25 research outputs found

    Omp25-dependent engagement of SLAMF1 by Brucella abortus in dendritic cells limits acute inflammation and favours bacterial persistence in vivo

    Get PDF
    The strategies by which intracellular pathogenic bacteria manipulate innate immunity to establish chronicity are poorly understood. Here, we show that Brucella abortus outer membrane protein Omp25 specifically binds the immune cell receptor SLAMF1 in vitro. The Omp25-dependent engagement of SLAMF1 by B. abortus limits NF-κB translocation in dendritic cells (DCs) with no impact on Brucella intracellular trafficking and replication. This in turn decreases pro-inflammatory cytokine secretion and impairs DC activation. The Omp25-SLAMF1 axis also dampens the immune response without affecting bacterial replication in vivo during the acute phase of Brucella infection in a mouse model. In contrast, at the chronic stage of infection, the Omp25/SLAMF1 engagement is essential for Brucella persistence. Interaction of a specific bacterial protein with an immune cell receptor expressed on the DC surface at the acute stage of infection is thus a powerful mechanism to support microbe settling in its replicative niche and progression to chronicity.Fil: Degos, Clara. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Hysenaj, Lisiena. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Gonzalez Espinoza, Gabriela. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Arce Gorvel, Vilma. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Gagnaire, Aurélie. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Papadopoulos, Alexia. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Pasquevich, Karina Alejandra. Universidad Nacional de San Martín. Instituto de Investigaciones Biotecnológicas. - Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Biotecnológicas; ArgentinaFil: Méresse, Stéphane. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Cassataro, Juliana. Universidad Nacional de San Martín. Instituto de Investigaciones Biotecnológicas. - Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Biotecnológicas; ArgentinaFil: Mémet, Sylvie. Inserm; Francia. Centre National de la Recherche Scientifique; FranciaFil: Gorvel, Jean Pierre. Inserm; Francia. Centre National de la Recherche Scientifique; Franci

    Endometrial Tumor Microenvironment Alters Human NK Cell Recruitment, and Resident NK Cell Phenotype and Function

    Get PDF
    Endometrial Cancer is the most common cancer in the female genital tract in developed countries, and with its increasing incidence due to risk factors such as aging and obesity tends to become a public health issue. However, its immune environment has been less characterized than in other tumors such as breast cancers. NK cells are cytotoxic innate lymphoid cells that are considered as a major anti-tumoral effector cell type which function is drastically altered in tumors which participates to tumor progression. Here we characterize tumor NK cells both phenotypically and functionally in the tumor microenvironment of endometrial cancer. For that, we gathered endometrial tumors, tumor adjacent healthy tissue, blood from matching patients and healthy donor blood to perform comparative analysis of NK cells. First we found that NK cells were impoverished in the tumor infiltrate. We then compared the phenotype of NK cells in the tumor and found that tumor resident CD103+ NK cells exhibited more co-inhibitory molecules such as Tigit, and TIM-3 compared to recruited CD103− NK cells and that the expression of these molecules increased with the severity of the disease. We showed that both chemokines (CXCL12, IP-10, and CCL27) and cytokines profiles (IL-1β and IL-6) were altered in the tumor microenvironment and might reduce NK cell function and recruitment to the tumor site. This led to hypothesize that the tumor microenvironment reduces resident NK cells cytotoxicity which we confirmed by measuring cytotoxic effector production and degranulation. Taken together, our results show that the tumor microenvironment reshapes NK cell phenotype and function to promote tumor progression

    Lipopolysaccharides with acylation defects potentiate TLR4 signaling and shape T cell responses

    Get PDF
    Lipopolysaccharides or endotoxins are components of Gram-negative enterobacteria that cause septic shock in mammals. However, a LPS carrying hexa-acyl lipid A moieties is highly endotoxic compared to a tetra-acyl LPS and the latter has been considered as an antagonist of hexa-acyl LPS-mediated TLR4 signaling. We investigated the relationship between the structure and the function of bacterial LPS in the context of human and mouse dendritic cell activation. Strikingly, LPS with acylation defects were capable of triggering a strong and early TLR4-dependent DC activation, which in turn led to the activation of the proteasome machinery dampening the pro-inflammatory cytokine secretion. Upon activation with tetra-acyl LPS both mouse and human dendritic cells triggered CD4(+) T and CD8(+) T cell responses and, importantly, human myeloid dendritic cells favored the induction of regulatory T cells. Altogether, our data suggest that LPS acylation controlled by pathogenic bacteria might be an important strategy to subvert adaptive immunity

    Escherichia coli α-Hemolysin Counteracts the Anti-Virulence Innate Immune Response Triggered by the Rho GTPase Activating Toxin CNF1 during Bacteremia

    Get PDF
    International audienceThe detection of the activities of pathogen-encoded virulence factors by the innate immune system has emerged as a new paradigm of pathogen recognition. Much remains to be determined with regard to the molecular and cellular components contributing to this defense mechanism in mammals and importance during infection. Here, we reveal the central role of the IL-1 beta signaling axis and Gr1+ cells in controlling the Escherichia coli burden in the blood in response to the sensing of the Rho GTPase-activating toxin CNF1. Consistently, this innate immune response is abrogated in caspase-1/11-impaired mice or following the treatment of infected mice with an IL-1 beta antagonist. In vitro experiments further revealed the synergistic effects of CNF1 and LPS in promoting the maturation/secretion of IL-1 beta and establishing the roles of Rac, ASC and caspase-1 in this pathway. Furthermore, we found that the Phi-hemolysin toxin inhibits IL-1 beta secretion without affecting the recruitment of Gr1+ cells. Here, we report the first example of anti-virulence-triggered immunity counteracted by a pore-forming toxin during bacteremia

    Control and modulation of the immune response by Brucella abortus

    No full text
    Brucella est une bactérie pathogène intracellulaire responsable d'une maladie, la brucellose. Sa capacité à établir une infection chronique est due aux mécanismes qu'elle déploie pour inhiber la réponse immunitaire. Parmi les cellules infectées, les cellules dendritiques (DC) et les macrophages (MO) jouent un rôle primordial dans l'induction de la réponse immunitaire. Nous avons étudié le rôle d'un récepteur des DC, des lymphocytes T (LT), MO : CD150. Il participe à l'activation des LT et il a été montré que CD150 est capable de reconnaître des protéines de l'enveloppe bactérienne, ce qui conduit à une augmentation de son expression à la membrane des MO. Nous avons découvert que l'expression de CD150 sur les DC dérivées de moelle osseuse (BMDC) augmente en présence d'extraits membranaire de Brucella, sauf quand ces derniers proviennent d'un mutant pour Omp25 (∆omp25). L'infection de BMDC par ∆omp25 active les BMDC en termes d'expression de molécules de co-stimulation, d'ARNm pro-inflammatoires (cytokines) et de translocation de NF-κB dans le noyau. En comparant avec une souche sauvage de Brucella, l'activation par ∆omp25 est plus importante. En absence de CD150 la translocation de NF-κB dans les BMDC infectées par la souche sauvage est aussi importante que celle induite par l'infection par ∆omp25. In vivo CD150 contrôle la réplication de Brucella dans la rate de souris infectées. Nous avons démontré que CD150 est capable de lier Omp25. Nous avons identifié un nouveau mécanisme par lequel Brucella inhibe l'activation des DC : la liaison d'Omp25 à CD150. Ce récepteur joue un double rôle puisqu'il est aussi crucial dans le contrôle de la survie de Brucella in vivo.Brucella is a pathogenic intracellular bacterium responsible for a disease, brucellosis. The ability of Brucella to establish a chronic infection is linked to the mechanism it uses to inhibit immune response. Among Brucella infected cells, dendritic cells (DC) and macrophages play a crucial role in the induction of an immune response. We studied the role of one receptor present at the DC, T cells, macrophages surface: CD150. This molecule participates at the T cell activation and it was shown recently that CD150 can recognize bacterial membrane proteins which lead to its own upregulation. We discovered that CD150 expression onto bone-marrow derived DC (BMDC) is increased when these cells are treated with Brucella membrane extracts, except when those extracts are coming from a mutant for Omp25(∆omp25), a Brucella membrane protein. BMDC infection with ∆omp25 leads to BMDC activation regarding co-stimulatory molecule expression, cytokines and chemokines secretion, pro-inflammatory mRNA expression and NF-κB translocation within the nucleus. In comparison with infection with the wild type strain, ∆omp25 induces a higher activation of BMDC. In absence of CD150, NF-κB translocation within the nucleus of infected-BMDC is the same between the wild type strain and ∆omp25. In vivo, CD150 controls Brucella replication in the spleen of infected mice, and Omp25 infection seems to trigger a higher inflammation in control mice. We finally demonstrate that CD150 binds Omp25. Here, we identified a new mechanism by which Brucella is able to inhibit DC activation: binding of Omp25 to CD150. This receptor plays a dual role since it is also required for controlling Brucella growth in mice

    Brucella discriminates between mouse dendritic cell subsets upon in vitro infection

    No full text
    International audienceBrucella is a Gram-negative bacterium responsible for brucellosis, a worldwide re-emerging zoonosis. Brucella has been shown to infect and replicate within Granulocyte macrophage colony-stimulating factor (GMCSF) in vitro grown bone marrow-derived dendritic cells (BMDC). In this cell model, Brucella can efficiently control BMDC maturation. However, it has been shown that Brucella infection in vivo induces spleen dendritic cells (DC) migration and maturation. As DCs form a complex network composed by several subpopulations, differences observed may be due to different interactions between Brucella and DC subsets. Here, we compare Brucella interaction with several in vitro BMDC models. The present study shows that Brucella is capable of replicating in all the BMDC models tested with a high infection rate at early time points in GMCSF-IL15 DCs and Flt3l DCs. GMCSF-IL15 DCs and Flt3l DCs are more activated than the other studied DC models and consequently intracellular bacteria are not efficiently targeted to the ER replicative niche. Interestingly, GMCSF-DC and GMCSF-Flt3l DC response to infection is comparable. However, the key difference between these 2 models concerns IL10 secretion by GMCSF DCs observed at 48h post-infection. IL10 secretion can explain the weak secretion of IL12p70 and TNF in the GMCSF-DC model and the low level of maturation observed when compared to GMCSF-IL15 DCs and Flt3l DCs. These models provide good tools to understand how Brucella induce DC maturation in vivo and may lead to new therapeutic design using DCs as cellular vaccines capable of enhancing immune response against pathogens

    LPS with acylation defects induce semi-mature mouse and human dendritic cells.

    No full text
    <p>Mouse BMDC were stimulated for 8 h (in grey) and 24 h (in black) with medium, <i>E. coli</i> LPS (either hexa-acyl, penta-acyl or tetra-acyl) and <i>Y. pestis</i> tetra-acyl LPS. All LPS were used at the concentration of 100 ng/ml. MHC II and co-stimulatory molecules up-regulation on the cell surface was measured by flow cytometry (A) and cytokine secretion was determined by ELISA (B). Data represent means ± standard errors of at least 5 independent experiments, **p<0.01, *p = 0.01 to 0.05. Human blood mDC were stimulated overnight with medium (in grey), hexa-acyl <i>E. coli</i> LPS (in red), tetra-acyl <i>E. coli</i> LPS (in blue) and <i>Y. pestis</i> tetra-acyl LPS (in orange). Surface expression of HLA-DR, CD83, CD40 and CD86 was analyzed by flow cytometry (C) and cytokine levels in the culture supernatants were measured by Luminex (D). Experiments were performed on 4 different donors. The data for one representative are shown. ***p<0.001, **p<0.01, *p = 0.01 to 0.05.</p

    Tetra-acyl LPS induce the activation of TLR4-dependent molecular pathways involved in mouse DC maturation.

    No full text
    <p>BMDC were activated with medium (grey), <i>E. coli</i> hexa-acyl LPS (dark blue), <i>E. coli</i> tetra-acyl LPS (purple) or <i>Y. pestis</i> tetra-acyl LPS (light blue) for 15 min, 30 min, 1 h and 2 h. NF-κB translocation was analyzed by confocal microscopy(A). Cells were fixed and stained for CD11c (in blue), MHC-II (in green) and NF-κB subunit p65/RelA (in red). The percentage of BMDC with translocated NF-κB into the nucleus was quantified (B). BMDC were stimulated for 30 min, 1 h, 4 h and 6 h with medium or different LPS. Cell lysates were subjected to SDS-PAGE and, after transfer to nitrocellulose, the membrane was probed with the antibodies against phospho-S6 (Ser235/236), S6 and an anti-actin antibody (C). Data represent means ± standard errors of at least 4 independent experiments, **p<0.01.</p

    LPS with acylation defects induce functional mouse and human dendritic cells.

    No full text
    <p>BMDC were incubated overnight with OVA and activated for 8 h with different LPS. Stimulated DC were co-cultured with T cells from OT-I and OT-II <i>Rag-2<sup>−/−</sup></i> mice (A). The proliferation of OT-I and OT-II T cells was assessed after 3 days of co-culture by CFSE decrease. For T cell activation assays, the expression of surface markers such as CD25 and CD62L was analyzed by flow cytometry. At least 3 independent experiments were performed and one representative is shown. (B) CFSE-labeled allogeneic naïve CD4<sup>+</sup> T and CD8<sup>+</sup> T cells were co-cultured with activated mDC for 7 days. Cell division was tested by measuring CFSE-dilution Experiments were performed on 4 different donors. Data for one representative experiment are shown.</p
    corecore