Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
J Fungi (Basel) ; 8(12)2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36547601

ABSTRACT

Bronchial epithelial cells (BEC) play a crucial role in innate immunity against inhaled fungi. Indeed, in response to microorganisms, BEC synthesize proinflammatory cytokines involved in the recruitment of neutrophils. We have recently shown that BEC exert antifungal activity against Aspergillus fumigatus by inhibiting filament growth. In the present study, we first analyzed the inflammatory and antifungal responses of BEC infected by several fungal species such as Aspergillus spp., Scedosporium apiospermum and Candida albicans, which are frequently isolated from the sputum of people with chronic pulmonary diseases. The airways of these patients, such as people with cystic fibrosis (pwCF), are mainly colonized by P. aeruginosa and secondary by fungal pathogens. We have previously demonstrated that BEC are capable of innate immune memory, allowing them to increase their inflammatory response against A. fumigatus following a previous contact with Pseudomonas aeruginosa flagellin. To identify the impact of bacteria exposure on BEC responses to other fungal infections, we extended the analysis of BEC innate immune memory to Aspergillus spp., Scedosporium apiospermum and Candida albicans infection. Our results show that BEC are able to recognize and respond to Aspergillus spp., S. apiospermum and C. albicans infection and that the modulation of BEC responses by pre-exposure to flagellin varies according to the fungal species encountered. Deepening our knowledge of the innate immune memory of BEC should open new therapeutic avenues to modulate the inflammatory response against polymicrobial infections observed in chronic pulmonary diseases such as CF.

2.
Front Immunol ; 12: 714027, 2021.
Article in English | MEDLINE | ID: mdl-34950129

ABSTRACT

In the coronavirus disease 2019 (COVID-19) health crisis, one major challenge is to identify the susceptibility factors of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) in order to adapt the recommendations for populations, as well as to reduce the risk of COVID-19 development in the most vulnerable people, especially patients with chronic respiratory diseases such as cystic fibrosis (CF). Airway epithelial cells (AECs) play a critical role in the modulation of both immune responses and COVID-19 severity. SARS-CoV-2 infects the airway through the receptor angiotensin-converting enzyme 2, and a host protease, transmembrane serine protease 2 (TMPRSS2), plays a major role in SARS-CoV-2 infectivity. Here, we show that Pseudomonas aeruginosa increases TMPRSS2 expression, notably in primary AECs with deficiency of the ion channel CF transmembrane conductance regulator (CFTR). Further, we show that the main component of P. aeruginosa flagella, the protein flagellin, increases TMPRSS2 expression in primary AECs and Calu-3 cells, through activation of Toll-like receptor-5 and p38 MAPK. This increase is particularly seen in Calu-3 cells deficient for CFTR and is associated with an intracellular increased level of SARS-CoV-2 infection, however, with no effect on the amount of virus particles released. Considering the urgency of the COVID-19 health crisis, this result may be of clinical significance for CF patients, who are frequently infected with and colonized by P. aeruginosa during the course of CF and might develop COVID-19.


Subject(s)
Cystic Fibrosis , Flagellin/metabolism , Pseudomonas Infections/complications , Respiratory Mucosa/virology , SARS-CoV-2/pathogenicity , Serine Endopeptidases/metabolism , Bacterial Proteins/metabolism , COVID-19/complications , Cells, Cultured , Humans , Pseudomonas aeruginosa , Respiratory Mucosa/metabolism
3.
Sci Rep ; 11(1): 16109, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373510

ABSTRACT

Scedosporium apiospermum is an emerging opportunistic fungal pathogen responsible for life-threatening infections in humans. Host-pathogen interactions often implicate lectins that have become therapeutic targets for the development of carbohydrate mimics for antiadhesive therapy. Here, we present the first report on the identification and characterization of a lectin from S. apiospermum named SapL1. SapL1 was found using bioinformatics as a homolog to the conidial surface lectin FleA from Aspergillus fumigatus known to play a role in the adhesion to host glycoconjugates present in human lung epithelium. In our strategy to obtain recombinant SapL1, we discovered the importance of osmolytes to achieve its expression in soluble form in bacteria. Analysis of glycan arrays indicates specificity for fucosylated oligosaccharides as expected. Submicromolar affinity was measured for fucose using isothermal titration calorimetry. We solved SapL1 crystal structure in complex with α-methyl-L-fucoside and analyzed its structural basis for fucose binding. We finally demonstrated that SapL1 binds to bronchial epithelial cells in a fucose-dependent manner. The information gathered here will contribute to the design and development of glycodrugs targeting SapL1.


Subject(s)
Fungal Proteins/metabolism , Lectins/metabolism , Scedosporium/metabolism , Amino Acid Sequence , Aspergillus fumigatus/metabolism , Binding Sites/physiology , Cells, Cultured , Epithelial Cells/metabolism , Fucose/metabolism , Glycoconjugates/metabolism , Host-Pathogen Interactions/physiology , Humans , Oligosaccharides/metabolism , Polysaccharides/metabolism
4.
Org Biomol Chem ; 19(14): 3234-3240, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33885578

ABSTRACT

Aspergillus fumigatus is a pathogenic fungus infecting the respiratory system and responsible for a variety of life-threatening lung diseases. A fucose-binding lectin named FleA which has a controversial role in A. fumigatus pathogenesis was recently identified. New chemical probes with high affinity and enzymatic stability are needed to explore the role of FleA in the infection process. In this study, we developed potent FleA antagonists based on optimized and non-hydrolysable thiofucoside ligands. We first synthesized a set of monovalent sugars showing micromolar affinity for FleA by isothermal titration calorimetry. The most potent derivative was co-crystallized with FleA to gain insights into the binding mode in operation. Its chemical multimerization on a cyclodextrin scaffold led to an hexavalent compound with a significantly enhanced binding affinity (Kd = 223 ± 21 nM) thanks to a chelate binding mode. The compound could probe the role of bronchial epithelial cells in a FleA-mediated response to tissue invasion.


Subject(s)
Aspergillus fumigatus/chemistry , Fucose/pharmacology , Lectins/antagonists & inhibitors , Sulfhydryl Compounds/pharmacology , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/pathogenicity , Dose-Response Relationship, Drug , Drug Design , Fucose/chemical synthesis , Fucose/chemistry , Lectins/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry
5.
Front Immunol ; 11: 1041, 2020.
Article in English | MEDLINE | ID: mdl-32528481

ABSTRACT

Aspergillus fumigatus is an environmental filamentous fungus that can be pathogenic for humans, wherein it is responsible for a large variety of clinical forms ranging from allergic diseases to life-threatening disseminated infections. The contamination occurs by inhalation of conidia present in the air, and the first encounter of this fungus in the human host is most likely with the bronchial epithelial cells. Although alveolar macrophages have been widely studied in the Aspergillus-lung interaction, increasing evidence suggests that bronchial epithelium plays a key role in responding to the fungus. This review focuses on the innate immune response of the bronchial epithelial cells against A. fumigatus, the predominant pathogenic species. We have also detailed the molecular interactants and the effects of the different modes of interaction between these cells and the fungus.


Subject(s)
Aspergillus fumigatus , Bronchi/immunology , Pulmonary Aspergillosis/immunology , Antimicrobial Cationic Peptides/biosynthesis , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Bronchi/microbiology , Cell Line , Cytokines/biosynthesis , Host Microbial Interactions/immunology , Humans , Immunity, Innate , Models, Immunological , Pathogen-Associated Molecular Pattern Molecules/immunology , Phagocytosis/immunology , Pulmonary Aspergillosis/microbiology , Receptors, Pattern Recognition/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Spores, Fungal/immunology , Spores, Fungal/pathogenicity
6.
J Infect Dis ; 221(6): 1000-1005, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31678998

ABSTRACT

Human bronchial epithelial cells play a key role in airway immune homeostasis. We hypothesized that these sentinel cells can remember a previous contact with pathogen compounds and respond nonspecifically to reinfection, a phenomenon called innate immune memory. We demonstrated that their preexposure to Pseudomonas aeruginosa flagellin modify their inflammatory response to a second, nonrelated stimulus, including live pathogens or lipopolysaccharide. Using histone acetyltransferase and methyltransferase inhibitors, we showed that this phenomenon relied on epigenetic regulation. This report is a major breakthrough in the field of multimicrobial respiratory tract infections, wherein control of inflammatory exacerbations is a major therapeutic issue.


Subject(s)
Immunologic Memory , Respiratory Mucosa/cytology , Epigenesis, Genetic , Epithelial Cells/immunology , Flagellin/immunology , Gene Expression Regulation/immunology , Humans , Inflammation , Lipopolysaccharides , Proof of Concept Study , Pseudomonas aeruginosa/immunology , RNA, Messenger , Respiratory Mucosa/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
7.
Front Immunol ; 9: 2823, 2018.
Article in English | MEDLINE | ID: mdl-30555487

ABSTRACT

In cystic fibrosis (CF), Pseudomonas aeruginosa (Pa) colonizes the lungs, leading to chronic inflammation of the bronchial epithelium. ChaC glutathione-specific γ-glutamylcyclotransferase 1 (CHAC1) mRNA is differentially expressed in primary human airway epithelial cells from bronchi (hAECBs) from patients with CF and healthy patients at baseline and upon infection with Pa. CHAC1 degrades glutathione and is associated with ER stress and apoptosis pathways. In this study, we examined the roles of CHAC1 in the inflammatory response and apoptosis in lung epithelial cells. First, we confirmed by reverse transcription quantitative polymerase chain reaction that CHAC1 mRNA was overexpressed in hAECBs from patients without CF compared with the expression in hAECBs from patients with CF upon Pa (PAK strain) infection. Moreover, the Pa virulence factors LPS and flagellin were shown to induce CHAC1 expression in cells from patients without CF. Using NCI-H292 lung epithelial cells, we found that LPS-induced CHAC1 mRNA expression was PERK-independent and involved ATF4. Additionally, using CHAC1 small interfering RNA, we showed that reduced CHAC1 expression in the context of LPS and flagellin stimulation was associated with modulation of inflammatory markers and alteration of NF-κB signaling. Finally, we showed that Pa was not able to induce apoptosis in NCI-H292 cells. Our results suggest that CHAC1 is involved in the regulation of inflammation in bronchial cells during Pa infection and may explain the excessive inflammation present in the respiratory tracts of patients with CF.


Subject(s)
Bronchi/immunology , Cystic Fibrosis/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , gamma-Glutamylcyclotransferase/immunology , A549 Cells , Adult , Aged , Bronchi/microbiology , Bronchi/pathology , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Humans , Male , Middle Aged , Pseudomonas Infections/genetics , Pseudomonas Infections/pathology
8.
Sci Rep ; 8(1): 15699, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356167

ABSTRACT

Aspergillus fumigatus is an environmental filamentous fungus that may act as an opportunistic pathogen causing a variety of diseases, including asthma or allergic bronchopulmonary aspergillosis, and infection, ranging from asymptomatic colonization to invasive pulmonary form, especially in immunocompromised patients. This fungus is characterized by different morphotypes including conidia which are the infective propagules able to germinate into hyphae. Due to their small size (2-3 µm), conidia released in the air can reach the lower respiratory tract. The objective of this study was to characterize the interactions between conidia and bronchial epithelial cells. To this end, we studied the role of bronchial epithelial cells, i.e., the BEAS-2B cell line and human primary cells, in conidial germination of a laboratory strain and three clinical strains of A. fumigatus. Microscopic observations and galactomannan measurements demonstrated that contact between epithelial cells and conidia leads to the inhibition of conidia germination. We demonstrated that this fungistatic process is not associated with the release of any soluble components nor internalization by the epithelial cells. We highlight that this antifungal process involves the phosphoinositide 3-kinase pathway on the host cellular side and the lectin FleA on the fungal side. Collectively, our results show that bronchial epithelial cells attenuate fungal virulence by inhibiting germination of extracellular conidia, thus preventing the morphological change from conidia to filaments, which is responsible for tissue invasion.


Subject(s)
Aspergillus fumigatus/pathogenicity , Bronchi/cytology , Epithelial Cells/metabolism , Epithelial Cells/parasitology , Lectins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Spores, Fungal/pathogenicity , Analysis of Variance , Animals , Cell Line , Cell Survival , Chi-Square Distribution , Fungal Proteins/metabolism , Galactose/analogs & derivatives , Humans , Lectins/chemical synthesis , Mannans/analysis , Microscopy , Spores, Fungal/cytology , Virulence
9.
Article in English | MEDLINE | ID: mdl-28611953

ABSTRACT

Pseudomonas aeruginosa (Pa) is the leading cause of chronic lung infection in Cystic Fibrosis (CF) patients. It is well recognized that CF epithelial cells fail to develop an appropriate response to infection, allowing bacterial colonization and a chronic inflammatory response. Since long non-coding RNAs (lncRNAs), are known to play a key role in regulating mammalian innate immune response, we hypothesized that CF cells exposed to Pa could express a specific lncRNA signature responsible of the maladaptative CF response. We analyzed transcriptomic datasets to compare the expression profiles of lncRNAs in primary CF and non-CF epithelial cells infected with Pa at 0, 2, 4, and 6 h of infection. Our analysis identified temporal expression signatures of 25, 73, 15, and 26 lncRNA transcripts differentially expressed at 0, 2, 4, and 6 h post-infection respectively, between CF and non-CF cells. In addition, we identified profiles specific to CF and non-CF cells. The differential expression of two candidate lncRNAs were independently validated using real-time PCR. We identified a specific CF signature of lncRNA expression in a context of Pa infection that could potentially play a role in the maladaptive immune response of CF patients.


Subject(s)
Bronchi/immunology , Cystic Fibrosis/immunology , Epithelial Cells/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/pathogenicity , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , Bronchi/microbiology , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Epithelial Cells/microbiology , Female , Gene Expression Regulation , Host-Parasite Interactions/immunology , Humans , Immunity, Innate , Lung/immunology , Lung/microbiology , Male , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Transcriptome
10.
mBio ; 7(3)2016 05 31.
Article in English | MEDLINE | ID: mdl-27247231

ABSTRACT

UNLABELLED: Overexpression of chromosomal resistance-nodulation-cell division (RND)-type efflux systems with broad substrate specificity contributes to multidrug resistance (MDR) in Acinetobacter baumannii We have shown that modulation of expression of the structural genes for the efflux systems AdeABC and AdeIJK confers MDR and results in numerous alterations of membrane-associated cellular functions, in particular biofilm formation. However, the contribution of these RND pumps to cell fitness and virulence has not yet been studied. The biological cost of an antibiotic resistance mechanism is a key parameter in determining its stability and dissemination. From an entirely sequenced susceptible clinical isolate, we have generated a set of isogenic derivatives having single point mutations resulting in overexpression of each efflux system or with every pump deleted by allelic replacement. We found that overproduction of the pumps results in a significant decrease in fitness of the bacterial host when measured by competition experiments in vitro Fitness and virulence were also evaluated in vivo both in systemic and pulmonary infection models in immunocompetent mice. A diminished competitiveness of the AdeABC-overexpressing mutant was observed only after intraperitoneal inoculation, but not after intranasal inoculation, the latter mimicking the most frequent type of human A. baumannii infection. However, in mice infected intranasally, this mutant was more virulent and stimulated an enhanced neutrophil activation in the lungs. Altogether, these data account for the observation that adeABC overexpression is common in MDR A. baumannii frequently found in ventilator-associated pneumonia. IMPORTANCE: Overproduction of the RND AdeABC efflux system is observed with a high incidence in multidrug-resistant Acinetobacter baumannii and results in increased resistance to several antibiotics of choice for the treatment of infections caused by this nosocomial pathogen. It was therefore important to study the biological cost of the overexpression of the adeABC structural operon which is normally tightly regulated. Fitness diminution of an overexpressing mutant detected in vitro and in vivo in a model that mimics sepsis was not observed in a pulmonary infection model in which the mutant was more virulent. This points out that increased virulence can occur independently from prolonged persistence in the infected organ and can account for the elevated incidence of this resistance mechanism in clinical isolates. The study also indicates that transposon libraries will identify only virulence genes that are expressed under physiological conditions but not those that are tightly regulated.


Subject(s)
Acinetobacter baumannii/genetics , Acinetobacter baumannii/pathogenicity , Drug Resistance, Multiple, Bacterial/genetics , Genetic Fitness , Membrane Transport Proteins/genetics , Acinetobacter Infections/microbiology , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Animals , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Division/genetics , Humans , Immunocompetence , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Neutrophils/immunology , Virulence
11.
PLoS One ; 10(10): e0140979, 2015.
Article in English | MEDLINE | ID: mdl-26485688

ABSTRACT

BACKGROUND AND AIMS: In cystic fibrosis (CF), Pseudomonas aeruginosa is not eradicated from the lower respiratory tract and is associated with epithelial inflammation that eventually causes tissue damage. To identify the molecular determinants of an effective response to P. aeruginosa infection, we performed a transcriptomic analysis of primary human bronchial epithelial cells from healthy donors (CTRL) 2, 4, and 6 h after induced P. aeruginosa infection. Compared to noninfected cells, infected cells showed changes in gene activity, which were most marked 6 h postinfection and usually consisted in upregulation. RESULTS: By comparing for each time point of infection, the transcriptomic response of epithelial cells from CF patients and healthy donors, we identified 851, 638, 667, and 980 differentially expressed genes 0, 2, 4, and 6 h postinfection, respectively. Gene selection followed by bioinformatic analysis showed that most of the differentially expressed genes, either up- or downregulated, were in the protein-binding and catalytic gene-ontology categories. Finally, we established that the protein products of the genes exhibiting the greatest differential upregulation (CSF2, CCL2, TNF, CSF3, MMP1, and MMP10) between CF patients and CTRL were produced in higher amounts by infected cells from CF patients versus CTRL. CONCLUSIONS: The differentially expressed genes in CF patients may constitute a signature for a detrimental inflammatory response and for an inefficient P. aeruginosa host-cell response.


Subject(s)
Bronchi/microbiology , Cystic Fibrosis/microbiology , Epithelial Cells/microbiology , Gene Expression Regulation , Pseudomonas Infections/genetics , Pseudomonas aeruginosa , Adult , Bronchi/pathology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Epithelial Cells/pathology , Female , Humans , Male , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Transcriptional Activation , Young Adult
12.
BMC Pulm Med ; 14: 100, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24909229

ABSTRACT

BACKGROUND: The aim was to measure flagellin concentrations in the expectorations of CF patients and to examine whether there are correlations with the level of respiratory insufficiency and inflammation. METHODS: Sputum samples from 31 adult patients chronically colonized with P. aeruginosa were collected and analysed for their content of flagellin and IL-8. Clinical data were extracted from patient files. RESULTS: Regardless of whether patients are colonized with mucoid strains or not, they carry clones of P. aeruginosa that express flagellin. While flagellin was present in airways of all of our CF patients, it is difficult to ascertain its contribution to inflammation (IL-8) and lung function deterioration. CONCLUSIONS: This is the first demonstration that flagellin is present in the sputum of patients. Thus, attempts to down regulate inflammation by the use of TLR5 (flagellin receptor) antagonists remain a possibility. However, this result needs to be extended to a larger number of patients to validate it for future research on this subject.


Subject(s)
Cystic Fibrosis/microbiology , Flagellin/analysis , Pseudomonas Infections/diagnosis , Pseudomonas aeruginosa/isolation & purification , Respiratory Insufficiency/diagnosis , Sputum/metabolism , Adult , Biomarkers/analysis , Cystic Fibrosis/diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-8/analysis , Male , Pseudomonas Infections/epidemiology , Sampling Studies , Severity of Illness Index , Sputum/microbiology
13.
Cell Microbiol ; 16(10): 1503-17, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24779433

ABSTRACT

Uropathogenic Escherichia coli (UPEC) colonizing kidneys is the main cause of acute pyelonephritis. TLR5 that senses flagellin was shown to be highly expressed in the bladder and to participate in host defence against flagellated UPEC, although its role in kidneys still remains elusive. Here we show that TLR5 is expressed in renal medullary collecting duct (MCD) cells, which represent a preferential site of UPEC adhesion. Flagellin, like lipopolysaccharide, stimulated the production of the chemoattractant chemokines CXCL1 and CXCL2, and subsequent migration capacity of neutrophils in cultured wild-type (WT) and Tlr4(-/-) MCDs, but not in Tlr5(-/-) MCDs. UPEC can translocate across intact MCD layers without altering tight junctions. Strikingly, the invasion capacity and transcellular translocation of the UPEC strain HT7 were significantly lower in Tlr5(-/-) than in WT MCDs. The non-motile HT7ΔfliC mutant lacking flagellin also exhibited much lower translocation capacities than the HT7 isolates. Finally, Tlr5(-/-) kidneys exhibited less infiltrating neutrophils than WT kidneys one day after the transurethral inoculation of HT7, and greater delayed renal bacterial loads in the day 4 post-infected Tlr5(-/-) kidneys. Overall, these findings indicate that the epithelial TLR5 participates to renal antibacterial defence, but paradoxically favours the translocation of UPEC across intact MCD cell layers.


Subject(s)
Escherichia coli Infections/immunology , Flagellin/immunology , Kidney Tubules, Collecting/immunology , Toll-Like Receptor 5/immunology , Uropathogenic Escherichia coli/pathogenicity , Animals , Bacterial Adhesion/physiology , Bacterial Load/immunology , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Escherichia coli Infections/microbiology , Female , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Pyelonephritis/immunology , Pyelonephritis/microbiology , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 5/genetics , Urinary Bladder/immunology , Urinary Bladder/metabolism , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology
14.
Am J Respir Crit Care Med ; 189(7): 799-811, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24555512

ABSTRACT

RATIONALE: Pseudomonas aeruginosa, a major problem pathogen responsible for severe infections in critically ill patients, triggers, through a functional type-3 secretion system (T3SS), the activation of an intracellular cytosolic sensor of innate immunity, NLRC4. Although the NLRC4-inflammasome-dependent response contributes to increased clearance of intracellular pathogens, it seems that NLRC4 inflammasome activation decreases the clearance of P. aeruginosa, a mainly extracellular pathogen. OBJECTIVES: We sought to determine the underlying mechanisms of this effect of the activation of NLRC4 by P. aeruginosa. METHODS: We established acute lung injury in wild-type and Nlrc4(-/-) mice using sublethal intranasal inocula of P. aeruginosa strain CHA expressing or not a functional T3SS. We studied 96-hour survival, lung injury, bacterial clearance from the lungs, cytokine secretion in bronchoalveolar lavage, lung antimicrobial peptide expression by quantitative polymerase chain reaction, and flow cytometry analysis of lung cells. MEASUREMENTS AND MAIN RESULTS: Nlrc4(-/-) mice showed enhanced bacterial clearance and decreased lung injury contributing to increased survival against extracellular P. aeruginosa strain expressing a functional T3SS. The mechanism involved decreased NLRC4-inflammasome-driven IL-18 secretion attenuating lung injury caused by excessive neutrophil recruitment. Additionally, in the lungs of Nlrc4(-/-) mice secretion of IL-17 by innate immune cells was increased and responsible for increased expression of lung epithelial antimicrobial peptides. Furthermore, IL-18 secretion was found to repress IL-17 and IL-17-driven lung antimicrobial peptide expression. CONCLUSIONS: We report a new role of the T3SS apparatus itself, independently of exotoxin translocation. Through NLRC4 inflammasome activation, the T3SS promotes IL-18 secretion, which dampens a beneficial IL-17-mediated antimicrobial host response.


Subject(s)
Acute Lung Injury/microbiology , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Inflammasomes/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Animals , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Caspase 1/metabolism , Cells, Cultured , Female , Flow Cytometry , Immunity, Innate , Interleukin-17/metabolism , Interleukin-18/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
J Infect Dis ; 210(2): 214-23, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24493823

ABSTRACT

Influenza A virus triggers a contagious respiratory disease that can cause considerable morbidity and mortality. Using an in vitro approach, we previously demonstrated that the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) plays a key role in influenza A virus-mediated immune response. However, the importance of RIG-I signaling in vivo has not been thoroughly examined, because of the lack of an appropriate mouse models. To circumvent this issue, we generated a new transgenic mouse overexpressing LGP2 (hereafter, "LGP2 TG mice"), a major regulator of the RIG-I signaling pathway. The time course of several parameters was compared in infected wild-type and LGP2 TG mice. We found that LGP2 TG mice displayed significantly reduced inflammatory mediators and a lower leukocyte infiltration into the bronchoalveolar airspace. More importantly, LGP2 TG mice had a significant survival advantage. Hence, our in vivo study reveals that LGP2 is a major downregulator of the influenza A virus-triggered detrimental inflammatory response.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/physiology , RNA Helicases/metabolism , Animals , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Gene Expression , Inflammation Mediators/analysis , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Signal Transduction , Survival Analysis
16.
PLoS One ; 8(12): e83077, 2013.
Article in English | MEDLINE | ID: mdl-24340081

ABSTRACT

Aspergillus fumigatus is an important allergen and opportunistic pathogen. Similarly to many other pathogens, it is able to produce lectins that may be involved in the host-pathogen interaction. We focused on the lectin AFL, which was prepared in recombinant form and characterized. Its binding properties were studied using hemagglutination and glycan array analysis. We determined the specificity of the lectin towards l-fucose and fucosylated oligosaccharides, including α1-6 linked core-fucose, which is an important marker for cancerogenesis. Other biologically relevant saccharides such as sialic acid, d-mannose or d-galactose were not bound. Blood group epitopes of the ABH and Lewis systems were recognized, Le(Y) being the preferred ligand among others. To provide a correlation between the observed functional characteristics and structural basis, AFL was crystallized in a complex with methyl-α,L-selenofucoside and its structure was solved using the SAD method. Six binding sites, each with different compositions, were identified per monomer and significant differences from the homologous AAL lectin were found. Structure-derived peptides were utilized to prepare anti-AFL polyclonal antibodies, which suggested the presence of AFL on the Aspergillus' conidia, confirming its expression in vivo. Stimulation of human bronchial cells by AFL led to IL-8 production in a dose-dependent manner. AFL thus probably contributes to the inflammatory response observed upon the exposure of a patient to A. fumigatus. The combination of affinity to human epithelial epitopes, production by conidia and pro-inflammatory activity is remarkable and shows that AFL might be an important virulence factor involved in an early stage of A. fumigatus infection.


Subject(s)
Aspergillus fumigatus/chemistry , Fucose/chemistry , Lectins/chemistry , Spores, Fungal/chemistry , Amino Acid Sequence , Aspergillosis/immunology , Binding Sites , Bronchi/cytology , Bronchi/microbiology , Epitopes/chemistry , Galactose/chemistry , Genome, Fungal , Hemagglutination , Host-Pathogen Interactions , Humans , Interleukin-8/metabolism , Mannose/chemistry , Molecular Sequence Data , N-Acetylneuraminic Acid/chemistry , Oligosaccharides/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Virulence Factors/chemistry
17.
PLoS Pathog ; 9(11): e1003716, 2013.
Article in English | MEDLINE | ID: mdl-24244155

ABSTRACT

α-(1,3)-Glucan is a major component of the cell wall of Aspergillus fumigatus, an opportunistic human fungal pathogen. There are three genes (AGS1, AGS2 and AGS3) controlling the biosynthesis of α-(1,3)-glucan in this fungal species. Deletion of all the three AGS genes resulted in a triple mutant that was devoid of α-(1,3)-glucan in its cell wall; however, its growth and germination was identical to that of the parental strain in vitro. In the experimental murine aspergillosis model, this mutant was less pathogenic than the parental strain. The AGS deletion resulted in an extensive structural modification of the conidial cell wall, especially conidial surface where the rodlet layer was covered by an amorphous glycoprotein matrix. This surface modification was responsible for viability reduction of conidia in vivo, which explains decrease in the virulence of triple agsΔ mutant.


Subject(s)
Aspergillosis/enzymology , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/pathogenicity , Cell Wall/enzymology , Fungal Polysaccharides/biosynthesis , Fungal Proteins/metabolism , Glucosyltransferases/metabolism , Animals , Aspergillosis/genetics , Aspergillosis/pathology , Aspergillus fumigatus/genetics , Cell Wall/genetics , Fungal Polysaccharides/genetics , Fungal Proteins/genetics , Gene Deletion , Glucosyltransferases/genetics , Humans , Mice, Knockout , Spores, Fungal/enzymology , Spores, Fungal/genetics
18.
PLoS One ; 7(7): e39888, 2012.
Article in English | MEDLINE | ID: mdl-22768318

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen involved in nosocomial infections. Flagellin is a P. aeruginosa virulence factor involved in host response to this pathogen. We examined the role of flagellin in P. aeruginosa-induced mucus secretion. Using a mouse model of pulmonary infection we showed that PAK, a wild type strain of P. aeruginosa, induced airway mucus secretion and mucin muc5ac expression at higher levels than its flagellin-deficient mutant (ΔFliC). PAK induced expression of MUC5AC and MUC2 in both human airway epithelial NCI-H292 cell line and in primary epithelial cells. In contrast, ΔFliC infection had lower to no effect on MUC5AC and MUC2 expressions. A purified P. aeruginosa flagellin induced MUC5AC expression in parallel to IL-8 secretion in NCI-H292 cells. Accordingly, ΔFliC mutant stimulated IL-8 secretion at significantly lower levels compared to PAK. Incubation of NCI-H292 cells with exogenous IL-8 induced MUC5AC expression and pre-incubation of these cells with an anti-IL-8 antibody abrogated flagellin-mediated MUC5AC expression. Silencing of TLR5 and Naip, siRNA inhibited both flagellin-induced MUC5AC expression and IL-8 secretion. Finally, inhibition of ERK abolished the expression of both PAK- and flagellin-induced MUC5AC. We conclude that: (i) flagellin is crucial in P. aeruginosa-induced mucus hyper-secretion through TLR5 and Naip pathways; (ii) this process is mediated by ERK and amplified by IL-8. Our findings help understand the mechanisms involved in mucus secretion during pulmonary infectious disease induced by P. aeruginosa, such as in cystic fibrosis.


Subject(s)
Flagellin/metabolism , Mucus/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , Animals , Cell Line , Female , Flagellin/immunology , Gene Expression Regulation/immunology , Humans , Interleukin-8/biosynthesis , Interleukin-8/immunology , Mice , Mucin 5AC/biosynthesis , Mucin 5AC/immunology , Mucin-2/biosynthesis , Mucin-2/immunology , Mucus/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Toll-Like Receptor 5/immunology , Toll-Like Receptor 5/metabolism
19.
J Infect Dis ; 206(6): 932-42, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22782952

ABSTRACT

BACKGROUND: Sepsis is characterized by a dysregulated inflammatory response followed by immunosuppression that favors the development of secondary infections. Toll-like receptors (TLRs) are major regulators of the host's response to infections. How variability in TLR signaling may impact the development of sepsis-induced immune dysfunction has not been established. We sought to establish the role of TLR2, TLR4, and TLR5 in postseptic mice with Pseudomonas aeruginosa pneumonia. METHODS: We used an experimental model of sublethal polymicrobial sepsis induced by cecal ligation and puncture (CLP). Wild-type, tlr2(-/-), tlr4(-/-), tlr5(-/-), tlr2 4(-/-) mice that underwent CLP were secondarily subjected to P. aeruginosa pulmonary infection. RESULTS: Postseptic wild-type and tlr4(-/-) and tlr5(-/-) mice displayed high susceptibility to P. aeruginosa pneumonia. In contrast, TLR2-deficient mice, either tlr2(-/-)or tlr2 4(-/-), that underwent CLP were resistant to the secondary pulmonary infection. As compared to wild-type mice, tlr2(-/-) mice displayed improvement in bacterial clearance, decreased bacteremic dissemination, and attenuated lung damage. Furthermore, tlr2(-/-) mice exhibited a pulmonary proinflammatory cytokine balance, with increased production of tumor necrosis factor α and decreased release of interleukin 10. CONCLUSIONS: In a model of secondary P. aeruginosa pneumonia in postseptic mice, TLR2 deficiency improves survival by promoting efficient bacterial clearance and restoring a proinflammatory cytokine balance in the lung.


Subject(s)
Pneumonia, Bacterial/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Sepsis/complications , Toll-Like Receptor 2/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Female , Inflammation/metabolism , Lung/cytology , Mice , Mice, Knockout , Neutrophils/metabolism , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , RNA/genetics , RNA/metabolism , Real-Time Polymerase Chain Reaction , Sepsis/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism
20.
Proc Natl Acad Sci U S A ; 109(5): 1619-24, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307620

ABSTRACT

A deficit in early clearance of Pseudomonas aeruginosa (P. aeruginosa) is crucial in nosocomial pneumonia and in chronic lung infections. Few studies have addressed the role of Toll-like receptors (TLRs), which are early pathogen associated molecular pattern receptors, in pathogen uptake and clearance by alveolar macrophages (AMs). Here, we report that TLR5 engagement is crucial for bacterial clearance by AMs in vitro and in vivo because unflagellated P. aeruginosa or different mutants defective in TLR5 activation were resistant to AM phagocytosis and killing. In addition, the clearance of PAK (a wild-type P. aeruginosa strain) by primary AMs was causally associated with increased IL-1ß release, which was dramatically reduced with PAK mutants or in WT PAK-infected primary TLR5(-/-) AMs, demonstrating the dependence of IL-1ß production on TLR5. We showed that this IL-1ß production was important in endosomal pH acidification and in inducing the killing of bacteria by AMs through asparagine endopeptidase (AEP), a key endosomal cysteine protease. In agreement, AMs from IL-1R1(-/-) and AEP(-/-) mice were unable to kill P. aeruginosa. Altogether, these findings demonstrate that TLR5 engagement plays a major role in P. aeruginosa internalization and in triggering IL-1ß formation.


Subject(s)
Endopeptidases/metabolism , Interleukin-1beta/metabolism , Macrophages, Alveolar/immunology , Phagocytosis , Pseudomonas aeruginosa/immunology , Toll-Like Receptor 5/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...