Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 17(820): eabg8145, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38261657

ABSTRACT

Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome-nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form "specks," and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant-associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1ß in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.


Subject(s)
Hereditary Autoinflammatory Diseases , Inflammasomes , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing , Intracellular Signaling Peptides and Proteins , Leukocytes, Mononuclear , NLR Family, Pyrin Domain-Containing 3 Protein , Syndrome
2.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Article in English | MEDLINE | ID: mdl-35007292

ABSTRACT

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Macrophages/microbiology , Vacuoles/microbiology , Virulence/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL
3.
Immunol Cell Biol ; 99(10): 1067-1076, 2021 11.
Article in English | MEDLINE | ID: mdl-34555867

ABSTRACT

The proinflammatory cytokine tumor necrosis factor (TNF) plays a central role in the host control of mycobacterial infections. Expression and release of TNF are tightly regulated, yet the molecular mechanisms that control the release of TNF by mycobacteria-infected host cells, in particular macrophages, are incompletely understood. Rab GTPases direct the transport of intracellular membrane-enclosed vesicles and are important regulators of macrophage cytokine secretion. Rab6b is known to be predominantly expressed in the brain where it functions in retrograde transport and anterograde vesicle transport for exocytosis. Whether it executes similar functions in the context of immune responses is unknown. Here we show that Rab6b is expressed by primary mouse macrophages, where it localized to the Golgi complex. Infection with Mycobacterium bovis bacille Calmette-Guérin (BCG) resulted in dynamic changes in Rab6b expression in primary mouse macrophages in vitro as well as in organs from infected mice in vivo. We further show that Rab6b facilitated TNF release by M. bovis BCG-infected macrophages, in the absence of discernible impact on Tnf messenger RNA and intracellular TNF protein expression. Our observations identify Rab6b as a positive regulator of M. bovis BCG-induced TNF trafficking and secretion by macrophages and positions Rab6b among the molecular machinery that orchestrates inflammatory cytokine responses by macrophages.


Subject(s)
Golgi Apparatus/immunology , Macrophages/immunology , Mycobacterium Infections , Tumor Necrosis Factor-alpha/immunology , rab GTP-Binding Proteins/immunology , Animals , Mice , Mycobacterium Infections/immunology , Mycobacterium bovis
4.
Nat Chem Biol ; 15(6): 556-559, 2019 06.
Article in English | MEDLINE | ID: mdl-31086327

ABSTRACT

Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and specific small-molecule inhibitor of the NLRP3 pathway, but its molecular target is not defined. Here, we show that MCC950 directly interacts with the Walker B motif within the NLRP3 NACHT domain, thereby blocking ATP hydrolysis and inhibiting NLRP3 activation and inflammasome formation.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/pharmacology , Adenosine Triphosphate/metabolism , Binding Sites/drug effects , Furans , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Hydrolysis/drug effects , Indenes , Inflammasomes/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sulfonamides , Sulfones/chemistry
5.
Curr Protoc Immunol ; 114: 14.40.1-14.40.29, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27479658

ABSTRACT

Inflammasomes are large protein complexes formed in response to cellular stresses that are platforms for recruitment and activation of caspase 1. Central to most inflammasome functions is the adapter molecule ASC (apoptosis-associated speck-like protein containing a caspase-recruitment domain) that links the inflammasome initiator protein to the recruited caspases. ASC is normally diffuse within the cell but within minutes of inflammasome activation relocates to a dense speck in the cytosol. The dramatic redistribution of ASC can be monitored by flow cytometry using parameters of fluorescence peak height and width when immunostained or tagged with a fluorescent protein. This can be used to define cells with active inflammasomes within populations of primary macrophages and monocytes, allowing quantification of responses and flow-sorting of responding cells. Protein structural requirements for ASC speck formation and recruitment of caspases to ASC specks can be assessed by expressing components in HEK293 cells. This provides rapid quantification of responding cell number and correlation with the expression level of inflammasome components within single cells. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Inflammasomes/metabolism , Macrophages/immunology , Monocytes/immunology , Animals , Apoptosis , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , HEK293 Cells , Humans , Single-Cell Analysis
6.
Immunobiology ; 221(2): 341-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26514298

ABSTRACT

NOD-like receptors (NLR) are a family of cytosolic pattern recognition receptors that include many key drivers of innate immune responses. NLRP12 is an emerging member of the NLR family that is closely related to the well-known inflammasome scaffold, NLRP3. Since its discovery, various functions have been proposed for NLRP12, including the positive regulation of dendritic cell (DC) and neutrophil migration and the inhibition of NF-κB and ERK signalling in DC and macrophages. We show here that NLRP12 is poorly expressed in murine macrophages and DC, but is strongly expressed in neutrophils. Using myeloid cells from WT and Nlrp12(-/)(-) mice, we show that, contrary to previous reports, NLRP12 does not suppress LPS- or infection-induced NF-κB or ERK activation in myeloid cells, and is not required for DC migration in vitro. Surprisingly, we found that Nlrp12 deficiency caused increased rather than decreased neutrophil migration towards the chemokine CXCL1 and the neutrophil parasite Leishmania major, revealing NLRP12 as a negative regulator of directed neutrophil migration under these conditions.


Subject(s)
Cell Movement/immunology , Chemokine CXCL1/immunology , Intracellular Signaling Peptides and Proteins/immunology , Neutrophils/immunology , Animals , Cell Movement/drug effects , Chemokine CXCL1/genetics , Dendritic Cells/immunology , Dendritic Cells/parasitology , Female , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Leishmania major/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Neutrophils/parasitology , Organ Specificity , Signal Transduction
7.
Immunobiology ; 217(12): 1325-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22898390

ABSTRACT

Macrophage pre-treatment with bacterial lipopolysaccharide (LPS) boosts subsequent activation of the NLRP3 inflammasome, which controls caspase-1-dependent pro-inflammatory cytokine maturation. Previous work has attributed this phenomenon (known as LPS 'priming') to LPS-dependent induction of NLRP3 expression. Whilst this plays a role, here we demonstrate that rapid LPS priming of NLRP3 inflammasome activation can occur independently of NLRP3 induction, since the priming effect of LPS is still apparent at short pre-treatment times in which NLRP3 protein expression remains unchanged. Furthermore, rapid LPS priming is still evident in Nlrp3(-/-) primary macrophages with NLRP3 expression reconstituted using a constitutive promoter. Similarly, we found that LPS potentiates AIM2 inflammasome activation to submaximal doses of cytosolic DNA without concomitant upregulation of AIM2 protein expression. Our data suggest that, in addition to augmenting NLRP3 inflammasome activity via NLRP3 induction, LPS boosts caspase-1 activation by the NLRP3 and AIM2 inflammasomes by an acute mechanism that is independent of inflammasome sensor induction.


Subject(s)
Carrier Proteins/genetics , Inflammasomes/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Animals , Carrier Proteins/agonists , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , DNA/genetics , DNA-Binding Proteins , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology
8.
Immunol Lett ; 134(2): 174-82, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-20933011

ABSTRACT

FCRL6 receptor is a more recently identified representative of the FCRL family. We generated a panel of mouse mAbs to baculovirus-derived recombinant FCRL6 protein. The clone 7B2 was found to specifically recognize a 63kDa protein expressed preferentially on the surface of CD8 T and CD56 NK cells in human peripheral blood and spleen. The clone 7B2 reacts with FCRL6 in Western blotting, FACS, and immunohistochemistry. In the T cell lineage, FCRL6 functions in antigen-experienced cells. Mitogenic stimulation of PB leukocytes in vitro resulted in an abrogation of the FCRL6 gene expression. We found a significant decrease in the FCRL6 gene expression in peripheral T cells of patients with certain autoimmune and blood diseases, and its upregulation at the late stages of HIV infection. Study of the FCRL6 association with signaling molecules showed its ability to recruit SHP-1, SHP-2, SHIP-1, and SHIP-2 phosphatases, and also adaptor protein Grb2 through phosphorylated cytoplasmic tyrosines. The current results demonstrate inhibitory potential of FCRL6 and suggest its possible involvement in modulation of CTL effector functions in various immune disorders.


Subject(s)
Carrier Proteins/immunology , Gene Expression Regulation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Alternative Splicing , Amino Acid Sequence , Autoimmune Diseases/immunology , Blood Cells/cytology , CD8-Positive T-Lymphocytes/immunology , Hematologic Diseases/immunology , Humans , Intracellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/immunology , Molecular Sequence Data , RNA, Messenger/immunology , Sequence Alignment , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...