Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nature ; 618(7967): 1072-1077, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37196676

ABSTRACT

Plasma membrane rupture (PMR) in dying cells undergoing pyroptosis or apoptosis requires the cell-surface protein NINJ11. PMR releases pro-inflammatory cytoplasmic molecules, collectively called damage-associated molecular patterns (DAMPs), that activate immune cells. Therefore, inhibiting NINJ1 and PMR may limit the inflammation that is associated with excessive cell death. Here we describe an anti-NINJ1 monoclonal antibody that specifically targets mouse NINJ1 and blocks oligomerization of NINJ1, preventing PMR. Electron microscopy studies showed that this antibody prevents NINJ1 from forming oligomeric filaments. In mice, inhibition of NINJ1 or Ninj1 deficiency ameliorated hepatocellular PMR induced with TNF plus D-galactosamine, concanavalin A, Jo2 anti-Fas agonist antibody or ischaemia-reperfusion injury. Accordingly, serum levels of lactate dehydrogenase, the liver enzymes alanine aminotransaminase and aspartate aminotransferase, and the DAMPs interleukin 18 and HMGB1 were reduced. Moreover, in the liver ischaemia-reperfusion injury model, there was an attendant reduction in neutrophil infiltration. These data indicate that NINJ1 mediates PMR and inflammation in diseases driven by aberrant hepatocellular death.


Subject(s)
Antibodies, Monoclonal , Cell Membrane , Inflammation , Liver , Nerve Growth Factors , Reperfusion Injury , Animals , Mice , Alanine Transaminase , Alarmins , Antibodies, Monoclonal/immunology , Aspartate Aminotransferases , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/immunology , Cell Adhesion Molecules, Neuronal/ultrastructure , Cell Death , Cell Membrane/pathology , Cell Membrane/ultrastructure , Concanavalin A , Galactosamine , Hepatocytes/pathology , Hepatocytes/ultrastructure , Inflammation/pathology , Lactate Dehydrogenases , Liver/pathology , Microscopy, Electron , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/deficiency , Nerve Growth Factors/immunology , Nerve Growth Factors/ultrastructure , Neutrophil Infiltration , Reperfusion Injury/pathology
2.
Immunity ; 49(3): 560-575.e6, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30170812

ABSTRACT

Signaling by Toll-like receptors (TLRs) on intestinal epithelial cells (IECs) is critical for intestinal homeostasis. To visualize epithelial expression of individual TLRs in vivo, we generated five strains of reporter mice. These mice revealed that TLR expression varied dramatically along the length of the intestine. Indeed, small intestine (SI) IECs expressed low levels of multiple TLRs that were highly expressed by colonic IECs. TLR5 expression was restricted to Paneth cells in the SI epithelium. Intestinal organoid experiments revealed that TLR signaling in Paneth cells or colonic IECs induced a core set of host defense genes, but this set did not include antimicrobial peptides, which instead were induced indirectly by inflammatory cytokines. This comprehensive blueprint of TLR expression and function in IECs reveals unexpected diversity in the responsiveness of IECs to microbial stimuli, and together with the associated reporter strains, provides a resource for further study of innate immunity.


Subject(s)
Colitis/immunology , Colon/pathology , Intestinal Mucosa/physiology , Intestine, Small/pathology , Paneth Cells/physiology , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , Colitis/chemically induced , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Homeostasis , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Receptor Cross-Talk , Signal Transduction , Toll-Like Receptor 5/metabolism
3.
Nature ; 591(7848): 131-136, 2021 03.
Article in English | MEDLINE | ID: mdl-33472215

ABSTRACT

Plasma membrane rupture (PMR) is the final cataclysmic event in lytic cell death. PMR releases intracellular molecules known as damage-associated molecular patterns (DAMPs) that propagate the inflammatory response1-3. The underlying mechanism of PMR, however, is unknown. Here we show that the cell-surface NINJ1 protein4-8, which contains two transmembrane regions, has an essential role in the induction of PMR. A forward-genetic screen of randomly mutagenized mice linked NINJ1 to PMR. Ninj1-/- macrophages exhibited impaired PMR in response to diverse inducers of pyroptotic, necrotic and apoptotic cell death, and were unable to release numerous intracellular proteins including HMGB1 (a known DAMP) and LDH (a standard measure of PMR). Ninj1-/- macrophages died, but with a distinctive and persistent ballooned morphology, attributable to defective disintegration of bubble-like herniations. Ninj1-/- mice were more susceptible than wild-type mice to infection with Citrobacter rodentium, which suggests a role for PMR in anti-bacterial host defence. Mechanistically, NINJ1 used an evolutionarily conserved extracellular domain for oligomerization and subsequent PMR. The discovery of NINJ1 as a mediator of PMR overturns the long-held idea that cell death-related PMR is a passive event.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Death , Cell Membrane/metabolism , Nerve Growth Factors/metabolism , Animals , Apoptosis , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Cell Death/genetics , Female , Humans , Macrophages , Male , Mice , Mutation , Necrosis , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Protein Multimerization , Pyroptosis/genetics
4.
Semin Immunol ; 70: 101841, 2023 11.
Article in English | MEDLINE | ID: mdl-37703611

ABSTRACT

Cells undergo an inflammatory programmed lytic cell death called 'pyroptosis' (with the Greek roots 'fiery'), often featuring morphological hallmarks such as large ballooning protrusions and subsequent bursting. Originally described as a caspase-1-dependent cell death in response to bacterial infection, pyroptosis has since been re-defined in 2018 as a cell death dependent on plasma membrane pores by a gasdermin (GSDM) family member [1,2]. GSDMs form pores in the plasma membrane as well as organelle membranes, thereby initiating membrane destruction and the rapid and lytic demise of a cell. The gasdermin family plays a profound role in the execution of pyroptosis in the context of infection, inflammation, tumor pathogenesis, and anti-tumor therapy. More recently, cell-death-independent functions for some of the GSDMs have been proposed. Therefore, a comprehensive understanding of gasdermin gene regulation, including mechanisms in both homeostatic conditions and during inflammation, is essential. In this review, we will summarize the role of gasdermins in pyroptosis and focus our discussion on the transcriptional and epigenetic mechanisms controlling the expression of GSDMs.


Subject(s)
Gasdermins , Neoplasm Proteins , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Apoptosis , Inflammation/metabolism , Epigenesis, Genetic , Inflammasomes/metabolism
5.
Immunity ; 47(5): 913-927.e6, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150239

ABSTRACT

Although apoptotic cells (ACs) contain nucleic acids that can be recognized by Toll-like receptors (TLRs), engulfment of ACs does not initiate inflammation in healthy organisms. Here we identified macrophage populations that continually engulf ACs in distinct tissues and found that these macrophages share characteristics compatible with immunologically silent clearance of ACs; such characteristics include high expression of AC recognition receptors, low expression of TLR9, and reduced TLR responsiveness to nucleic acids. Removal of the macrophages from tissues resulted in loss of many of these characteristics and the ability to generate inflammatory responses to AC-derived nucleic acids, suggesting that cues from the tissue microenvironment program macrophages for silent AC clearance. The transcription factors KLF2 and KLF4 control the expression of many genes within this AC clearance program. The coordinated expression of AC receptors with genes that limit responses to nucleic acids might ensure maintenance of homeostasis and thus represent a central feature of tissue macrophages.


Subject(s)
Apoptosis , Macrophages/immunology , Animals , Female , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/physiology , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 7/physiology , Toll-Like Receptor 9/physiology
6.
PLoS Pathog ; 17(10): e1009967, 2021 10.
Article in English | MEDLINE | ID: mdl-34648590

ABSTRACT

Cell death plays a critical role in inflammatory responses. During pyroptosis, inflammatory caspases cleave Gasdermin D (GSDMD) to release an N-terminal fragment that generates plasma membrane pores that mediate cell lysis and IL-1 cytokine release. Terminal cell lysis and IL-1ß release following caspase activation can be uncoupled in certain cell types or in response to particular stimuli, a state termed hyperactivation. However, the factors and mechanisms that regulate terminal cell lysis downstream of GSDMD cleavage remain poorly understood. In the course of studies to define regulation of pyroptosis during Yersinia infection, we identified a line of Card19-deficient mice (Card19lxcn) whose macrophages were protected from cell lysis and showed reduced apoptosis and pyroptosis, yet had wild-type levels of caspase activation, IL-1 secretion, and GSDMD cleavage. Unexpectedly, CARD19, a mitochondrial CARD-containing protein, was not directly responsible for this, as an independently-generated CRISPR/Cas9 Card19 knockout mouse line (Card19Null) showed no defect in macrophage cell lysis. Notably, Card19 is located on chromosome 13, immediately adjacent to Ninj1, which was recently found to regulate cell lysis downstream of GSDMD activation. RNA-seq and western blotting revealed that Card19lxcn BMDMs have significantly reduced NINJ1 expression, and reconstitution of Ninj1 in Card19lxcn immortalized BMDMs restored their ability to undergo cell lysis in response to caspase-dependent cell death stimuli. Card19lxcn mice exhibited increased susceptibility to Yersinia infection, whereas independently-generated Card19Null mice did not, demonstrating that cell lysis itself plays a key role in protection against bacterial infection, and that the increased infection susceptibility of Card19lxcn mice is attributable to loss of NINJ1. Our findings identify genetic targeting of Card19 being responsible for off-target effects on the adjacent gene Ninj1, disrupting the ability of macrophages to undergo plasma membrane rupture downstream of gasdermin cleavage and impacting host survival and bacterial control during Yersinia infection.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Macrophages/metabolism , Nerve Growth Factors/metabolism , Yersinia Infections/pathology , Animals , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Pyroptosis/physiology , Yersinia Infections/metabolism
7.
PLoS Biol ; 17(9): e3000354, 2019 09.
Article in English | MEDLINE | ID: mdl-31525186

ABSTRACT

The nucleotide-binding-domain (NBD)-and leucine-rich repeat (LRR)-containing (NLR) family, pyrin-domain-containing 3 (NLRP3) inflammasome drives pathological inflammation in a suite of autoimmune, metabolic, malignant, and neurodegenerative diseases. Additionally, NLRP3 gain-of-function point mutations cause systemic periodic fever syndromes that are collectively known as cryopyrin-associated periodic syndrome (CAPS). There is significant interest in the discovery and development of diarylsulfonylurea Cytokine Release Inhibitory Drugs (CRIDs) such as MCC950/CRID3, a potent and selective inhibitor of the NLRP3 inflammasome pathway, for the treatment of CAPS and other diseases. However, drug discovery efforts have been constrained by the lack of insight into the molecular target and mechanism by which these CRIDs inhibit the NLRP3 inflammasome pathway. Here, we show that the NAIP, CIITA, HET-E, and TP1 (NACHT) domain of NLRP3 is the molecular target of diarylsulfonylurea inhibitors. Interestingly, we find photoaffinity labeling (PAL) of the NACHT domain requires an intact (d)ATP-binding pocket and is substantially reduced for most CAPS-associated NLRP3 mutants. In concordance with this finding, MCC950/CRID3 failed to inhibit NLRP3-driven inflammatory pathology in two mouse models of CAPS. Moreover, it abolished circulating levels of interleukin (IL)-1ß and IL-18 in lipopolysaccharide (LPS)-challenged wild-type mice but not in Nlrp3L351P knock-in mice and ex vivo-stimulated mutant macrophages. These results identify wild-type NLRP3 as the molecular target of MCC950/CRID3 and show that CAPS-related NLRP3 mutants escape efficient MCC950/CRID3 inhibition. Collectively, this work suggests that MCC950/CRID3-based therapies may effectively treat inflammation driven by wild-type NLRP3 but not CAPS-associated mutants.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/genetics , Furans/pharmacology , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Cytokines/antagonists & inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings , Humans , Indenes , Lipopolysaccharides , Macrophages/drug effects , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Protein Domains , Sulfones
9.
Nature ; 526(7575): 666-71, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26375259

ABSTRACT

Intracellular lipopolysaccharide from Gram-negative bacteria including Escherichia coli, Salmonella typhimurium, Shigella flexneri, and Burkholderia thailandensis activates mouse caspase-11, causing pyroptotic cell death, interleukin-1ß processing, and lethal septic shock. How caspase-11 executes these downstream signalling events is largely unknown. Here we show that gasdermin D is essential for caspase-11-dependent pyroptosis and interleukin-1ß maturation. A forward genetic screen with ethyl-N-nitrosourea-mutagenized mice links Gsdmd to the intracellular lipopolysaccharide response. Macrophages from Gsdmd(-/-) mice generated by gene targeting also exhibit defective pyroptosis and interleukin-1ß secretion induced by cytoplasmic lipopolysaccharide or Gram-negative bacteria. In addition, Gsdmd(-/-) mice are protected from a lethal dose of lipopolysaccharide. Mechanistically, caspase-11 cleaves gasdermin D, and the resulting amino-terminal fragment promotes both pyroptosis and NLRP3-dependent activation of caspase-1 in a cell-intrinsic manner. Our data identify gasdermin D as a critical target of caspase-11 and a key mediator of the host response against Gram-negative bacteria.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Inflammasomes/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Caspases, Initiator , Cell Line , Female , Gram-Negative Bacteria/immunology , Humans , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mutation/genetics , Necrosis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins , Protein Processing, Post-Translational/drug effects , Sepsis/microbiology , Signal Transduction/genetics , Survival Analysis
11.
Immunol Rev ; 265(1): 75-84, 2015 May.
Article in English | MEDLINE | ID: mdl-25879285

ABSTRACT

As a front line of defense against pathogenic microbes, our body employs a primitive, yet highly sophisticated and potent innate immune response pathway collectively referred to as the inflammasome. Innate immune cells, epithelial cells, and many other cell types are capable of detecting infection or tissue injury and mounting a coordinated molecular defense. For example, Gram-negative bacteria are specifically detected via a surveillance mechanism that involves activation of extracellular receptors such as Toll-like receptors (TLRs) followed by intracellular recognition and activation of pathways such as caspase-11 (caspase-4/5 in humans). Importantly, lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is a strong trigger of these pathways. Extracellular LPS primarily stimulates TLR4, which can serve as a priming signal for expression of inflammasome components. Intracellular LPS can then trigger caspase-11-dependent inflammasome activation in the cytoplasm. Here, we briefly review the burgeoning caspase-11-dependent non-canonical inflammasome field, focusing mainly on the innate sensing of LPS.


Subject(s)
Bacterial Infections/metabolism , Caspases, Initiator/metabolism , Caspases/metabolism , Multiprotein Complexes/metabolism , Animals , Bacterial Infections/immunology , Caspases/immunology , Caspases, Initiator/immunology , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Mice , Multiprotein Complexes/immunology , Signal Transduction , Toll-Like Receptor 4/metabolism
12.
PLoS Pathog ; 12(12): e1006035, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911947

ABSTRACT

Type III secretion systems (T3SS) are central virulence factors for many pathogenic Gram-negative bacteria, and secreted T3SS effectors can block key aspects of host cell signaling. To counter this, innate immune responses can also sense some T3SS components to initiate anti-bacterial mechanisms. The Yersinia pestis T3SS is particularly effective and sophisticated in manipulating the production of pro-inflammatory cytokines IL-1ß and IL-18, which are typically processed into their mature forms by active caspase-1 following inflammasome formation. Some effectors, like Y. pestis YopM, may block inflammasome activation. Here we show that YopM prevents Y. pestis induced activation of the Pyrin inflammasome induced by the RhoA-inhibiting effector YopE, which is a GTPase activating protein. YopM blocks YopE-induced Pyrin-mediated caspase-1 dependent IL-1ß/IL-18 production and cell death. We also detected YopM in a complex with Pyrin and kinases RSK1 and PKN1, putative negative regulators of Pyrin. In contrast to wild-type mice, Pyrin deficient mice were also highly susceptible to an attenuated Y. pestis strain lacking YopM, emphasizing the importance of inhibition of Pyrin in vivo. A complex interplay between the Y. pestis T3SS and IL-1ß/IL-18 production is evident, involving at least four inflammasome pathways. The secreted effector YopJ triggers caspase-8- dependent IL-1ß activation, even when YopM is present. Additionally, the presence of the T3SS needle/translocon activates NLRP3 and NLRC4-dependent IL-1ß generation, which is blocked by YopK, but not by YopM. Taken together, the data suggest YopM specificity for obstructing the Pyrin pathway, as the effector does not appear to block Y. pestis-induced NLRP3, NLRC4 or caspase-8 dependent caspase-1 processing. Thus, we identify Y. pestis YopM as a microbial inhibitor of the Pyrin inflammasome. The fact that so many of the Y. pestis T3SS components are participating in regulation of IL-1ß/IL-18 release suggests that these effects are essential for maximal control of innate immunity during plague.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , Animals , Disease Models, Animal , Mice , Mice, Knockout , Yersinia pestis/immunology
13.
Immunity ; 28(3): 381-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18342009

ABSTRACT

Muramyl dipeptide (MDP), a product of bacterial cell-wall peptidoglycan, activates innate immune cells by stimulating nucleotide-binding oligomerization domain containing 2 (NOD2) -dependent activation of the transcription factor NFkappaB and transcription of proinflammatory genes. A20 is a ubiquitin-modifying enzyme that restricts tumor necrosis factor (TNF) receptor and Toll-like receptor (TLR) -induced signals. We now show that MDP induces ubiquitylation of receptor- interacting protein 2 (RIP2) in primary macrophages. A20-deficient cells exhibit dramatically amplified responses to MDP, including increased RIP2 ubiquitylation, prolonged NFkappaB signaling, and increased production of proinflammatory cytokines. In addition, in vivo responses to MDP are exaggerated in A20-deficient mice and in chimeric mice bearing A20-deficient hematopoietic cells. These exaggerated responses occur independently of the TLR adaptors MyD88 and TRIF as well as TNF signals. These findings indicate that A20 directly restricts NOD2 induced signals in vitro and in vivo, and provide new insights into how these signals are physiologically restricted.


Subject(s)
Cysteine Endopeptidases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Macrophages/metabolism , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin/metabolism
14.
Nature ; 457(7231): 906-9, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19060883

ABSTRACT

Proteins that directly regulate tumour necrosis factor receptor (TNFR) signalling have critical roles in regulating cellular activation and survival. ABIN-1 (A20 binding and inhibitor of NF-kappaB) is a novel protein that is thought to inhibit NF-kappaB signalling. Here we show that mice deficient for ABIN-1 die during embryogenesis with fetal liver apoptosis, anaemia and hypoplasia. ABIN-1 deficient cells are hypersensitive to tumour necrosis factor (TNF)-induced programmed cell death, and TNF deficiency rescues ABIN-1 deficient embryos. ABIN-1 inhibits caspase 8 recruitment to FADD (Fas-associated death domain-containing protein) in TNF-induced signalling complexes, preventing caspase 8 cleavage and programmed cell death. Moreover, ABIN-1 directly binds polyubiquitin chains and this ubiquitin sensing activity is required for ABIN-1's anti-apoptotic activity. These studies provide insights into how ubiquitination and ubiquitin sensing proteins regulate cellular and organismal survival.


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/metabolism , Embryonic Development/physiology , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , DNA-Binding Proteins/chemistry , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Alignment , Tumor Necrosis Factor-alpha/metabolism
15.
Nature ; 456(7222): 658-62, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-18820679

ABSTRACT

Mammalian Toll-like receptors (TLRs) 3, 7, 8 and 9 initiate immune responses to infection by recognizing microbial nucleic acids; however, these responses come at the cost of potential autoimmunity owing to inappropriate recognition of self nucleic acids. The localization of TLR9 and TLR7 to intracellular compartments seems to have a role in facilitating responses to viral nucleic acids while maintaining tolerance to self nucleic acids, yet the cell biology regulating the transport and localization of these receptors remains poorly understood. Here we define the route by which TLR9 and TLR7 exit the endoplasmic reticulum and travel to endolysosomes in mouse macrophages and dendritic cells. The ectodomains of TLR9 and TLR7 are cleaved in the endolysosome, such that no full-length protein is detectable in the compartment where ligand is recognized. Notably, although both the full-length and cleaved forms of TLR9 are capable of binding ligand, only the processed form recruits MyD88 on activation, indicating that this truncated receptor, rather than the full-length form, is functional. Furthermore, conditions that prevent receptor proteolysis, including forced TLR9 surface localization, render the receptor non-functional. We propose that ectodomain cleavage represents a strategy to restrict receptor activation to endolysosomal compartments and prevent TLRs from responding to self nucleic acids.


Subject(s)
Protein Processing, Post-Translational , Toll-Like Receptor 9/chemistry , Toll-Like Receptor 9/metabolism , Animals , Cell Line , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endoplasmic Reticulum/metabolism , Female , Golgi Apparatus/metabolism , Ligands , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Phagosomes/metabolism , Protein Structure, Tertiary , Protein Transport , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 7/metabolism
16.
Sci Signal ; 12(582)2019 05 21.
Article in English | MEDLINE | ID: mdl-31113851

ABSTRACT

Gasdermin-D (GSDMD) is cleaved by caspase-1, caspase-4, and caspase-11 in response to canonical and noncanonical inflammasome activation. Upon cleavage, GSDMD oligomerizes and forms plasma membrane pores, resulting in interleukin-1ß (IL-1ß) secretion, pyroptotic cell death, and inflammatory pathologies, including periodic fever syndromes and septic shock-a plague on modern medicine. Here, we showed that IRF2, a member of the interferon regulatory factor (IRF) family of transcription factors, was essential for the transcriptional activation of GSDMD. A forward genetic screen with N-ethyl-N-nitrosourea (ENU)-mutagenized mice linked IRF2 to inflammasome signaling. GSDMD expression was substantially attenuated in IRF2-deficient macrophages, endothelial cells, and multiple tissues, which corresponded with reduced IL-1ß secretion and inhibited pyroptosis. Mechanistically, IRF2 bound to a previously uncharacterized but unique site within the GSDMD promoter to directly drive GSDMD transcription for the execution of pyroptosis. Disruption of this single IRF2-binding site abolished signaling by both the canonical and noncanonical inflammasomes. Together, our data illuminate a key transcriptional mechanism for expression of the gene encoding GSDMD, a critical mediator of inflammatory pathologies.


Subject(s)
Interferon Regulatory Factor-2/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphate-Binding Proteins/genetics , Pyroptosis/genetics , Transcription, Genetic/genetics , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Interferon Regulatory Factor-2/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins/metabolism , Signal Transduction/genetics , Transcriptional Activation/genetics
17.
Sci Rep ; 8(1): 3788, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491424

ABSTRACT

The NLRC4 inflammasome recognizes bacterial flagellin and components of the type III secretion apparatus. NLRC4 stimulation leads to caspase-1 activation followed by a rapid lytic cell death known as pyroptosis. NLRC4 is linked to pathogen-free auto-inflammatory diseases, suggesting a role for NLRC4 in sterile inflammation. Here, we show that NLRC4 activates an alternative cell death program morphologically similar to apoptosis in caspase-1-deficient BMDMs. By performing an unbiased genome-wide CRISPR/Cas9 screen with subsequent validation studies in gene-targeted mice, we highlight a critical role for caspase-8 and ASC adaptor in an alternative apoptotic pathway downstream of NLRC4. Furthermore, caspase-1 catalytically dead knock-in (Casp1 C284A KI) BMDMs genetically segregate pyroptosis and apoptosis, and confirm that caspase-1 does not functionally compete with ASC for NLRC4 interactions. We show that NLRC4/caspase-8-mediated apoptotic cells eventually undergo plasma cell membrane damage in vitro, suggesting that this pathway can lead to secondary necrosis. Unexpectedly, we found that DFNA5/GSDME, a member of the pore-forming gasdermin family, is dispensable for the secondary necrosis that follows NLRC4-mediated apoptosis in macrophages. Together, our data confirm the existence of an alternative caspase-8 activation pathway diverging from the NLRC4 inflammasome in primary macrophages.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , CARD Signaling Adaptor Proteins/physiology , Calcium-Binding Proteins/metabolism , Caspase 1/physiology , Caspase 8/physiology , Inflammasomes/metabolism , Macrophages/pathology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , CRISPR-Cas Systems , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Genome , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
J Exp Med ; 215(9): 2279-2288, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30135078

ABSTRACT

Intracellular LPS sensing by caspase-4/5/11 triggers proteolytic activation of pore-forming gasdermin D (GSDMD), leading to pyroptotic cell death in Gram-negative bacteria-infected cells. Involvement of caspase-4/5/11 and GSDMD in inflammatory responses, such as lethal sepsis, makes them highly desirable drug targets. Using knock-in (KI) mouse strains, we herein provide genetic evidence to show that caspase-11 auto-cleavage at the inter-subunit linker is essential for optimal catalytic activity and subsequent proteolytic cleavage of GSDMD. Macrophages from caspase-11-processing dead KI mice (Casp11Prc D285A/D285A ) exhibit defective caspase-11 auto-processing and phenocopy Casp11-/- and caspase-11 enzymatically dead KI (Casp11Enz C254A/C254A ) macrophages in attenuating responses to cytoplasmic LPS or Gram-negative bacteria infection. GsdmdD276A/D276A KI macrophages also fail to cleave GSDMD and are hypo-responsive to inflammasome stimuli, confirming that the GSDMD Asp276 residue is a nonredundant and indispensable site for proteolytic activation of GSDMD. Our data highlight the role of caspase-11 self-cleavage as a critical regulatory step for GSDMD processing and response against Gram-negative bacteria.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Caspases/immunology , Inflammasomes/immunology , Macrophages/immunology , Proteolysis , Animals , Apoptosis Regulatory Proteins/genetics , Caspases/genetics , Caspases, Initiator , Gene Knock-In Techniques , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/pathology , Inflammasomes/genetics , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Phosphate-Binding Proteins
19.
Science ; 362(6418): 1064-1069, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30361383

ABSTRACT

Limited proteolysis of gasdermin D (GSDMD) generates an N-terminal pore-forming fragment that controls pyroptosis in macrophages. GSDMD is processed via inflammasome-activated caspase-1 or -11. It is currently unknown whether macrophage GSDMD can be processed by other mechanisms. Here, we describe an additional pathway controlling GSDMD processing. The inhibition of TAK1 or IκB kinase (IKK) by the Yersinia effector protein YopJ elicits RIPK1- and caspase-8-dependent cleavage of GSDMD, which subsequently results in cell death. GSDMD processing also contributes to the NLRP3 inflammasome-dependent release of interleukin-1ß (IL-1ß). Thus, caspase-8 acts as a regulator of GSDMD-driven cell death. Furthermore, this study establishes the importance of TAK1 and IKK activity in the control of GSDMD cleavage and cytotoxicity.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 8/metabolism , Host-Pathogen Interactions , I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinases/metabolism , Plague/immunology , Animals , Bacterial Proteins/metabolism , Caspase 8/genetics , Cell Death , Humans , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Plague/enzymology , Plague/pathology , Proteolysis
20.
Trends Cell Biol ; 24(6): 360-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24439965

ABSTRACT

Over the past decade we have learned much about nucleic acid recognition by the innate immune system and in particular by Toll-like receptors (TLRs). These receptors localize to endosomal compartments where they are poised to recognize microbial nucleic acids. Multiple regulatory mechanisms function to limit responses to self DNA or RNA, and breakdowns in these mechanisms can contribute to autoimmune or inflammatory disorders. In this review we discuss our current understanding of the cell biology of TLRs involved in nucleic acid recognition and how localization and trafficking of these receptors regulates their function.


Subject(s)
Endosomes/metabolism , Toll-Like Receptors/metabolism , Animals , Endoplasmic Reticulum/metabolism , Humans , Protein Transport , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL