Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Eur J Immunol ; 53(2): e2249985, 2023 02.
Article in English | MEDLINE | ID: mdl-36427489

ABSTRACT

Flagellin-induced NAIP/NLRC4 inflammasome activation and pyroptosis are critical events restricting Legionella pneumophila infection. However, the cellular and molecular dynamics of the in vivo responses against this bacterium are still unclear. We have found temporal coordination of two independent innate immunity pathways in controlling Legionella infection, the inflammasome activation and the CCR2-mediated Mo-DC recruitment. Inflammasome activation was an important player at the early stage of infection by lowering the numbers of bacteria for an efficient bacterial clearance conferred by the Mo-DC at the late stage of the infection. Mo-DC emergence highly depended on CCR2-signaling and dispensed inflammasome activation and pyroptosis. Also, Mo-DC compartment did not rely on the inflammasome machinery to deliver proper immune responses and was the most abundant cytokine-producing among the monocyte-derived cells in the infected lung. Importantly, when the CCR2- and NLRC4-dependent axes of response were simultaneously ablated, we observed an aggravated bacterial burden in the lung of infected mice. Taken together, we showed that inflammasome activation and CCR2-mediated immune response interplay in distinct pathways to restrict pulmonary bacterial infection. These findings extend our understanding of the in vivo integration and cooperation of different innate immunity arms in controlling infectious agents.


Subject(s)
Dendritic Cells , Inflammasomes , Legionella pneumophila , Legionnaires' Disease , Monocytes , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Dendritic Cells/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Legionella pneumophila/immunology , Legionnaires' Disease/genetics , Legionnaires' Disease/immunology , Macrophages , Mice, Knockout , Monocytes/metabolism , Receptors, CCR2/metabolism
2.
PLoS Pathog ; 13(8): e1006502, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28771586

ABSTRACT

Legionella pneumophila is a Gram-negative, flagellated bacterium that survives in phagocytes and causes Legionnaires' disease. Upon infection of mammalian macrophages, cytosolic flagellin triggers the activation of Naip/NLRC4 inflammasome, which culminates in pyroptosis and restriction of bacterial replication. Although NLRC4 and caspase-1 participate in the same inflammasome, Nlrc4-/- mice and their macrophages are more permissive to L. pneumophila replication compared with Casp1/11-/-. This feature supports the existence of a pathway that is NLRC4-dependent and caspase-1/11-independent. Here, we demonstrate that caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in response to flagellin-positive bacteria. Accordingly, caspase-8 is activated in Casp1/11-/- macrophages in a process dependent on flagellin, Naip5, NLRC4 and ASC. Silencing caspase-8 in Casp1/11-/- cells culminated in macrophages that were as susceptible as Nlrc4-/- for the restriction of L. pneumophila replication. Accordingly, macrophages and mice deficient in Asc/Casp1/11-/- were more susceptible than Casp1/11-/- and as susceptible as Nlrc4-/- for the restriction of infection. Mechanistically, we found that caspase-8 activation triggers gasdermin-D-independent pore formation and cell death. Interestingly, caspase-8 is recruited to the Naip5/NLRC4/ASC inflammasome in wild-type macrophages, but it is only activated when caspase-1 or gasdermin-D is inhibited. Our data suggest that caspase-8 activation in the Naip5/NLRC4/ASC inflammasome enable induction of cell death when caspase-1 or gasdermin-D is suppressed.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Caspase 1/immunology , Caspase 8/immunology , Inflammasomes/immunology , Legionnaires' Disease/immunology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins , Caspase 1/metabolism , Caspase 8/metabolism , Disease Models, Animal , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins , Legionella pneumophila , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Apoptosis-Inhibitory Protein , Phosphate-Binding Proteins , Real-Time Polymerase Chain Reaction
3.
J Infect Dis ; 215(3): 440-451, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27932612

ABSTRACT

BACKGROUND: Legionella longbeachae (Llo) and Legionella pneumophila (Lpn) are the most common pneumonia-causing agents of the genus. Although both species can be lethal to humans and are highly prevalent, little is known about the molecular pathogenesis of Llo infections. In murine models of infection, Lpn infection is self-limited, whereas Llo infection is lethal. METHODS: We used mouse macrophages, human macrophages, human epithelial cells, and mouse infections in vivo to evaluate multiple parameters of the infection. RESULTS: We determined that the Llo Dot/Icm secretion system is critical for virulence. Different than Lpn, Llo disseminates and the animals develop a severe pulmonary failure, as demonstrated by lung mechanics and blood oxygenation assays. As compared to Lpn, Llo is immunologically silent and fails to trigger the production of cytokines in human pulmonary epithelial cells and in mouse and human macrophages. Infections in Tnfr1-/-, Ifng-/-, and Il12p40-/- mice supported the participation of cytokines for the resistance phenotype. CONCLUSIONS: Both Lpn and Llo require the Dot/Icm system for pathogenesis, but the infection outcome is strikingly different. Llo is immunologically silent, highly virulent, and lethal. The differences reported herein may reflect unappreciated clinical differences in patients infected with Lpn or Llo.


Subject(s)
Legionella longbeachae/immunology , Legionella longbeachae/pathogenicity , Legionellosis/immunology , Animals , Cytokines/metabolism , Disease Resistance/immunology , Female , Humans , Legionella pneumophila/immunology , Legionellosis/microbiology , Legionellosis/pathology , Legionellosis/physiopathology , Leukocytes, Mononuclear , Lung/physiopathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Species Specificity , Virulence
4.
J Infect Dis ; 211(2): 322-30, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25104770

ABSTRACT

Legionella pneumophila, the etiological agent of Legionnaires' disease, triggers activation of multiple innate immune pathways that lead to the restriction of bacterial replication in vivo. Despite the critical role for MyD88 in infection clearance, the receptors and mechanisms responsible for MyD88-mediated pulmonary bacterial clearance are still unclear. Here, we used flagellin mutants of L. pneumophila, which bypass the NAIP5/NLRC4-mediated restriction of bacterial replication, to assess the receptors involved in MyD88-mediated pulmonary bacterial clearance. By systematically comparing pulmonary clearance of L. pneumophila in C57BL/6 MyD88(-/-), TLR2(-/-), TLR3(-/-), TLR4(-/-), TLR9(-/-), IL-1R(-/-), and IL-18(-/-) mice, we found that, while the knockout of a single Toll-like receptor or interleukin 18 resulted only in minor impairment of bacterial clearance, deficiency in the interleukin 1 (IL-1) receptor led to a significant impairment. IL-1/MyD88-mediated pulmonary bacterial clearance occurs via processes involving the recruitment of neutrophils. Collectively, our data contribute to the understanding of the effector mechanisms involved in MyD88-mediated pulmonary bacterial clearance.


Subject(s)
Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Lung/immunology , Myeloid Differentiation Factor 88/metabolism , Neutrophil Infiltration , Receptors, Interleukin-1/metabolism , Animals , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL