Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
EMBO J ; 43(6): 904-930, 2024 Mar.
Article En | MEDLINE | ID: mdl-38337057

Mitochondrial outer membrane permeabilisation (MOMP) is often essential for apoptosis, by enabling cytochrome c release that leads to caspase activation and rapid cell death. Recently, MOMP has been shown to be inherently pro-inflammatory with emerging cellular roles, including its ability to elicit anti-tumour immunity. Nonetheless, how MOMP triggers inflammation and how the cell regulates this remains poorly defined. We find that upon MOMP, many proteins localised either to inner or outer mitochondrial membranes are ubiquitylated in a promiscuous manner. This extensive ubiquitylation serves to recruit the essential adaptor molecule NEMO, leading to the activation of pro-inflammatory NF-κB signalling. We show that disruption of mitochondrial outer membrane integrity through different means leads to the engagement of a similar pro-inflammatory signalling platform. Therefore, mitochondrial integrity directly controls inflammation, such that permeabilised mitochondria initiate NF-κB signalling.


NF-kappa B , Ubiquitin , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Ubiquitin/metabolism , Mitochondrial Membranes/metabolism , Mitochondria/metabolism , Apoptosis/physiology , Inflammation/metabolism
2.
Biochem Soc Trans ; 51(1): 457-472, 2023 02 27.
Article En | MEDLINE | ID: mdl-36815695

Cytosolic DNA is recognized by the innate immune system as a potential threat. During apoptotic cell death, mitochondrial DNA (mtDNA) release activates the DNA sensor cyclic GMP-AMP synthase (cGAS) to promote a pro-inflammatory type I interferon response. Inflammation following mtDNA release during apoptotic cell death can be exploited to engage anti-tumor immunity and represents a potential avenue for cancer therapy. Additionally, various studies have described leakage of mtDNA, independent of cell death, with different underlying cues such as pathogenic infections, changes in mtDNA packaging, mtDNA stress or reduced mitochondrial clearance. The interferon response in these scenarios can be beneficial but also potentially disadvantageous, as suggested by a variety of disease phenotypes. In this review, we discuss mechanisms underlying mtDNA release governed by cell death pathways and summarize release mechanisms independent of cell death. We further highlight the similarities and differences in mtDNA release pathways, outlining gaps in our knowledge and questions for further research. Together, a deeper understanding of how and when mtDNA is released may enable the development of drugs to specifically target or inhibit mtDNA release in different disease settings.


DNA, Mitochondrial , Mitochondria , Humans , DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Inflammation/metabolism , Apoptosis , Interferons/metabolism
3.
Dev Cell ; 57(10): 1211-1225.e6, 2022 05 23.
Article En | MEDLINE | ID: mdl-35447090

Mitochondrial dysfunction is interconnected with cancer. Nevertheless, how defective mitochondria promote cancer is poorly understood. We find that mitochondrial dysfunction promotes DNA damage under conditions of increased apoptotic priming. Underlying this process, we reveal a key role for mitochondrial dynamics in the regulation of DNA damage and genome instability. The ability of mitochondrial dynamics to regulate oncogenic DNA damage centers upon the control of minority mitochondrial outer membrane permeabilization (MOMP), a process that enables non-lethal caspase activation leading to DNA damage. Mitochondrial fusion suppresses minority MOMP and its associated DNA damage by enabling homogeneous mitochondrial expression of anti-apoptotic BCL-2 proteins. Finally, we find that mitochondrial dysfunction inhibits pro-apoptotic BAX retrotranslocation, causing BAX mitochondrial localization and thereby promoting minority MOMP. Unexpectedly, these data reveal oncogenic effects of mitochondrial dysfunction that are mediated via mitochondrial dynamics and caspase-dependent DNA damage.


Caspases , Mitochondrial Dynamics , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Caspases/metabolism , DNA Damage , Genomic Instability , Humans , bcl-2-Associated X Protein/metabolism
4.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article En | MEDLINE | ID: mdl-34260403

Injection of effector proteins to block host innate immune signaling is a common strategy used by many pathogenic organisms to establish an infection. For example, pathogenic Yersinia species inject the acetyltransferase YopJ into target cells to inhibit NF-κB and MAPK signaling. To counteract this, detection of YopJ activity in myeloid cells promotes the assembly of a RIPK1-caspase-8 death-inducing platform that confers antibacterial defense. While recent studies revealed that caspase-8 cleaves the pore-forming protein gasdermin D to trigger pyroptosis in macrophages, whether RIPK1 activates additional substrates downstream of caspase-8 to promote host defense is unclear. Here, we report that the related gasdermin family member gasdermin E (GSDME) is activated upon detection of YopJ activity in a RIPK1 kinase-dependent manner. Specifically, GSDME promotes neutrophil pyroptosis and IL-1ß release, which is critical for anti-Yersinia defense. During in vivo infection, IL-1ß neutralization increases bacterial burden in wild-type but not Gsdme-deficient mice. Thus, our study establishes GSDME as an important mediator that counteracts pathogen blockade of innate immune signaling.


Immunity, Innate , Macrophages/metabolism , Neoplasm Proteins/metabolism , Neutrophils/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Yersinia pseudotuberculosis/physiology , 3T3 Cells , Animals , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Pyroptosis , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/microbiology
5.
Nat Commun ; 11(1): 3276, 2020 06 24.
Article En | MEDLINE | ID: mdl-32581219

The human non-canonical inflammasome controls caspase-4 activation and gasdermin-D-dependent pyroptosis in response to cytosolic bacterial lipopolysaccharide (LPS). Since LPS binds and oligomerizes caspase-4, the pathway is thought to proceed without dedicated LPS sensors or an activation platform. Here we report that interferon-induced guanylate-binding proteins (GBPs) are required for non-canonical inflammasome activation by cytosolic Salmonella or upon cytosolic delivery of LPS. GBP1 associates with the surface of cytosolic Salmonella seconds after bacterial escape from their vacuole, initiating the recruitment of GBP2-4 to assemble a GBP coat. The GBP coat then promotes the recruitment of caspase-4 to the bacterial surface and caspase activation, in absence of bacteriolysis. Mechanistically, GBP1 binds LPS with high affinity through electrostatic interactions. Our findings indicate that in human epithelial cells GBP1 acts as a cytosolic LPS sensor and assembles a platform for caspase-4 recruitment and activation at LPS-containing membranes as the first step of non-canonical inflammasome signaling.


Caspases, Initiator/metabolism , Cytosol/microbiology , GTP-Binding Proteins/metabolism , Lipopolysaccharides/metabolism , Salmonella/metabolism , Cell Line , Enzyme Activation , Epithelial Cells/metabolism , HeLa Cells , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Protein Binding , Pyroptosis , Static Electricity
6.
Life Sci Alliance ; 3(6)2020 06.
Article En | MEDLINE | ID: mdl-32345661

Caspase-1 drives a lytic inflammatory cell death named pyroptosis by cleaving the pore-forming cell death executor gasdermin-D (GSDMD). Gsdmd deficiency, however, only delays cell lysis, indicating that caspase-1 controls alternative cell death pathways. Here, we show that in the absence of GSDMD, caspase-1 activates apoptotic initiator and executioner caspases and triggers a rapid progression into secondary necrosis. GSDMD-independent cell death required direct caspase-1-driven truncation of Bid and generation of caspase-3 p19/p12 by either caspase-8 or caspase-9. tBid-induced mitochondrial outer membrane permeabilization was also required to drive SMAC release and relieve inhibitor of apoptosis protein inhibition of caspase-3, thereby allowing caspase-3 auto-processing to the fully active p17/p12 form. Our data reveal that cell lysis in inflammasome-activated Gsdmd-deficient cells is caused by a synergistic effect of rapid caspase-1-driven activation of initiator caspases-8/-9 and Bid cleavage, resulting in an unusually fast activation of caspase-3 and immediate transition into secondary necrosis. This pathway might be advantageous for the host in counteracting pathogen-induced inhibition of GSDMD but also has implications for the use of GSDMD inhibitors in immune therapies for caspase-1-dependent inflammatory disease.


Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/deficiency , Caspase 1/deficiency , Intracellular Signaling Peptides and Proteins/deficiency , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Phosphate-Binding Proteins/deficiency , Signal Transduction/genetics , Animals , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Caspase 1/genetics , Cells, Cultured , Gene Editing , Gene Knockout Techniques , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Necrosis/genetics , Necrosis/metabolism , Phosphate-Binding Proteins/genetics , Pyroptosis/genetics , Transfection
7.
EMBO J ; 38(10)2019 05 15.
Article En | MEDLINE | ID: mdl-30902848

Pyroptosis is a form of lytic inflammatory cell death driven by inflammatory caspase-1, caspase-4, caspase-5 and caspase-11. These caspases cleave and activate the pore-forming protein gasdermin D (GSDMD) to induce membrane damage. By contrast, apoptosis is driven by apoptotic caspase-8 or caspase-9 and has traditionally been classified as an immunologically silent form of cell death. Emerging evidence suggests that therapeutics designed for cancer chemotherapy or inflammatory disorders such as SMAC mimetics, TAK1 inhibitors and BH3 mimetics promote caspase-8 or caspase-9-dependent inflammatory cell death and NLRP3 inflammasome activation. However, the mechanism by which caspase-8 or caspase-9 triggers cell lysis and NLRP3 activation is still undefined. Here, we demonstrate that during extrinsic apoptosis, caspase-1 and caspase-8 cleave GSDMD to promote lytic cell death. By engineering a novel Gsdmd D88A knock-in mouse, we further demonstrate that this proinflammatory function of caspase-8 is counteracted by caspase-3-dependent cleavage and inactivation of GSDMD at aspartate 88, and is essential to suppress GSDMD-dependent cell lysis during caspase-8-dependent apoptosis. Lastly, we provide evidence that channel-forming glycoprotein pannexin-1, but not GSDMD or GSDME promotes NLRP3 inflammasome activation during caspase-8 or caspase-9-dependent apoptosis.


Apoptosis/physiology , Connexins/physiology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Tissue Proteins/physiology , 3T3 Cells , Animals , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cells, Cultured , Embryo, Mammalian , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/metabolism , Phosphate-Binding Proteins/metabolism , Protein Binding , Protein Multimerization , Receptors, Estrogen/metabolism , Signal Transduction/physiology
8.
Science ; 362(6417): 956-960, 2018 11 23.
Article En | MEDLINE | ID: mdl-30467171

Pyroptosis is a lytic form of cell death that is induced by inflammatory caspases upon activation of the canonical or noncanonical inflammasome pathways. These caspases cleave gasdermin D (GSDMD) to generate an N-terminal GSDMD fragment, which executes pyroptosis by forming membrane pores. We found that calcium influx through GSDMD pores serves as a signal for cells to initiate membrane repair by recruiting the endosomal sorting complexes required for transport (ESCRT) machinery to damaged membrane areas, such as the plasma membrane. Inhibition of the ESCRT-III machinery strongly enhances pyroptosis and interleukin-1ß release in both human and murine cells after canonical or noncanonical inflammasome activation. These results not only attribute an anti-inflammatory role to membrane repair by the ESCRT-III system but also provide insight into general cellular survival mechanisms during pyroptosis.


Calcium Signaling , Calcium/metabolism , Caspases/metabolism , Cell Membrane/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Neoplasm Proteins/metabolism , Pyroptosis , Aniline Compounds/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Caspases/genetics , Caspases, Initiator , Cell Survival , Cells, Cultured , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Macrophages , Mice , Neoplasm Proteins/genetics , Phosphate-Binding Proteins , Xanthenes/metabolism
9.
Article En | MEDLINE | ID: mdl-30234022

Francisella tularensis is the causative agent of the life-threatening disease tularemia. However, the molecular tools to study Francisella are limited. Especially, expression plasmids are sparse and difficult to use, as they are unstable and prone to spontaneous loss. Most Francisella expression plasmids lack inducible promoters making it difficult to control gene expression levels. In addition, available expression plasmids are mainly designed for F. tularensis, however, genetic differences including restriction-modification systems impede the use of these plasmids in F. novicida, which is often used as a model organism to study Francisella pathogenesis. Here we report construction and characterization of two mobilizable plasmids (pFNMB1 and pFNMB2) designed for regulated gene expression in F. novicida. pFNMB plasmids contain a tetracycline inducible promoter to control gene expression levels and oriT for RP4 mediated mobilization. We show that both plasmids are stably maintained in bacteria for more than 40 generations over 4 days of culturing in the absence of selection against plasmid loss. Expression levels are dependent on anhydrotetracycline concentration and homogeneous in a bacterial population. pFNMB1 and pFNMB2 plasmids differ in the sequence between promoter and translation start site and thus allow to reach different maximum levels of protein expression. We used pFNMB1 and pFNMB2 for complementation of Francisella Pathogenicity Island mutants ΔiglF, ΔiglI, and ΔiglC in-vitro and pFNMB1 to complement ΔiglI mutant in bone marrow derived macrophages.


Francisella/genetics , Gene Expression Regulation, Bacterial , Genetic Engineering/methods , Genetic Vectors , Genetics, Microbial/methods , Plasmids , Gene Deletion , Genes, Bacterial , Genetic Complementation Test , Genomic Instability , Genomic Islands , Interspersed Repetitive Sequences , Promoter Regions, Genetic , Transcriptional Activation/drug effects
10.
Eur J Immunol ; 48(4): 584-592, 2018 04.
Article En | MEDLINE | ID: mdl-29274245

The pro-inflammatory cytokine IL-1ß is well known for its role in host defense and the initiation of potent inflammatory responses. It is processed from its inactive pro-form by the inflammatory caspase-1 into its mature bioactive form, which is then released from the cell via an unconventional secretion mechanism. Recently, gasdermin-D has been identified as a new target of caspase-1. After proteolytical cleavage of gasdermin-D, the N-terminal fragment induces pyroptosis, a lytic cell death, by forming large permeability pores in the plasma membrane. Here we show using the murine system that gasdermin-D is required for IL-1ß secretion by macrophages, dendritic cells and partially in neutrophils, and that secretion is a cell-lysis-independent event. Liposome transport assays in vitro further demonstrate that gasdermin-D pores are large enough to allow the direct release of IL-1ß. Moreover, IL-18 and other small soluble cytosolic proteins can also be released in a lysis-independent but gasdermin-D-dependent mode, suggesting that the gasdermin-D pores allow passive the release of cytosolic proteins in a size-dependent manner.


Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Interleukin-1beta/metabolism , Liposomes/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 1/genetics , Cells, Cultured , Dendritic Cells/metabolism , Intracellular Signaling Peptides and Proteins , Macrophages/metabolism , Mice , Mice, Knockout , Phosphate-Binding Proteins , Protein Transport/physiology , Pyroptosis/physiology
11.
Eur J Immunol ; 48(2): 230-238, 2018 02.
Article En | MEDLINE | ID: mdl-29148036

Pyrin, encoded by the MEFV gene, is an intracellular pattern recognition receptor that assembles inflammasome complexes in response to pathogen infections. Mutations in the MEFV gene have been linked to autoinflammatory diseases such as familial Mediterranean fever (FMF) or pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). Recent insights have now revealed how pyrin is activated during infection, providing a molecular basis for the understanding of such disease-causing mutations in pyrin. Interestingly, pyrin does not directly recognize molecular patterns (pathogen- or host-derived danger molecules), but rather responds to disturbances in cytoplasmic homeostasis caused by the infection. In the case of pyrin, these perturbations, recently defined as 'homeostasis-altering molecular processes' (HAMPs), are processes leading to the inactivation of the RhoA GTPase. This review attempts to combine early observation and findings with the most recent discoveries on how pyrin detects inactivation of RhoA to shed light on the function and mechanism of pyrin activation.


Familial Mediterranean Fever/immunology , Infections/immunology , Inflammasomes/metabolism , Pyrin/genetics , Sweet Syndrome/immunology , rhoA GTP-Binding Protein/metabolism , Animals , Familial Mediterranean Fever/genetics , Homeostasis , Humans , Infections/genetics , Pyroptosis , Sweet Syndrome/genetics
12.
Cell Rep ; 21(13): 3846-3859, 2017 12 26.
Article En | MEDLINE | ID: mdl-29281832

Inflammasomes activate the protease caspase-1, which cleaves interleukin-1ß and interleukin-18 to generate the mature cytokines and controls their secretion and a form of inflammatory cell death called pyroptosis. By generating mice expressing enzymatically inactive caspase-1C284A, we provide genetic evidence that caspase-1 protease activity is required for canonical IL-1 secretion, pyroptosis, and inflammasome-mediated immunity. In caspase-1-deficient cells, caspase-8 can be activated at the inflammasome. Using mice either lacking the pyroptosis effector gasdermin D (GSDMD) or expressing caspase-1C284A, we found that GSDMD-dependent pyroptosis prevented caspase-8 activation at the inflammasome. In the absence of GSDMD-dependent pyroptosis, the inflammasome engaged a delayed, alternative form of lytic cell death that was accompanied by the release of large amounts of mature IL-1 and contributed to host protection. Features of this cell death modality distinguished it from apoptosis, suggesting it may represent a distinct form of pro-inflammatory regulated necrosis.


Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , Pyroptosis , Animals , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Enzyme Activation/drug effects , Francisella/physiology , Immunity, Innate , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Phosphate-Binding Proteins , Pyroptosis/drug effects
13.
EMBO J ; 35(16): 1766-78, 2016 08 15.
Article En | MEDLINE | ID: mdl-27418190

Pyroptosis is a lytic type of cell death that is initiated by inflammatory caspases. These caspases are activated within multi-protein inflammasome complexes that assemble in response to pathogens and endogenous danger signals. Pyroptotic cell death has been proposed to proceed via the formation of a plasma membrane pore, but the underlying molecular mechanism has remained unclear. Recently, gasdermin D (GSDMD), a member of the ill-characterized gasdermin protein family, was identified as a caspase substrate and an essential mediator of pyroptosis. GSDMD is thus a candidate for pyroptotic pore formation. Here, we characterize GSDMD function in live cells and in vitro We show that the N-terminal fragment of caspase-1-cleaved GSDMD rapidly targets the membrane fraction of macrophages and that it induces the formation of a plasma membrane pore. In vitro, the N-terminal fragment of caspase-1-cleaved recombinant GSDMD tightly binds liposomes and forms large permeability pores. Visualization of liposome-inserted GSDMD at nanometer resolution by cryo-electron and atomic force microscopy shows circular pores with variable ring diameters around 20 nm. Overall, these data demonstrate that GSDMD is the direct and final executor of pyroptotic cell death.


Caspase 1/metabolism , Cell Membrane/metabolism , Neoplasm Proteins/metabolism , Pyroptosis , Cell Line , Cryoelectron Microscopy , Humans , Intracellular Signaling Peptides and Proteins , Liposomes/chemistry , Liposomes/ultrastructure , Microscopy, Atomic Force , Permeability , Phosphate-Binding Proteins
14.
J Virol ; 88(22): 13029-46, 2014 Nov.
Article En | MEDLINE | ID: mdl-25165113

UNLABELLED: Influenza A virus (IAV) uses the low pH in late endocytic vacuoles as a cue for penetration by membrane fusion. Here, we analyzed the prefusion reactions that prepare the core for uncoating after it has been delivered to the cytosol. We found that this priming process occurs in two steps that are mediated by the envelope-embedded M2 ion channel. The first weakens the interactions between the matrix protein, M1, and the viral ribonucleoprotein bundle. It involves a conformational change in a linker sequence and the C-terminal domain of M1 after exposure to a pH below 6.5. The second step is triggered by a pH of <6.0 and by the influx of K(+) ions. It causes additional changes in M1 as well as a loss of stability in the viral ribonucleoprotein bundle. Our results indicate that both the switch from Na(+) to K(+) in maturing endosomes and the decreasing pH are needed to prime IAV cores for efficient uncoating and infection of the host cell. IMPORTANCE: The entry of IAV involves several steps, including endocytosis and fusion at late endosomes. Entry also includes disassembly of the viral core, which is composed of the viral ribonucleoproteins and the RNA genome. We have found that the uncoating process of IAV is initiated long before the core is delivered into the cytosol. M2, an ion channel in the viral membrane, is activated when the virus passes through early endosomes. Here, we show that protons entering the virus through M2 cause a conformational change in the matrix protein, M1. This weakens interactions between M1 and the viral ribonucleoproteins. A second change was found to occur when the virus enters late endosomes. The preacidified core is then exposed to a high concentration of K(+), which affects the interactions between the ribonucleoproteins. Thus, when cores are finally delivered to the cytosol, they are already partially destabilized and, therefore, uncoating competent and infectious.


Endosomes/metabolism , Endosomes/virology , Influenza A virus/drug effects , Influenza A virus/physiology , Potassium/metabolism , Viral Matrix Proteins/metabolism , Virus Uncoating/drug effects , Animals , Humans , Hydrogen-Ion Concentration , Protein Binding/drug effects , Protein Conformation/drug effects
...