Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 7246, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945612

ABSTRACT

NLRP3 induces caspase-1-dependent pyroptotic cell death to drive inflammation. Aberrant activity of NLRP3 occurs in many human diseases. NLRP3 activation induces ASC polymerization into a single, micron-scale perinuclear punctum. Higher resolution imaging of this signaling platform is needed to understand how it induces pyroptosis. Here, we apply correlative cryo-light microscopy and cryo-electron tomography to visualize ASC/caspase-1 in NLRP3-activated cells. The puncta are composed of branched ASC filaments, with a tubular core formed by the pyrin domain. Ribosomes and Golgi-like or endosomal vesicles permeate the filament network, consistent with roles for these organelles in NLRP3 activation. Mitochondria are not associated with ASC but have outer-membrane discontinuities the same size as gasdermin D pores, consistent with our data showing gasdermin D associates with mitochondria and contributes to mitochondrial depolarization.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Electron Microscope Tomography , Gasdermins , Caspase 1/metabolism , Caspases/metabolism , Pyroptosis , Organelles/metabolism
2.
Cell Rep ; 36(8): 109614, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433041

ABSTRACT

Zoonotic pathogens, such as COVID-19, reside in animal hosts before jumping species to infect humans. The Carnivora, like mink, carry many zoonoses, yet how diversity in host immune genes across species affect pathogen carriage is poorly understood. Here, we describe a progressive evolutionary downregulation of pathogen-sensing inflammasome pathways in Carnivora. This includes the loss of nucleotide-oligomerization domain leucine-rich repeat receptors (NLRs), acquisition of a unique caspase-1/-4 effector fusion protein that processes gasdermin D pore formation without inducing rapid lytic cell death, and the formation of a caspase-8 containing inflammasome that inefficiently processes interleukin-1ß. Inflammasomes regulate gut immunity, but the carnivorous diet has antimicrobial properties that could compensate for the loss of these immune pathways. We speculate that the consequences of systemic inflammasome downregulation, however, can impair host sensing of specific pathogens such that they can reside undetected in the Carnivora.


Subject(s)
Carnivora/metabolism , Evolution, Molecular , Inflammasomes/metabolism , Zoonoses/pathology , Animals , Caspase 1/genetics , Caspase 1/metabolism , Caspase 8/metabolism , Caspases, Initiator/genetics , Caspases, Initiator/metabolism , Cell Death , Cell Line , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NLR Proteins/genetics , NLR Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salmonella typhi/pathogenicity , Zoonoses/immunology , Zoonoses/parasitology
3.
Nat Microbiol ; 5(12): 1588-1597, 2020 12.
Article in English | MEDLINE | ID: mdl-33106673

ABSTRACT

Pattern recognition receptors (PRRs) expressed in antigen-presenting cells are thought to shape pathogen-specific immunity by inducing secretion of costimulatory cytokines during T-cell activation, yet data to support this notion in vivo are scarce. Here, we show that the cytosolic PRR Nod-like Receptor CARD 4 (NLRC4) suppresses, rather than facilitates, effector and memory CD4+ T-cell responses against Salmonella in mice. NLRC4 negatively regulates immunological memory by preventing delayed activation of the cytosolic PRR NLR pyrin domain 3 (NLRP3) that would otherwise amplify the production of cytokines important for the generation of Th1 immunity such as intereukin-18. Consistent with a role for NLRC4 in memory immunity, primary challenge with Salmonella expressing flagellin modified to largely evade NLRC4 recognition notably increases protection against lethal rechallenge. This finding suggests flagellin modification to reduce NLRC4 activation enhances protective immunity, which could have important implications for vaccine development against flagellated microbial pathogens.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Flagellin/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Female , Flagellin/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Interleukin-18/genetics , Interleukin-18/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , T-Lymphocytes/immunology , Th1 Cells/immunology
4.
J Biol Chem ; 295(31): 10857-10867, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32111741

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) encodes a complex protein that includes kinase and GTPase domains. Genome-wide association studies have identified dominant LRRK2 alleles that predispose their carriers to late-onset idiotypic Parkinson's disease (PD) and also to autoimmune disorders such as Crohn's disease. Considerable evidence indicates that PD initiation and progression involve activation of innate immune functions in microglia, which are brain-resident macrophages. Here we asked whether LRRK2 modifies inflammatory signaling and how this modification might contribute to PD and Crohn's disease. We used RNA-Seq-based high-resolution transcriptomics to compare gene expression in activated primary macrophages derived from WT and Lrrk2 knockout mice. Remarkably, expression of a single gene, Rap guanine nucleotide exchange factor 3 (Rapgef3), was strongly up-regulated in the absence of LRRK2 and down-regulated in its presence. We observed similar regulation of Rapgef3 expression in cells treated with a highly specific inhibitor of LRRK2 protein kinase activity. Rapgef3 encodes an exchange protein, activated by cAMP 1 (EPAC-1), a guanine nucleotide exchange factor that activates the small GTPase Rap-1. Rap-1 mediates cell adhesion, polarization, and directional motility, and our results indicate that LRRK2 modulates chemotaxis of microglia and macrophages. Dominant PD-associated LRRK2 alleles may suppress EPAC-1 activity, further restricting motility and preventing efficient migration of microglia to sites of neuronal damage. Functional analysis in vivo in a subclinical infection model also indicated that Lrrk2 subtly modifies the inflammatory response. These results indicate that LRRK2 modulates the expression of genes involved in murine immune cell chemotaxis.


Subject(s)
Cell Adhesion , Cell Polarity , Chemotaxis , Gene Expression Regulation , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Macrophage Activation , Macrophages/enzymology , Animals , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Mice , Mice, Knockout , Microglia/enzymology , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 111(20): 7403-8, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24803432

ABSTRACT

Pathogen recognition by nucleotide-binding oligomerization domain-like receptor (NLR) results in the formation of a macromolecular protein complex (inflammasome) that drives protective inflammatory responses in the host. It is thought that the number of inflammasome complexes forming in a cell is determined by the number of NLRs being activated, with each NLR initiating its own inflammasome assembly independent of one another; however, we show here that the important foodborne pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) simultaneously activates at least two NLRs, whereas only a single inflammasome complex is formed in a macrophage. Both nucleotide-binding domain and leucine-rich repeat caspase recruitment domain 4 and nucleotide-binding domain and leucine-rich repeat pyrin domain 3 are simultaneously present in the same inflammasome, where both NLRs are required to drive IL-1ß processing within the Salmonella-infected cell and to regulate the bacterial burden in mice. Superresolution imaging of Salmonella-infected macrophages revealed a macromolecular complex with an outer ring of apoptosis-associated speck-like protein containing a caspase activation and recruitment domain and an inner ring of NLRs, with active caspase effectors containing the pro-IL-1ß substrate localized internal to the ring structure. Our data reveal the spatial localization of different components of the inflammasome and how different members of the NLR family cooperate to drive robust IL-1ß processing during Salmonella infection.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/microbiology , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Caspase 8/metabolism , Inflammasomes/physiology , Macrophages/microbiology , Animals , Apoptosis , Enzyme Activation , HEK293 Cells , Humans , Inflammation , Interleukin-1beta/metabolism , Mice , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Salmonella typhimurium
6.
Immunology ; 111(2): 230-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15027909

ABSTRACT

Scrapie is a transmissible spongiform encephalopathy in which there is an accumulation of the abnormal form of the prion protein, PrPsc, in the lymphoreticular system and nervous system. There is a particular accumulation of PrPsc on follicular dendritic cells within the germinal centre of B-cell follicles. Because accumulation of PrPsc in the nervous system leads to neuronal cell loss we have examined PrPsc accumulation in the prescapular and mesenteric lymph nodes in relation to lymph node architecture of scrapie-challenged sheep. We demonstrate that an accumulation of PrPsc in the lymph node fails to result in gross defects in the microanatomy and phenotype of T- and B-cell areas in the lymph nodes.


Subject(s)
Lymph Nodes/immunology , Scrapie/immunology , Animals , B-Lymphocytes/pathology , Female , Immunophenotyping , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Mesentery , PrPSc Proteins/metabolism , Scapula , Scrapie/metabolism , Scrapie/pathology , Sheep , T-Lymphocyte Subsets/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...