Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Gut Microbes ; 15(2): 2249960, 2023 12.
Article En | MEDLINE | ID: mdl-37655966

Over 90% of epidemic non-bacterial gastroenteritis are caused by human noroviruses (NoVs), which persist in a substantial subset of people allowing their spread worldwide. This has led to a significant number of endemic cases and up to 70,000 children deaths in developing countries. NoVs are primarily transmitted through the fecal-oral route. To date, studies have focused on the influence of the gut microbiota on enteric viral clearance by mucosal immunity. In this study, the use of mouse norovirus S99 (MNoV_S99) and CR6 (MNoV_CR6), two persistent strains, allowed us to provide evidence that the norovirus-induced exacerbation of colitis severity relied on bacterial sensing by nucleotide-binding oligomerization domain 2 (Nod2). Consequently, Nod2-deficient mice showed reduced levels of gravity of Dextran sodium sulfate (DSS)-induced colitis with both viral strains. And MNoV_CR6 viremia was heightened in Nod2-/- mice in comparison with animals hypomorphic for Atg16l1, which are prone to aggravated inflammation under DSS. Accordingly, the infection of macrophages derived from WT mice promoted the phosphorylation of Signal Transducer and Activator of Transcription 1 (STAT1) and NOD2's expression levels. Higher secretion of Tumor Necrosis Factor alpha (TNFα) following NOD2 activation and better viral clearance were measured in these cells. By contrast, reduced levels of pSTAT1 and blunted downstream secretion of TNFα were found in Nod2-deficient macrophages infected by MNoV_S99. Hence, our results uncover a previously unidentified virus-host-bacterial interplay that may represent a novel therapeutic target for treating noroviral origin gastroenteritis that may be linked with susceptibility to several common illnesses such as Crohn's disease.


Caliciviridae Infections , Colitis , Gastroenteritis , Gastrointestinal Microbiome , Nod2 Signaling Adaptor Protein , Animals , Mice , Caliciviridae Infections/immunology , Colitis/chemically induced , Colitis/virology , Gastroenteritis/immunology , Gastroenteritis/virology , Nod2 Signaling Adaptor Protein/metabolism
2.
Biofilm ; 5: 100131, 2023 Dec.
Article En | MEDLINE | ID: mdl-37252226

Biofilms are highly tolerant to antimicrobials and host immune defense, enabling pathogens to thrive in hostile environments. The diversity of microbial biofilm infections requires alternative and complex treatment strategies. In a previous work we demonstrated that the human Atrial Natriuretic Peptide (hANP) displays a strong anti-biofilm activity toward Pseudomonas aeruginosa and that the binding of hANP by the AmiC protein supports this effect. This AmiC sensor has been identified as an analog of the human natriuretic peptide receptor subtype C (h-NPRC). In the present study, we evaluated the anti-biofilm activity of the h-NPRC agonist, osteocrin (OSTN), a hormone that displays a strong affinity for the AmiC sensor at least in vitro. Using molecular docking, we identified a pocket in the AmiC sensor that OSTN reproducibly docks into, suggesting that OSTN might possess an anti-biofilm activity as well as hANP. This hypothesis was validated since we observed that OSTN dispersed established biofilm of P. aeruginosa PA14 strain at the same concentrations as hANP. However, the OSTN dispersal effect is less marked than that observed for the hANP (-61% versus -73%). We demonstrated that the co-exposure of P. aeruginosa preformed biofilm to hANP and OSTN induced a biofilm dispersion with a similar effect to that observed with hANP alone suggesting a similar mechanism of action of these two peptides. This was confirmed by the observation that OSTN anti-biofilm activity requires the activation of the complex composed by the sensor AmiC and the regulator AmiR of the ami pathway. Using a panel of both P. aeruginosa laboratory reference strains and clinical isolates, we observed that the OSTN capacity to disperse established biofilms is highly variable from one strain to another. Taken together, these results show that similarly to the hANP hormone, OSTN has a strong potential to be used as a tool to disperse P. aeruginosa biofilms.

3.
Nat Commun ; 14(1): 78, 2023 01 05.
Article En | MEDLINE | ID: mdl-36604442

Carbapenemase-producing Enterobacterales (CPE) are spreading rapidly in hospital settings. Asymptomatic CPE gut colonisation may be associated with dysbiosis and gut-lung axis alterations, which could impact lung infection outcomes. In this study, in male C57BL/6JRj mice colonised by CPE, we characterise the resulting gut dysbiosis, and analyse the lung immune responses and outcomes of subsequent Pseudomonas aeruginosa lung infection. Asymptomatic gut colonisation by CPE leads to a specific gut dysbiosis and increases the severity of P. aeruginosa lung infection through lower numbers of alveolar macrophages and conventional dendritic cells. CPE-associated dysbiosis is characterised by a near disappearance of the Muribaculaceae family and lower levels of short-chain fatty acids. Faecal microbiota transplantation restores immune responses and outcomes of lung infection outcomes, demonstrating the involvement of CPE colonisation-induced gut dysbiosis in altering the immune gut-lung axis, possibly mediated by microbial metabolites such as short-chain fatty acids.


Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Mice , Male , Klebsiella pneumoniae , Dysbiosis , Mice, Inbred C57BL , Lung , Fatty Acids, Volatile
4.
J Crohns Colitis ; 17(1): 111-122, 2023 Jan 27.
Article En | MEDLINE | ID: mdl-35917251

BACKGROUND AND AIMS: NOD2 has emerged as a critical player in the induction of both Th1 and Th2 responses for potentiation and polarisation of antigen-dependent immunity. Loss-of-function mutations in the NOD2-encoding gene and deregulation of its downstream signalling pathway have been linked to Crohn's disease. Although it is well documented that NOD2 is capable of sensing bacterial muramyl dipeptide, it remains counter-intuitive to link development of overt intestinal inflammation to a loss of bacterial-induced inflammatory response. We hypothesised that a T helper bias could also contribute to an autoimmune-like colitis different from inflammation that is fully fledged by Th1 type cells. METHODS: An oedematous bowel wall with a mixed Th1/Th2 response was induced in mice by intrarectal instillation of the haptenating agent oxazolone. Survival and clinical scoring were evaluated. At several time points after instillation, colonic damage was assessed by macroscopic and microscopic observations. To evaluate the involvement of NOD2 in immunochemical phenomena, quantitative polymerase chain reaction [PCR] and flow cytometry analysis were performed. Bone marrow chimera experimentation allowed us to evaluate the role of haematopoietic/non-hematopoietic NOD2-expressing cells. RESULTS: Herein, we identified a key regulatory circuit whereby NOD2-mediated sensing of a muramyl dipeptide [MDP] by radio-resistant cells improves colitis with a mixed Th1/Th2 response that is induced by oxazolone. Genetic ablation of either Nod2 or Ripk2 precipitated oxazolone colitis that is predominantly linked to a lack of interferon-gamma. Bone marrow chimera experiments revealed that inactivation of Nod2 signalling in non-haematopoietic cells is causing a biased M1-M2 polarisation of macrophages and a decreased frequency of splenic regulatory T cells that correlates with an impaired activation of CD4 + T cells within mesenteric lymph nodes. Mechanistically, mice were protected from oxazolone-induced colitis upon administration of MDP in an interleukin-1- and interleukin-23-dependent manner. CONCLUSIONS: These findings indicate that Nod2 signalling may prevent pathological conversion of T helper cells for maintenance of tissue homeostasis.


Colitis , Oxazolone , Mice , Animals , Oxazolone/adverse effects , Acetylmuramyl-Alanyl-Isoglutamine/adverse effects , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Colitis/metabolism , Inflammation , Signal Transduction , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism
5.
Front Cell Infect Microbiol ; 12: 859181, 2022.
Article En | MEDLINE | ID: mdl-35782124

Bacteria of the genus Achromobacter are environmental germs, with an unknown reservoir. It can become opportunistic pathogens in immunocompromised patients, causing bacteremia, meningitis, pneumonia, or peritonitis. In recent years, Achromobacter xylosoxidans has emerged with increasing incidence in patients with cystic fibrosis (CF). Recent studies showed that A. xylosoxidans is involved in the degradation of the respiratory function of patients with CF. The respiratory ecosystem of patients with CF is colonized by bacterial species that constantly fight for space and access to nutrients. The type VI secretion system (T6SS) empowers this constant bacterial antagonism, and it is used as a virulence factor in several pathogenic bacteria. This study aimed to investigate the prevalence of the T6SS genes in A. xylosoxidans isolated in patients with CF. We also evaluated clinical and molecular characteristics of T6SS-positive A. xylosoxidans strains. We showed that A. xylosoxidans possesses a T6SS gene cluster and that some environmental and clinical isolates assemble a functional T6SS nanomachine. A. xylosoxidans T6SS is used to target competing bacteria, including other CF-specific pathogens. Finally, we demonstrated the importance of the T6SS in the internalization of A. xylosoxidans in lung epithelial cells and that the T6SS protein Hcp is detected in the sputum of patients with CF. Altogether, these results suggest for the first time a role of T6SS in CF-lung colonization by A. xylosoxidans and opens promising perspective to target this virulence determinant as innovative theranostic options for CF management.


Achromobacter denitrificans , Cystic Fibrosis , Gram-Negative Bacterial Infections , Type VI Secretion Systems , Achromobacter denitrificans/genetics , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Ecosystem , Gram-Negative Bacterial Infections/microbiology , Humans , Lung , Type VI Secretion Systems/genetics , Virulence Factors/genetics
6.
Adv Sci (Weinh) ; 9(7): e2103262, 2022 03.
Article En | MEDLINE | ID: mdl-35032112

Pseudomonas aeruginosa biofilms cause chronic, antibiotic tolerant infections in wounds and lungs. Numerous recent studies demonstrate that bacteria can detect human communication compounds through specific sensor/receptor tools that modulate bacterial physiology. Consequently, interfering with these mechanisms offers an exciting opportunity to directly affect the infection process. It is shown that the human hormone Atrial Natriuretic Peptide (hANP) both prevents the formation of P. aeruginosa biofilms and strongly disperses established P. aeruginosa biofilms. This hANP action is dose-dependent with a strong effect at low nanomolar concentrations and takes effect in 30-120 min. Furthermore, although hANP has no antimicrobial effect, it acts as an antibiotic adjuvant. hANP enhances the antibiofilm action of antibiotics with diverse modes of action, allowing almost full biofilm eradication. The hANP effect requires the presence of the P. aeruginosa sensor AmiC and the AmiR antiterminator regulator, indicating a specific mode of action. These data establish the activation of the ami pathway as a potential mechanism for P. aeruginosa biofilm dispersion. hANP appears to be devoid of toxicity, does not enhance bacterial pathogenicity, and acts synergistically with antibiotics. These data show that hANP is a promising powerful antibiofilm weapon against established P. aeruginosa biofilms in chronic infections.


Atrial Natriuretic Factor , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Atrial Natriuretic Factor/metabolism , Atrial Natriuretic Factor/pharmacology , Biofilms , Humans , Pseudomonas aeruginosa/metabolism , Virulence
7.
J Mol Diagn ; 23(2): 234-241, 2021 02.
Article En | MEDLINE | ID: mdl-33259953

Bacterial vaginosis (BV) diagnosis in pregnancy is based on the Nugent score, which consists of semiquantitation of bacterial morphotypes. Limited data exist concerning molecular-based diagnosis in asymptomatic pregnant women. Using high-throughput quantitative PCR, 34 microorganisms were screened in asymptomatic pregnant women and compared with the Nugent score. Three-hundred and four vaginal samples had a Nugent score <7 (69.9%) and 131, a Nugent score ≥7 (30.1%), consistent with BV. More pregnant women with BV share Atopobiumvaginae, bacterial vaginosis associated bacteria-2, Gardnerella spp., Mobiluncus curtisii, Mo. mulieris, Mycoplasma hominis, Ureaplasma urealyticum, Prevotella bivia, Megasphaera 1, and Megasphaera 2 in their vaginal sample. Fewer pregnant women with BV share Lactobacillus crispatus, L. gasseri, L. jensenii, and Enterococcus faecalis in their vaginal sample (P < 0.001). Classification and regression tree analysis was performed to determine which combinations of detected bacteria optimally diagnose BV in this population. A set of only four bacteria of 34 microorganisms (A. vaginae, Gardnerella spp., L. crispatus, and P. bivia) was the best combination to identify BV in a cohort of asymptomatic pregnant women, with a sensitivity of 77.1%, and specificity of 97.0% compared with the Nugent score. The quantitative PCR in the present study responds to the limits of the Nugent score by implementing an easily reproducible quantitative assay to assess the absence of BV in pregnancy.


Pregnant Women , Real-Time Polymerase Chain Reaction/methods , Vaginosis, Bacterial/diagnosis , Adult , Female , Humans , Regression Analysis , Vaginosis, Bacterial/microbiology
8.
Crit Care ; 24(1): 611, 2020 10 15.
Article En | MEDLINE | ID: mdl-33076936

BACKGROUND: Gut dysbiosis due to the adverse effects of antibiotics affects outcomes of lung infection. Previous murine models relied on significant depletion of both gut and lung microbiota, rendering the analysis of immune gut-lung cross-talk difficult. Here, we study the effects of antibiotic-induced gut dysbiosis without lung dysbiosis on lung immunity and the consequences on acute P. aeruginosa lung infection. METHODS: C57BL6 mice received 7 days oral vancomycin-colistin, followed by normal regimen or fecal microbial transplant or Fms-related tyrosine kinase 3 ligand (Flt3-Ligand) over 2 days, and then intra-nasal P. aeruginosa strain PAO1. Gut and lung microbiota were studied by next-generation sequencing, and lung infection outcomes were studied at 24 h. Effects of vancomycin-colistin on underlying immunity and bone marrow progenitors were studied in uninfected mice by flow cytometry in the lung, spleen, and bone marrow. RESULTS: Vancomycin-colistin administration induces widespread cellular immunosuppression in both the lung and spleen, decreases circulating hematopoietic cytokine Flt3-Ligand, and depresses dendritic cell bone marrow progenitors leading to worsening of P. aeruginosa lung infection outcomes (bacterial loads, lung injury, and survival). Reversal of these effects by fecal microbial transplant shows that these alterations are related to gut dysbiosis. Recombinant Flt3-Ligand reverses the effects of antibiotics on subsequent lung infection. CONCLUSIONS: These results show that gut dysbiosis strongly impairs monocyte/dendritic progenitors and lung immunity, worsening outcomes of P. aeruginosa lung infection. Treatment with a fecal microbial transplant or immune stimulation by Flt3-Ligand both restore lung cellular responses to and outcomes of P. aeruginosa following antibiotic-induced gut dysbiosis.


Anti-Bacterial Agents/adverse effects , Dysbiosis/complications , Immunosuppression Therapy/adverse effects , Pneumonia/etiology , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Dysbiosis/etiology , Dysbiosis/physiopathology , Immunosuppression Therapy/methods , Lung/microbiology , Lung/physiopathology , Mice, Inbred C57BL , Microbiota/drug effects , Pneumonia/physiopathology , Pseudomonas aeruginosa/drug effects , Vancomycin/adverse effects , Vancomycin/pharmacology
9.
Article En | MEDLINE | ID: mdl-30962336

While antibiotic use is a risk factor of carbapenemase-producing Enterobacteriaceae (CPE) acquisition, the importance of timing of antibiotic administration relative to CPE exposure remains unclear. In a murine model of gut colonization by New Delhi metallo-beta-lactamase-1 (NDM-1)-producing Klebsiella pneumoniae, a single injection of clindamycin within at most 1 week before or after CPE exposure induced colonization persisting up to 100 days. The timing of antibiotic administration relative to CPE exposure may be relevant to infection control and antimicrobial stewardship approaches.


Anti-Bacterial Agents/administration & dosage , Bacterial Proteins/metabolism , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae/drug effects , Gastrointestinal Microbiome/drug effects , beta-Lactamases/metabolism , Animals , Disease Models, Animal , Enterobacteriaceae/metabolism , Infection Control/methods , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Mice , Microbial Sensitivity Tests/methods
10.
Article En | MEDLINE | ID: mdl-30533763

Pseudomonas aeruginosa infections are challenging due to intrinsic and acquired resistance mechanisms. We report here the draft genome sequences of two multidrug-resistant strains-PAL0.1, isolated from the airways of an intensive care unit (ICU) patient with ventilator-associated pneumonia, and PAL1.1, isolated from blood cultures of an ICU patient with sepsis.

11.
Article En | MEDLINE | ID: mdl-30533912

Carbapenemase-producing Klebsiella pneumoniae represents an emerging public health issue. Here, we present the draft whole-genome sequences of K. pneumoniae clinical strains KPL0.1 (OXA-48 carbapenemase) and KPL0.2 (NDM-1 carbapenemase). These genome sequences should help in investigating pathophysiological mechanisms of digestive colonization or infection with these highly resistant bacteria.

12.
Org Biomol Chem ; 15(47): 10037-10043, 2017 Dec 06.
Article En | MEDLINE | ID: mdl-29165489

The synthesis of eight perylenediimide-based glycoclusters was readily performed from hexa- and tetra-propargylated cores through azide-alkyne "click" conjugation. Variations in the carbohydrate epitope (Glc, Gal, Man, Fuc) and the linker arm provided molecular diversity. Interactions with LecA and LecB, two proteins involved in the adhesion of Pseudomonas aeruginosa to host tissues, were evaluated by microcalorimetry (ITC). In both cases high affinities were obtained with Kd values in the nanomolar range. Further evaluation of their anti-adhesive properties using cultured epithelial cells demonstrated their potent anti-adhesive activities against Pseudomonas aeruginosa with only 30-40% residual adhesion observed. The fluorescence properties of the PDI core were then investigated by confocal microscopy on cell-bacteria cultures. However, the red fluorescence signal of the PDI-based glycocluster was too weak to provide significant data. The present study provides another type of anti-adhesive glycocluster against bacterial infection with a large aromatic PDI core.


Adhesins, Bacterial/drug effects , Glycoconjugates/pharmacology , Imides/pharmacology , Lectins/antagonists & inhibitors , Perylene/analogs & derivatives , Pseudomonas aeruginosa/drug effects , Binding Sites/drug effects , Calorimetry , Cell Adhesion/drug effects , Glycoconjugates/chemical synthesis , Glycoconjugates/chemistry , Imides/chemical synthesis , Imides/chemistry , Ligands , Molecular Structure , Perylene/chemical synthesis , Perylene/chemistry , Perylene/pharmacology , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/cytology
13.
Int Immunol ; 29(8): 377-384, 2017 08 01.
Article En | MEDLINE | ID: mdl-28992059

While NLRC4-dependent sensing of intracellular Gram-negative pathogens such as Salmonella enterica serovar typhimurium is a beneficial host response, NLRC4-dependent sensing of the Pseudomonas aeruginosa type 3 secretion system (T3SS) has been shown to be involved in pathogenicity. In mice, different pathogen-associated microbial patterns are sensed by the combination of the NLRC4-inflammasome with different neuronal apoptosis inhibitory proteins (NAIPs). NAIP2 is involved in sensing PscI, an inner-rod protein of the P. aeruginosa T3SS. Surprisingly, only a single human NAIP (hNAIP) has been found. Moreover, there is no description of hNAIP-NLRC4 inflammasome recognition of T3SS inner-rod proteins in humans. Here, we show that the P. aeruginosa T3SS inner-rod protein PscI and needle protein PscF are both sensed by the hNAIP-NLRC4 inflammasome in human macrophages and PBMCs from healthy donors, allowing caspase-1 and IL-1ß maturation and resulting in a robust inflammatory response. TLR4 and TLR2 are involved in redundantly sensing these two T3SS components.


CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Inflammasomes/metabolism , Macrophages/immunology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Type III Secretion Systems/metabolism , Animals , Carrier Proteins/metabolism , Caspase 1/metabolism , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins , Interleukin-1beta/metabolism , Macrophages/microbiology , Mice , Pathogen-Associated Molecular Pattern Molecules/immunology , THP-1 Cells , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Type III Secretion Systems/immunology
14.
EMBO J ; 36(13): 1869-1887, 2017 07 03.
Article En | MEDLINE | ID: mdl-28483816

Bacterial pathogens often subvert the innate immune system to establish a successful infection. The direct inhibition of downstream components of innate immune pathways is particularly well documented but how bacteria interfere with receptor proximal events is far less well understood. Here, we describe a Toll/interleukin 1 receptor (TIR) domain-containing protein (PumA) of the multi-drug resistant Pseudomonas aeruginosa PA7 strain. We found that PumA is essential for virulence and inhibits NF-κB, a property transferable to non-PumA strain PA14, suggesting no additional factors are needed for PumA function. The TIR domain is able to interact with the Toll-like receptor (TLR) adaptors TIRAP and MyD88, as well as the ubiquitin-associated protein 1 (UBAP1), a component of the endosomal-sorting complex required for transport I (ESCRT-I). These interactions are not spatially exclusive as we show UBAP1 can associate with MyD88, enhancing its plasma membrane localization. Combined targeting of UBAP1 and TLR adaptors by PumA impedes both cytokine and TLR receptor signalling, highlighting a novel strategy for innate immune evasion.


Carrier Proteins/antagonists & inhibitors , Immune Evasion , Membrane Glycoproteins/antagonists & inhibitors , Myeloid Differentiation Factor 88/antagonists & inhibitors , Pseudomonas aeruginosa/pathogenicity , Receptors, Interleukin-1/antagonists & inhibitors , Toll-Like Receptors/antagonists & inhibitors , Virulence Factors/metabolism , Bacterial Proteins/metabolism , Cell Line , Epithelial Cells/immunology , Epithelial Cells/microbiology , Humans , Pseudomonas aeruginosa/immunology
15.
Sci Rep ; 7: 41178, 2017 01 24.
Article En | MEDLINE | ID: mdl-28117457

We have previously shown that the eukaryotic C-type natriuretic peptide hormone (CNP) regulates Pseudomonas aeruginosa virulence and biofilm formation after binding on the AmiC sensor, triggering the amiE transcription. Herein, the involvement of the aliphatic amidase AmiE in P. aeruginosa virulence regulation has been investigated. The proteome analysis of an AmiE over-producing strain (AmiE+) revealed an expression change for 138 proteins, including some that are involved in motility, synthesis of quorum sensing compounds and virulence regulation. We observed that the AmiE+ strain produced less biofilm compared to the wild type, and over-produced rhamnolipids. In the same line, AmiE is involved in P. aeruginosa motilities (swarming and twitching) and production of the quorum sensing molecules N-acyl homoserine lactones and Pseudomonas Quinolone Signal (PQS). We observed that AmiE overproduction reduced levels of HCN and pyocyanin causing a decreased virulence in different hosts (i.e. Dictyostelium discoideum and Caenorhabditis elegans). This phenotype was further confirmed in a mouse model of acute lung infection, in which AmiE overproduction resulted in an almost fully virulence decrease. Taken together, our data suggest that, in addition to its role in bacterial secondary metabolism, AmiE is involved in P. aeruginosa virulence regulation by modulating pilus synthesis and cell-to-cell communication.


Amidohydrolases/metabolism , Pseudomonas Infections/enzymology , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity , Virulence Factors , Animals , Biofilms , Caenorhabditis elegans/microbiology , Dictyostelium/microbiology , Female , Lung/microbiology , Male , Mice, Inbred C57BL , Proteome , Pseudomonas Infections/microbiology , Quorum Sensing , Virulence
16.
BMC Microbiol ; 16(1): 137, 2016 07 08.
Article En | MEDLINE | ID: mdl-27392067

BACKGROUND: Pseudomonas aeruginosa (Pa) is a Gram-negative bacteria frequently involved in healthcare-associated pneumonia with poor clinical outcome. To face the announced post-antibiotic era due to increasing resistance and lack of new antibiotics, new treatment strategies have to be developed. Immunomodulation of the host response involved in outcome could be an alternative therapeutic target in Pa-induced lung infection. Kynurenines are metabolites resulting from tryptophan catabolism and are known for their immunomodulatory properties. Pa catabolizes tryptophan through the kynurenine pathway. Interestingly, many host cells also possess the kynurenine pathway, whose metabolites are known to control immune system homeostasis. Thus, bacterial metabolites may interfere with the host's immune response. However, the kynurenine pathway in Pa, including functional enzymes, types and amounts of secreted metabolites remains poorly known. Using liquid chromatography coupled to mass spectrometry and different strains of Pa, we determined types and levels of metabolites produced by Pa ex vivo in growth medium, and the relevance of this production in vivo in a murine model of acute lung injury. RESULTS: Ex vivo, Pa secretes clinically relevant kynurenine levels (µM to mM). Pa also secretes kynurenic acid and 3-OH-kynurenine, suggesting that the bacteria possess both a functional kynurenine aminotransferase and kynurenine monooxygenase. The bacterial kynurenine pathway is the major pathway leading to anthranilate production both ex vivo and in vivo. In the absence of the anthranilate pathway, the kynurenine pathway leads to kynurenic acid production. CONCLUSION: Pa produces and secretes several metabolites of the kynurenine pathway. Here, we demonstrate the existence of new metabolic pathways leading to synthesis of bioactive molecules, kynurenic acid and 3-OH-kynurenine in Pa. The kynurenine pathway in Pa is critical to produce anthranilate, a crucial precursor of some Pa virulence factors. Metabolites (anthranilate, kynurenine, kynurenic acid) are produced at sustained levels both ex vivo and in vivo leading to a possible immunomodulatory interplay between bacteria and host. These data may imply that pulmonary infection with bacteria highly expressing the kynurenine pathway enzymes could influence the equilibrium of the host's tryptophan metabolic pathway, known to be involved in the immune response to infection. Further studies are needed to explore the effects of these metabolic changes on the pathophysiology of Pa infection.


Pseudomonas aeruginosa/metabolism , Tryptophan/metabolism , Acute Lung Injury/microbiology , Animals , Disease Models, Animal , Host-Pathogen Interactions , Immunity, Innate , Kynurenic Acid/metabolism , Kynurenine/metabolism , Metabolic Networks and Pathways , Mice , Murinae , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/enzymology , Transaminases/metabolism , ortho-Aminobenzoates/metabolism
17.
Medicine (Baltimore) ; 95(5): e2668, 2016 Feb.
Article En | MEDLINE | ID: mdl-26844497

Bacterial vaginosis (BV) is considered as a trigger for an inflammatory response that could promote adverse pregnancy outcome (APO). We hypothesized that BV-related inflammation could be counterbalanced by anti-inflammatory and mucosal homeostatic responses that could participate in pregnancy outcomes.A total of 402 vaginal self-samples from pregnant women in their first trimester were screened by Nugent score. In this population, we enrolled 23 pregnant women with BV but without APO, 5 pregnant women with BV and developing APO, 21 pregnant women with intermediate flora, and 28 random control samples from pregnant women without BV or APO.BV without APO in pregnant women was associated with 28-fold interleukin-8, 5-fold interleukin-10, and 40-fold interleukin-22 increases in expression compared to controls. BV associated with APO in pregnant women shared 4-fold increase in tumor necrosis factor, 100-fold decrease in interleukin-10, and no variation in interleukin-22 expressions compared to controls. Next-generation sequencing of vaginal microbiota revealed a shift from obligate anaerobic bacteria dominance in BV without APO pregnant women to Lactobacillus dominance microbiota in BV with APO.Our results show that the anti-inflammatory and mucosal homeostatic responses to BV may determine outcome of pregnancy in the setting of BV possibly through effects on the vaginal microbiota.


Pregnancy Complications, Infectious/immunology , Vaginosis, Bacterial/immunology , Adult , Female , Homeostasis , Humans , Immunity, Mucosal , Pilot Projects , Pregnancy , Pregnancy Outcome
18.
Vet Res ; 46: 91, 2015 Sep 04.
Article En | MEDLINE | ID: mdl-26338794

The murine norovirus (MNV) is a recently discovered mouse pathogen, representing the most common contaminant in laboratory mouse colonies. Nevertheless, the effects of MNV infection on biomedical research are still unclear. We tested the hypothesis that MNV infection could alter immune response in mice with acute lung infection. Here we report that co-infection with MNV increases survival of mice with Pseudomonas aeruginosa acute lung injury and decreases in vivo production of pro-inflammatory cytokines. Our results suggest that MNV infection can deeply modify the parameters studied in conventional models of infection and lead to false conclusions in experimental models.


Caliciviridae Infections/immunology , Cytokines/metabolism , Immunomodulation , Norovirus/physiology , Pneumonia/immunology , Pseudomonas Infections/immunology , Rodent Diseases/immunology , Acute Disease , Animals , Caliciviridae Infections/virology , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Male , Mice , Mice, Inbred C57BL , Pneumonia/microbiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Rodent Diseases/microbiology , Rodent Diseases/virology
19.
Mol Microbiol ; 96(2): 419-36, 2015 Apr.
Article En | MEDLINE | ID: mdl-25614137

The export of bacterial toxins across the bacterial envelope requires the assembly of complex, membrane-embedded protein architectures. Pseudomonas aeruginosa employs type III secretion (T3S) injectisome to translocate exotoxins directly into the cytoplasm of a target eukaryotic cell. This multi-protein channel crosses two bacterial membranes and extends further as a needle through which the proteins travel. We show in this work that PscI, proposed to form the T3S system (T3SS) inner rod, possesses intrinsic properties to polymerize into flexible and regularly twisted fibrils and activates IL-1ß production in mouse bone marrow macrophages in vitro. We also found that point mutations within C-terminal amphipathic helix of PscI alter needle assembly in vitro and T3SS function in cell infection assays, suggesting that this region is essential for an efficient needle assembly. The overexpression of PscF partially compensates for the absence of the inner rod in PscI-deficient mutant by forming a secretion-proficient injectisome. All together, we propose that the polymerized PscI in P. aeruginosa optimizes the injectisome function by anchoring the needle within the envelope-embedded complex of the T3S secretome and - contrary to its counterpart in Salmonella - is not involved in substrate switching.


Bacterial Proteins/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/metabolism , Type III Secretion Systems/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice , Polymerization , Protein Transport , Pseudomonas Infections/genetics , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Type III Secretion Systems/chemistry , Type III Secretion Systems/genetics
20.
J Med Chem ; 57(24): 10275-89, 2014 Dec 26.
Article En | MEDLINE | ID: mdl-25419855

Pseudomonas aeruginosa lung infections are a major cause of death in cystic fibrosis and hospitalized patients. Treating these infections is becoming difficult due to the emergence of conventional antimicrobial multiresistance. While monosaccharides have proved beneficial against such bacterial lung infection, the design of several multivalent glycosylated macromolecules has been shown to be also beneficial on biofilm dispersion. In this study, calix[4]arene-based glycoclusters functionalized with galactosides or fucosides have been synthesized. The characterization of their inhibitory properties on Pseudomonas aeruginosa aggregation, biofilm formation, adhesion on epithelial cells, and destruction of alveolar tissues were performed. The antiadhesive properties of the designed glycoclusters were demonstrated through several in vitro bioassays. An in vivo mouse model of lung infection provided an almost complete protection against Pseudomonas aeruginosa with the designed glycoclusters.


Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Biofilms/drug effects , Calixarenes/chemistry , Lung/drug effects , Pseudomonas Infections/drug therapy , Respiratory Tract Infections/drug therapy , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Animals , Anti-Bacterial Agents/chemistry , Cells, Cultured , Fluorescent Antibody Technique , Glycosylation , Humans , Lectins/chemistry , Lectins/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Models, Chemical , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Respiratory Tract Infections/microbiology
...