella tularensis/genetics , Francisella tularensis/ultrastructure , Gas Chromatography-Mass Spectrometry , Humans , Macrophages/microbiology , Molecular Structure , Mutagenesis, Site-Directed , Phosphorylation , Potassium Chloride/pharmacology , Protein Processing, Post-Translational , Proteomics , Tandem Mass Spectrometry , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/drug effects , Type VI Secretion Systems/genetics
12.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Article in English | MEDLINE | ID: mdl-29630665

ABSTRACT

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Endothelial Protein C Receptor/metabolism , Endothelium, Vascular/pathology , Membrane Proteins/metabolism , Meningococcal Infections/complications , Microvessels/pathology , Protein C/metabolism , Purpura Fulminans/etiology , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Bacterial Adhesion , Blood Coagulation/physiology , Cells, Cultured , Endothelial Protein C Receptor/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology , Humans , Membrane Proteins/genetics , Meningococcal Infections/microbiology , Microvessels/metabolism , Microvessels/microbiology , Neisseria meningitidis/physiology , Protein C/genetics , Purpura Fulminans/metabolism , Purpura Fulminans/pathology
13.
Cell Microbiol ; 21(11): e13063, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31167044

ABSTRACT

Neisseria meningitidis is a Gram-negative bacterium that asymptomatically colonises the nasopharynx of humans. For an unknown reason, N. meningitidis can cross the nasopharyngeal barrier and invade the bloodstream where it becomes one of the most harmful extracellular bacterial pathogen. This infectious cycle involves the colonisation of two different environments. (a) In the nasopharynx, N. meningitidis grow on the top of mucus-producing epithelial cells surrounded by a complex microbiota. To survive and grow in this challenging environment, the meningococcus expresses specific virulence factors such as polymorphic toxins and MDAΦ. (b) Meningococci have the ability to survive in the extra cellular fluids including blood and cerebrospinal fluid. The interaction of N. meningitidis with human endothelial cells leads to the formation of typical microcolonies that extend overtime and promote vascular injury, disseminated intravascular coagulation, and acute inflammation. In this review, we will focus on the interplay between N. meningitidis and these two different niches at the cellular and molecular level and discuss the use of inhibitors of piliation as a potent therapeutic approach.


Subject(s)
Meningococcal Infections/microbiology , Nasopharynx/microbiology , Neisseria meningitidis/pathogenicity , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Blood Vessels/microbiology , Endothelial Cells/pathology , Epithelial Cells/pathology , Host Microbial Interactions , Humans , Inovirus/growth & development , Inovirus/pathogenicity , Meningococcal Infections/blood , Meningococcal Infections/cerebrospinal fluid , Neisseria meningitidis/metabolism , Virulence Factors
14.
J Infect Dis ; 220(12): 1967-1976, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31420648

ABSTRACT

Staphylococcus aureus is a leading cause of both acute and chronic infections in humans. The importance of the pentose phosphate pathway (PPP) during S. aureus infection is currently largely unexplored. In the current study, we focused on one key PPP enzyme, transketolase (TKT). We showed that inactivation of the unique gene encoding TKT activity in S. aureus USA300 (∆tkt) led to drastic metabolomic changes. Using time-lapse video imaging and mice infection, we observed a major defect of the ∆tkt strain compared with wild-type strain in early intracellular proliferation and in the ability to colonize kidneys. Transcriptional activity of the 2 master regulators sigma B and RpiRc was drastically reduced in the ∆tkt mutant during host cells invasion. The concomitant increased RNAIII transcription suggests that TKT-or a functional PPP-strongly influences the ability of S. aureus to proliferate within host cells by modulating key transcriptional regulators.


Subject(s)
Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Stress, Physiological , Transketolase/metabolism , Animals , Carbon/metabolism , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation, Bacterial , Gene Silencing , Genes, Bacterial , Humans , Kidney/metabolism , Kidney/microbiology , Metabolomics/methods , Mice , Mutation , Phenotype , Signal Transduction , Staphylococcus aureus/enzymology , Stress, Physiological/genetics , Transketolase/genetics
15.
Clin Infect Dis ; 69(11): 1937-1945, 2019 11 13.
Article in English | MEDLINE | ID: mdl-30753350

ABSTRACT

BACKGROUND: Chronic lung infection in cystic fibrosis (CF) patients by Staphylococcus aureus is a well-established epidemiological fact. Indeed, S. aureus is the most commonly identified pathogen in the lungs of CF patients. Improving our understanding of the mechanisms associated with the persistence of S. aureus is therefore an important issue. METHODS: We selected pairs of sequential S. aureus isolates from 3 patients with CF and from 1 patient with non-CF chronic lung disease. We used a combination of genomic, proteomic, and metabolomic approaches with functional assays for in-depth characterization of S. aureus long-term persistence. RESULTS: In this study, we show that late S. aureus isolates from CF patients have an increased ability for intracellular survival in CF bronchial epithelial-F508del cells compared to ancestral early isolates. Importantly, the increased ability to persist intracellularly was confirmed for S. aureus isolates within the own-patient F508del epithelial cells. An increased ability to form biofilm was also demonstrated. Furthermore, we identified the underlying genetic modifications that induce altered protein expression profiles and notable metabolic changes. These modifications affect several metabolic pathways and virulence regulators that could constitute therapeutic targets. CONCLUSIONS: Our results strongly suggest that the intracellular environment might constitute an important niche of persistence and relapse necessitating adapted antibiotic treatments.


Subject(s)
Staphylococcus aureus/drug effects , Adaptation, Physiological/drug effects , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Cell Line , Cells, Cultured , Chromatography, Liquid , Humans , Proteogenomics/methods , Proteomics/methods , Tandem Mass Spectrometry
16.
Infect Immun ; 84(10): 3017-23, 2016 10.
Article in English | MEDLINE | ID: mdl-27481255

ABSTRACT

Meningococcal septic shock is associated with profound vasoplegia, early and severe myocardial dysfunction, and extended skin necrosis responsible for a specific clinical entity designated purpura fulminans (PF). PF represents 90% of fatal meningococcal infections. One characteristic of meningococcal PF is the myocardial dysfunction that occurs in the early phase of sepsis. Furthermore, hemodynamic studies have shown that the prognosis of meningococcal sepsis is directly related to the degree of impairment of cardiac contractility during the initial phase of the disease. To gain insight into a potential interaction of Neisseria meningitidis with the myocardial microvasculature, we modified a previously described humanized mouse model by grafting human myocardial tissue to SCID mice. We then infected the grafted mice with N. meningitides Using the humanized SCID mouse model, we demonstrated that N. meningitidis targets the human myocardial tissue vasculature, leading to the formation of blood thrombi, infectious vasculitis, and vascular leakage. These results suggest a novel mechanism of myocardial injury in the course of severe N. meningitidis sepsis that is likely to participate in primary myocardial dysfunction.


Subject(s)
Heart/microbiology , Meningococcal Infections/microbiology , Microvessels/microbiology , Animals , Bacteremia/microbiology , Disease Models, Animal , Endothelial Cells/microbiology , Endothelial Cells/pathology , Female , Humans , Meningococcal Infections/pathology , Mice, SCID , Myocardium , Neisseria meningitidis , Shock, Septic/blood , Vasculitis/pathology , Venous Thrombosis/pathology
17.
Cell Microbiol ; 15(4): 512-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23189983

ABSTRACT

The brain and meningeal spaces are protected from bacterial invasion by the blood-brain barrier, formed by specialized endothelial cells and tight intercellular junctional complexes. However, once in the bloodstream, Neisseria meningitidis crosses this barrier in about 60% of the cases. This highlights the particular efficacy with which N. meningitidis targets the brain vascular cell wall. The first step of central nervous system invasion is the direct interaction between bacteria and endothelial cells. This step is mediated by the type IV pili, which induce a remodelling of the endothelial monolayer, leading to the opening of the intercellular space. In this review, strategies used by the bacteria to survive in the bloodstream, to colonize the brain vasculature and to cross the blood-brain barrier will be discussed.


Subject(s)
Blood-Brain Barrier/microbiology , Brain/microbiology , Cerebrospinal Fluid/microbiology , Endothelium/microbiology , Neisseria meningitidis/physiology , Blood-Brain Barrier/immunology , Fimbriae, Bacterial/metabolism , Host-Pathogen Interactions , Neisseria meningitidis/growth & development
18.
Handb Exp Pharmacol ; 219: 361-74, 2014.
Article in English | MEDLINE | ID: mdl-24292839

ABSTRACT

In the context of host-pathogen interaction, host cell receptors and signaling pathways are essential for both invading pathogens, which exploit them for their own profit, and the defending organism, which activates early mechanism of defense, known as innate immunity, to block the aggression. Because of their central role as scaffolding proteins downstream of activated receptors, ß-arrestins are involved in multiple signaling pathways activated in host cells by pathogens. Some of these pathways participate in the innate immunity and the inflammatory response. Other ß-arrestin-dependent pathways are actually hijacked by microbes and toxins to penetrate into host cells and spread in the organism.


Subject(s)
Arrestins/metabolism , Host-Pathogen Interactions/physiology , Signal Transduction/physiology , Animals , Bacteria/pathogenicity , Humans , Immunity, Innate/physiology , Viruses/pathogenicity , beta-Arrestins
19.
J Infect Dis ; 208(10): 1590-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23840047

ABSTRACT

Neisseria meningitidis is a strict human pathogen that closely interacts with human endothelial cells via type IV pili in vitro. To decipher whether this interaction plays a role in vivo, we set up an experimental model of fulminant meningococcemia in human skin grafted SCID mice using the wild-type strain 2C4.3. Human skin and mouse tissues were sampled 24 hours after bacterial challenge for histopathology, immunohistochemistry and ultrastructural analysis. In all infected mice, N. meningitidis targeted the human vasculature, leading to bacterial and blood thrombi, infectious vasculitis and vascular leakage. Mouse vessels, including brain vessels, remained unaffected by the infectious and thrombotic process, and a nonpiliated Δ pilE derivative of 2C4.3 failed to target human graft vessels and to induce vascular damages. These data demonstrate that N. meningitidis targets human endothelial cells in vivo and that this interaction triggers the vascular damages that characterize purpura fulminans.


Subject(s)
Microvessels/microbiology , Neisseria meningitidis/physiology , Purpura Fulminans/etiology , Purpura Fulminans/pathology , Animals , Bacterial Adhesion , Endothelial Cells/microbiology , Endothelial Cells/pathology , Endothelium, Vascular/microbiology , Endothelium, Vascular/pathology , Female , Fimbriae, Bacterial/physiology , Heterografts , Humans , Meningococcal Infections/complications , Meningococcal Infections/microbiology , Mice , Microvessels/pathology , Skin/blood supply , Skin/pathology , Skin Transplantation
20.
Sci Rep ; 14(1): 7797, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38565565

ABSTRACT

Bacterial pathogens adapt and replicate within host cells, while host cells develop mechanisms to eliminate them. Using a dual proteomic approach, we characterized the intra-macrophage proteome of the facultative intracellular pathogen, Francisella novicida. More than 900 Francisella proteins were identified in infected macrophages after a 10-h infection. Biotin biosynthesis-related proteins were upregulated, emphasizing the role of biotin-associated genes in Francisella replication. Conversely, proteins encoded by the Francisella pathogenicity island (FPI) were downregulated, supporting the importance of the F. tularensis Type VI Secretion System for vacuole escape, not cytosolic replication. In the host cell, over 300 proteins showed differential expression among the 6200 identified during infection. The most upregulated host protein was cis-aconitate decarboxylase IRG1, known for itaconate production with antimicrobial properties in Francisella. Surprisingly, disrupting IRG1 expression did not impact Francisella's intracellular life cycle, suggesting redundancy with other immune proteins or inclusion in larger complexes. Over-representation analysis highlighted cell-cell contact and actin polymerization in macrophage deregulated proteins. Using flow cytometry and live cell imaging, we demonstrated that merocytophagy involves diverse cell-to-cell contacts and actin polymerization-dependent processes. These findings lay the groundwork for further exploration of merocytophagy and its molecular mechanisms in future research.Data are available via ProteomeXchange with identifier PXD035145.


Subject(s)
Francisella tularensis , Tularemia , Animals , Francisella tularensis/genetics , Actins/metabolism , Biotin/metabolism , Proteomics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Macrophages/metabolism , Life Cycle Stages , Tularemia/microbiology , Genomic Islands
SELECTION OF CITATIONS
SEARCH DETAIL