Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38991619

ABSTRACT

Mounting evidence indicates that proteotoxic stress is a primary activator of the CARD8 inflammasome, but the complete array of signals that control this inflammasome have not yet been established. Notably, we recently discovered that several hydrophobic radical-trapping antioxidants (RTAs), including JSH-23, potentiate CARD8 inflammasome activation through an unknown mechanism. Here, we report that these RTAs directly alkylate several cysteine residues in the N-terminal disordered region of CARD8. These hydrophobic modifications destabilize the repressive CARD8 N-terminal fragment and accelerate its proteasome-mediated degradation, thereby releasing the inflammatory CARD8 C-terminal fragment from autoinhibition. Consistently, we also found that unrelated (non-RTA) hydrophobic electrophiles as well as genetic mutation of the CARD8 cysteine residues to isoleucines similarly potentiate inflammasome activation. Overall, our results not only provide further evidence that protein folding stress is a key CARD8 inflammasome-activating signal, but also indicate that the N-terminal cysteines can play key roles in tuning the response to this stress.

2.
Cell Rep ; 42(1): 111965, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36649711

ABSTRACT

NLRP1 and CARD8 are related pattern-recognition receptors (PRRs) that detect intracellular danger signals and form inflammasomes. Both undergo autoproteolysis, generating N-terminal (NT) and C-terminal (CT) fragments. The proteasome-mediated degradation of the NT releases the CT from autoinhibition, but the stimuli that trigger NT degradation have not been fully elucidated. Here, we show that several distinct agents that interfere with protein folding, including aminopeptidase inhibitors, chaperone inhibitors, and inducers of the unfolded protein response, accelerate NT degradation. However, these agents alone do not trigger inflammasome formation because the released CT fragments are physically sequestered by the serine dipeptidase DPP9. We show that DPP9-binding ligands must also be present to disrupt these complexes and allow the CT fragments to oligomerize into inflammasomes. Overall, these results indicate that NLRP1 and CARD8 detect a specific perturbation that induces both protein folding stress and DPP9 ligand accumulation.


Subject(s)
Adaptor Proteins, Signal Transducing , Inflammasomes , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , NLR Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Protein Folding , CARD Signaling Adaptor Proteins/metabolism
3.
J Biol Chem ; 298(7): 102032, 2022 07.
Article in English | MEDLINE | ID: mdl-35580636

ABSTRACT

CARD8 is a pattern-recognition receptor that forms a caspase-1-activating inflammasome. CARD8 undergoes constitutive autoproteolysis, generating an N-terminal (NT) fragment with a disordered region and a ZU5 domain and a C-terminal (CT) fragment with UPA and CARD domains. Dipeptidyl peptidase 8 and dipeptidyl peptidase 9 inhibitors, including Val-boroPro, accelerate the degradation of the NT fragment via a poorly characterized proteasome-mediated pathway, thereby releasing the inflammatory CT fragment from autoinhibition. Here, we show that the core 20S proteasome, which degrades disordered and misfolded proteins independent of ubiquitin modification, controls activation of the CARD8 inflammasome. In unstressed cells, we discovered that the 20S proteasome degrades just the NT disordered region, leaving behind the folded ZU5, UPA, and CARD domains to act as an inhibitor of inflammasome assembly. However, in Val-boroPro-stressed cells, we show the 20S proteasome degrades the entire NT fragment, perhaps due to ZU5 domain unfolding, freeing the CT fragment from autoinhibition. Taken together, these results show that the susceptibility of the CARD8 NT domain to 20S proteasome-mediated degradation controls inflammasome activation.


Subject(s)
CARD Signaling Adaptor Proteins , Inflammasomes , Proteasome Endopeptidase Complex , CARD Signaling Adaptor Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism
4.
Nat Chem Biol ; 18(5): 565-574, 2022 05.
Article in English | MEDLINE | ID: mdl-35165443

ABSTRACT

Inflammasomes are multiprotein complexes that sense intracellular danger signals and induce pyroptosis. CARD8 and NLRP1 are related inflammasomes that are repressed by the enzymatic activities and protein structures of the dipeptidyl peptidases 8 and 9 (DPP8/9). Potent DPP8/9 inhibitors such as Val-boroPro (VbP) activate both NLRP1 and CARD8, but chemical probes that selectively activate only one have not been identified. Here we report a small molecule called CQ31 that selectively activates CARD8. CQ31 inhibits the M24B aminopeptidases prolidase (PEPD) and Xaa-Pro aminopeptidase 1 (XPNPEP1), leading to the accumulation of proline-containing peptides that inhibit DPP8/9 and thereby activate CARD8. NLRP1 is distinct from CARD8 in that it directly contacts DPP8/9's active site; these proline-containing peptides, unlike VbP, do not disrupt this repressive interaction and thus do not activate NLRP1. We expect that CQ31 will now become a valuable tool to study CARD8 biology.


Subject(s)
CARD Signaling Adaptor Proteins , Inflammasomes , Aminopeptidases/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Neoplasm Proteins , Proline
5.
Cell Rep ; 33(2): 108264, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053349

ABSTRACT

Several cytosolic pattern-recognition receptors (PRRs) form multiprotein complexes called canonical inflammasomes in response to intracellular danger signals. Canonical inflammasomes recruit and activate caspase-1 (CASP1), which in turn cleaves and activates inflammatory cytokines and gasdermin D (GSDMD), inducing pyroptotic cell death. Inhibitors of the dipeptidyl peptidases DPP8 and DPP9 (DPP8/9) activate both the human NLRP1 and CARD8 inflammasomes. NLRP1 and CARD8 have different N-terminal regions but have similar C-terminal regions that undergo autoproteolysis to generate two non-covalently associated fragments. Here, we show that DPP8/9 inhibition activates a proteasomal degradation pathway that targets disordered and misfolded proteins for destruction. CARD8's N terminus contains a disordered region of ∼160 amino acids that is recognized and destroyed by this degradation pathway, thereby freeing its C-terminal fragment to activate CASP1 and induce pyroptosis. Thus, CARD8 serves as an alarm to signal the activation of a degradation pathway for disordered and misfolded proteins.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/metabolism , Intrinsically Disordered Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Animals , Boronic Acids/pharmacology , Dipeptides/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , HEK293 Cells , Humans , Lysine/metabolism , Mice , Proteolysis , Proteostasis , RAW 264.7 Cells , THP-1 Cells
6.
ACS Chem Biol ; 14(11): 2424-2429, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31525884

ABSTRACT

Inflammasomes are multiprotein complexes formed in response to pathogens. NLRP1 and CARD8 are related proteins that form inflammasomes, but the pathogen-associated signal(s) and the molecular mechanisms controlling their activation have not been established. Inhibitors of the serine dipeptidyl peptidases DPP8 and DPP9 (DPP8/9) activate both NLRP1 and CARD8. Interestingly, DPP9 binds directly to NLRP1 and CARD8, and this interaction may contribute to the inhibition of NLRP1. Here, we use activity-based probes, reconstituted inflammasome assays, and mass spectrometry-based proteomics to further investigate the DPP9-CARD8 interaction. We show that the DPP9-CARD8 interaction, unlike the DPP9-NLRP1 interaction, is not disrupted by DPP9 inhibitors or CARD8 mutations that block autoproteolysis. Moreover, wild-type, but not catalytically inactive mutant, DPP9 rescues CARD8-mediated cell death in DPP9 knockout cells. Together, this work reveals that DPP9's catalytic activity and not its binding to CARD8 restrains the CARD8 inflammasome and thus suggests the binding interaction likely serves some other biological purpose.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Dipeptidases/metabolism , HEK293 Cells , Humans , Mutation , NLR Proteins , Organofluorophosphonates/metabolism , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation , Signal Transduction
7.
Cell Death Dis ; 10(8): 587, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31383852

ABSTRACT

Intracellular pathogenic structures or activities stimulate the formation of inflammasomes, which recruit and activate caspase-1 and trigger an inflammatory form of cell death called pyroptosis. The well-characterized mammalian inflammasome sensor proteins all detect one specific type of signal, for example double-stranded DNA or bacterial flagellin. Remarkably, NLRP1 was the first protein discovered to form an inflammasome, but the pathogenic signal that NLRP1 detects has not yet been identified. NLRP1 is highly polymorphic, even among inbred rodent strains, and it has been suggested that these diverse NLRP1 alleles may have evolved to detect entirely different stimuli. Intriguingly, inhibitors of the serine proteases DPP8 and DPP9 (DPP8/9) were recently shown to activate human NLRP1, its homolog CARD8, and several mouse NLRP1 alleles. Here, we show now that DPP8/9 inhibitors activate all functional rodent NLRP1 alleles, indicating that DPP8/9 inhibition induces a signal detected by all NLRP1 proteins. Moreover, we discovered that the NLRP1 allele sensitivities to DPP8/9 inhibitor-induced and Toxoplasma gondii-induced pyroptosis are strikingly similar, suggesting that DPP8/9 inhibition phenocopies a key activity of T. gondii. Overall, this work indicates that the highly polymorphic NLRP1 inflammasome indeed senses a specific signal like the other mammalian inflammasomes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alleles , Apoptosis Regulatory Proteins/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Bacterial/pharmacology , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/pharmacology , Boronic Acids/pharmacology , Dipeptides/pharmacology , Female , HEK293 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nerve Tissue Proteins/metabolism , Pyroptosis/drug effects , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Rats, Zucker , Serine Proteinase Inhibitors/pharmacology , Toxoplasma/immunology , Transfection
8.
Science ; 364(6435): 82-85, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30872531

ABSTRACT

Intracellular pathogens and danger signals trigger the formation of inflammasomes, which activate inflammatory caspases and induce pyroptosis. The anthrax lethal factor metalloprotease and small-molecule DPP8/9 inhibitors both activate the NLRP1B inflammasome, but the molecular mechanism of NLRP1B activation is unknown. In this study, we used genome-wide CRISPR-Cas9 knockout screens to identify genes required for NLRP1B-mediated pyroptosis. We discovered that lethal factor induces cell death via the N-end rule proteasomal degradation pathway. Lethal factor directly cleaves NLRP1B, inducing the N-end rule-mediated degradation of the NLRP1B N terminus and freeing the NLRP1B C terminus to activate caspase-1. DPP8/9 inhibitors also induce proteasomal degradation of the NLRP1B N terminus but not via the N-end rule pathway. Thus, N-terminal degradation is the common activation mechanism of this innate immune sensor.


Subject(s)
Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/metabolism , Inflammasomes/metabolism , Proteolysis , Pyroptosis/physiology , Animals , Apoptosis Regulatory Proteins/genetics , CRISPR-Cas Systems , Caspase 1/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Gene Knockout Techniques , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Mice , Proteasome Endopeptidase Complex/metabolism , Pyroptosis/genetics , RAW 264.7 Cells , Serine Proteinase Inhibitors/pharmacology , THP-1 Cells , Ubiquitin-Protein Ligases/genetics
9.
Nat Med ; 24(8): 1151-1156, 2018 08.
Article in English | MEDLINE | ID: mdl-29967349

ABSTRACT

Small-molecule inhibitors of the serine dipeptidases DPP8 and DPP9 (DPP8/9) induce a lytic form of cell death called pyroptosis in mouse and human monocytes and macrophages1,2. In mouse myeloid cells, Dpp8/9 inhibition activates the inflammasome sensor Nlrp1b, which in turn activates pro-caspase-1 to mediate cell death3, but the mechanism of DPP8/9 inhibitor-induced pyroptosis in human myeloid cells is not yet known. Here we show that the CARD-containing protein CARD8 mediates DPP8/9 inhibitor-induced pro-caspase-1-dependent pyroptosis in human myeloid cells. We further show that DPP8/9 inhibitors induce pyroptosis in the majority of human acute myeloid leukemia (AML) cell lines and primary AML samples, but not in cells from many other lineages, and that these inhibitors inhibit human AML progression in mouse models. Overall, this work identifies an activator of CARD8 in human cells and indicates that its activation by small-molecule DPP8/9 inhibitors represents a new potential therapeutic strategy for AML.


Subject(s)
Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Protease Inhibitors/therapeutic use , Pyroptosis/drug effects , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Cell Line, Tumor , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Disease Progression , HEK293 Cells , Humans , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology
10.
Cell Chem Biol ; 25(3): 262-267.e5, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29396289

ABSTRACT

Val-boroPro (PT-100, Talabostat) induces powerful anti-tumor immune responses in syngeneic cancer models, but its mechanism of action has not yet been established. Val-boroPro is a non-selective inhibitor of post-proline-cleaving serine proteases, and the inhibition of the highly related cytosolic serine proteases Dpp8 and Dpp9 (Dpp8/9) by Val-boroPro was recently demonstrated to trigger an immunostimulatory form of programmed cell death known as pyroptosis selectively in monocytes and macrophages. Here we show that Dpp8/9 inhibition activates the inflammasome sensor protein Nlrp1b, which in turn activates pro-caspase-1 to mediate pyroptosis. This work reveals a previously unrecognized mechanism for activating an innate immune pattern recognition receptor and suggests that Dpp8/9 serve as an intracellular checkpoint to restrain Nlrp1b and the innate immune system.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Dipeptidases/metabolism , Inflammasomes/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Boronic Acids/chemistry , Boronic Acids/metabolism , Boronic Acids/pharmacology , Caspase 1/metabolism , Dipeptidases/antagonists & inhibitors , Dipeptides/chemistry , Dipeptides/metabolism , Dipeptides/pharmacology , Female , HEK293 Cells , Humans , Immunity, Innate/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Pyroptosis/drug effects , RAW 264.7 Cells
11.
Nat Chem Biol ; 13(1): 46-53, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27820798

ABSTRACT

Val-boroPro (Talabostat, PT-100), a nonselective inhibitor of post-proline cleaving serine proteases, stimulates mammalian immune systems through an unknown mechanism of action. Despite this lack of mechanistic understanding, Val-boroPro has attracted substantial interest as a potential anticancer agent, reaching phase 3 trials in humans. Here we show that Val-boroPro stimulates the immune system by triggering a proinflammatory form of cell death in monocytes and macrophages known as pyroptosis. We demonstrate that the inhibition of two serine proteases, DPP8 and DPP9, activates the pro-protein form of caspase-1 independent of the inflammasome adaptor ASC. Activated pro-caspase-1 does not efficiently process itself or IL-1ß but does cleave and activate gasdermin D to induce pyroptosis. Mice lacking caspase-1 do not show immune stimulation after treatment with Val-boroPro. Our data identify what is to our knowledge the first small molecule that induces pyroptosis and reveals a new checkpoint that controls the activation of the innate immune system.


Subject(s)
Boronic Acids/pharmacology , Caspase 1/metabolism , Dipeptidases/antagonists & inhibitors , Dipeptides/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Macrophages/drug effects , Pyroptosis/drug effects , Serine Proteinase Inhibitors/pharmacology , Animals , Boronic Acids/chemistry , Caspase 1/deficiency , Cell Line , Dipeptidases/metabolism , Dipeptides/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Dose-Response Relationship, Drug , Humans , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/pathology , Macrophages/enzymology , Macrophages/pathology , Mice , Molecular Conformation , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
12.
Biochemistry ; 54(9): 1717-28, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25715079

ABSTRACT

The intrinsically disordered protein (IDP) stathmin plays an important regulatory role in cytoskeletal maintenance through its helical binding to tubulin and microtubules. However, it lacks a stable fold in the absence of its binding partner. Although stathmin has been a focus of research over the past two decades, the solution-phase conformational dynamics of this IDP are poorly understood. It has been reported that stathmin is purely monomeric in solution and that it bears a short helical region of persistent foldedness, which may act to nucleate helical folding in the C-terminal direction. Here we report a comprehensive study of the structural equilibria local to this region in stathmin that contradicts these two claims. Using the technique of electron paramagnetic resonance (EPR) spectroscopy on spin-labeled stathmin mutants in the solution-phase and when immobilized on Sepharose solid support, we show that all sites in the helical nucleation region of stathmin exhibit multiple spectral components that correspond to dynamic states of differing mobilities and stabilities. Importantly, a state with relatively low mobility dominates each spectrum with an average population greater than 50%, which we suggest corresponds to an oligomerized state of the protein. This is in contrast to a less populated, more mobile state, which likely represents a helically folded monomeric state of stathmin, and a highly mobile state, which we propose is the random coil conformer of the protein. Our interpretation of the EPR data is confirmed by further characterization of the protein using the techniques of native and SDS PAGE, gel filtration chromatography, and multiangle and dynamic light scattering, all of which show the presence of oligomeric stathmin in solution. Collectively, these data suggest that stathmin exists in a diverse equilibrium of states throughout the purported helical nucleation region and that this IDP exhibits a propensity toward oligomerization.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Stathmin/chemistry , Amino Acid Sequence , Circular Dichroism , Electron Spin Resonance Spectroscopy , Humans , Protein Folding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Stathmin/metabolism , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...