Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
2.
PLoS Pathog ; 11(8): e1005078, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26241037

ABSTRACT

Differential modulation of NF-κB during meningococcal infection is critical in innate immune response to meningococcal disease. Non-invasive isolates of Neisseria meningitidis provoke a sustained NF-κB activation in epithelial cells. However, the hyperinvasive isolates of the ST-11 clonal complex (ST-11) only induce an early NF-κB activation followed by a sustained activation of JNK and apoptosis. We show that this temporal activation of NF-κB was caused by specific cleavage at the C-terminal region of NF-κB p65/RelA component within the nucleus of infected cells. This cleavage was mediated by the secreted 150 kDa meningococcal ST-11 IgA protease carrying nuclear localisation signals (NLS) in its α-peptide moiety that allowed efficient intra-nuclear transport. In a collection of non-ST-11 healthy carriage isolates lacking NLS in the α-peptide, secreted IgA protease was devoid of intra-nuclear transport. This part of iga polymorphism allows non-invasive isolates lacking NLS, unlike hyperinvasive ST-11 isolates of N. meningitides habouring NLS in their α-peptide, to be carried asymptomatically in the human nasopharynx through selective eradication of their ability to induce apoptosis in infected epithelial cells.


Subject(s)
Bacterial Proteins/metabolism , Cell Nucleus/metabolism , Meningococcal Infections/metabolism , Serine Endopeptidases/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/physiology , Apoptosis/immunology , Bacterial Proteins/immunology , Cell Line , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Meningococcal Infections/immunology , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Neisseria meningitidis/immunology , Neisseria meningitidis/metabolism , Neisseria meningitidis/pathogenicity , Nuclear Localization Signals , Real-Time Polymerase Chain Reaction , Transfection
3.
iScience ; 27(8): 110441, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39104410

ABSTRACT

Coordinating immune responses - humoral and cellular - is vital for protection against severe Covid-19. Our study evaluates a multicytokine CD4+T cell signature's predictive for post-vaccinal serological and CD8+T cell responses. A cytokine signature composed of four cytokines (IL-2, TNF-α, IP10, IL-9) excluding IFN-γ, and generated through machine learning, effectively predicted the CD8+T cell response following mRNA-1273 or BNT162b2 vaccine administration. Its applicability extends to murine vaccination models, encompassing diverse immunization routes (such as intranasal) and vaccine platforms (including adjuvanted proteins). Notably, we found correlation between CD4+T lymphocyte-produced IL-21 and the humoral response. Consequently, we propose a test that offers a rapid overview of integrated immune responses. This approach holds particular relevance for scenarios involving immunocompromised patients because they often have low cell counts (lymphopenia) or pandemics. This study also underscores the pivotal role of CD4+T cells during a vaccine response and highlights their value in vaccine immunomonitoring.

4.
Mol Immunol ; 118: 52-59, 2020 02.
Article in English | MEDLINE | ID: mdl-31855807

ABSTRACT

BACKGROUND: Interleukin (IL)-22 is a cytokine involved in tissue protection and repair following lung pathologies. Interferon (IFN)-λ cytokines displayed similar properties during viral infection and a synergy of action between these two players has been documented in the intestine. We hypothesize that during Pseudomonas aeruginosa challenge, IL-22 up-regulates IFN-λ and that IFN-λ exhibits protective functions during Pseudomonas aeruginosa acute pneumonia model in mice. METHODS: Using an in vitro human alveolar epithelial cell line A549, we assessed the ability of IL-22 to enhance IFN-λ expression during infection. IFN-λ protective function was evaluated in an acute mouse pneumonia model. RESULTS: We first demonstrated in murine lungs that only type-II alveolar cells express IL-22 receptor and that IL-22 treatment of A549 cell line up-regulates IFN-λ expression. In a murine acute pneumonia model, IL-22 administration maintained significant IFN-λ levels in the broncho-alveolar fluids whereas IL-22 neutralization abolished IFN-λ up-regulation. In vivo administration of IFN-λ during Pseudomonas aeruginosa pneumonia improves mice outcome by dampening neutrophil recruitment and decreasing epithelium damages. DISCUSSION: We show here that IL-22 regulates IFN-λ levels during Pseudomonas aeruginosa pneumonia.


Subject(s)
Interferons/immunology , Interleukins/immunology , Pneumonia/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , A549 Cells , Alveolar Epithelial Cells/immunology , Animals , Bronchi/immunology , Cell Line, Tumor , Cytokines/immunology , Disease Models, Animal , Female , Humans , Lung/immunology , Mice , Neutrophil Infiltration/immunology , Receptors, Interleukin/immunology , Up-Regulation/immunology , Interleukin-22
5.
Sci Rep ; 7(1): 11010, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28887540

ABSTRACT

Pseudomonas aeruginosa is a major threat for immune-compromised patients. Bacterial pneumonia can induce uncontrolled and massive neutrophil recruitment ultimately leading to acute respiratory distress syndrome and epithelium damage. Interleukin-22 plays a central role in the protection of the epithelium. In this study, we aimed to evaluate the role of interleukin-22 and its soluble receptor IL-22BP in an acute Pseudomonas aeruginosa pneumonia model in mice. In this model, we noted a transient increase of IL-22 during Pseudomonas aeruginosa challenge. Using an antibody-based approach, we demonstrated that IL-22 neutralisation led to increased susceptibility to infection and to lung damage correlated with an increase in neutrophil accumulation in the lungs. On the contrary, rIL-22 administration or IL-22BP neutralisation led to a decrease in mouse susceptibility and lung damage associated with a decrease in neutrophil accumulation. This study demonstrated that the IL-22/IL-22BP system plays a major role during Pseudomonas aeruginosa pneumonia by moderating neutrophil accumulation in the lungs that ultimately leads to epithelium protection.


Subject(s)
Interleukins/analysis , Lung/pathology , Neutrophil Infiltration , Pneumonia, Bacterial/pathology , Pseudomonas Infections/pathology , Receptors, Interleukin/analysis , Animals , Disease Models, Animal , Mice , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL