Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Annu Rev Cell Dev Biol ; 30: 337-56, 2014.
Article in English | MEDLINE | ID: mdl-25150011

ABSTRACT

Cell turnover is a fundamental feature in metazoans. Cells can die passively, as a consequence of severe damage to their structural integrity, or actively, owing to a more confined biological disruption such as DNA damage. Passive cell death is uncontrolled and often harmful to the organism. In contrast, active cell death is tightly regulated and serves to support the organism's life. Apoptosis-the primary form of regulated cell death-is relatively well defined. Necroptosis-an alternative, distinct kind of regulated cell death discovered more recently-is less well understood. Apoptosis and necroptosis can be triggered either from within the cell or by extracellular stimuli. Certain signaling components, including several death ligands and receptors, can regulate both processes. Whereas apoptosis is triggered and executed via intracellular proteases called caspases, necroptosis is suppressed by caspase activity. Here we highlight current understanding of the key signaling mechanisms that control regulated cell death.


Subject(s)
Cell Death/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/physiology , Caspases/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Enzyme Activation , Humans , Models, Biological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factors/physiology
2.
J Biol Chem ; 299(6): 104792, 2023 06.
Article in English | MEDLINE | ID: mdl-37150321

ABSTRACT

Necroptosis is a form of regulated cell death triggered by various host and pathogen-derived molecules during infection and inflammation. The essential step leading to necroptosis is phosphorylation of the mixed lineage kinase domain-like protein by receptor-interacting protein kinase 3. Caspase-8 cleaves receptor-interacting protein kinases to block necroptosis, so synthetic caspase inhibitors are required to study this process in experimental models. However, it is unclear how caspase-8 activity is regulated in a physiological setting. The active site cysteine of caspases is sensitive to oxidative inactivation, so we hypothesized that oxidants generated at sites of inflammation can inhibit caspase-8 and promote necroptosis. Here, we discovered that hypothiocyanous acid (HOSCN), an oxidant generated in vivo by heme peroxidases including myeloperoxidase and lactoperoxidase, is a potent caspase-8 inhibitor. We found HOSCN was able to promote necroptosis in mouse fibroblasts treated with tumor necrosis factor. We also demonstrate purified caspase-8 was inactivated by low concentrations of HOSCN, with the predominant product being a disulfide-linked dimer between Cys360 and Cys409 of the large and small catalytic subunits. We show oxidation still occurred in the presence of reducing agents, and reduction of the dimer was slow, consistent with HOSCN being a powerful physiological caspase inhibitor. While the initial oxidation product is a dimer, further modification also occurred in cells treated with HOSCN, leading to higher molecular weight caspase-8 species. Taken together, these findings indicate major disruption of caspase-8 function and suggest a novel mechanism for the promotion of necroptosis at sites of inflammation.


Subject(s)
Caspase 8 , Necroptosis , Oxidants , Tumor Necrosis Factors , Animals , Mice , Caspase 8/chemistry , Caspase 8/metabolism , Inflammation/metabolism , Necroptosis/drug effects , Oxidants/metabolism , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Tumor Necrosis Factors/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Peroxidase , Lactoperoxidase , Catalytic Domain
3.
Cell ; 137(4): 604-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450509

ABSTRACT

Caspase-8 is activated at the plasma membrane by the death-inducing signaling complex (DISC). Jin et al. (2009) show that polyubiquitination of caspase-8, rather than targeting it for proteasomal degradation, is critical for sustaining caspase-8 activity after dissociation from the DISC.


Subject(s)
Caspase 8/metabolism , Cullin Proteins/metabolism , Apoptosis , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Humans , Ubiquitination
4.
Int J Mol Sci ; 25(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38791309

ABSTRACT

The protein scaffold that includes the caspases is ancient and found in all domains of life. However, the stringent specificity that defines the caspase biologic function is relatively recent and found only in multicellular animals. During the radiation of the Chordata, members of the caspase family adopted roles in immunity, events coinciding with the development of substrates that define the modern innate immune response. This review focuses on the switch from the non-inflammatory cellular demise of apoptosis to the highly inflammatory innate response driven by distinct members of the caspase family, and the interplay between these two regulated cell death pathways.


Subject(s)
Caspases , Immunity, Innate , Pyroptosis , Humans , Caspases/metabolism , Animals , Evolution, Molecular , Apoptosis
5.
J Biol Chem ; 298(6): 101931, 2022 06.
Article in English | MEDLINE | ID: mdl-35427646

ABSTRACT

Pyroptosis is a mechanism of inflammatory cell death mediated by the activation of the prolytic protein gasdermin D by caspase-1, caspase-4, and caspase-5 in human, and caspase-1 and caspase-11 in mouse. In addition, caspase-1 amplifies inflammation by proteolytic activation of cytokine interleukin-1ß (IL-1ß). Modern mammals of the order Carnivora lack the caspase-1 catalytic domain but express an unusual version of caspase-4 that can activate both gasdermin D and IL-1ß. Seeking to understand the evolutionary origin of this caspase, we utilized the large amount of data available in public databases to perform ancestral sequence reconstruction of an inflammatory caspase of a Carnivora ancestor. We expressed the catalytic domain of this putative ancestor in Escherichia coli, purified it, and compared its substrate specificity on synthetic and protein substrates to extant caspases. We demonstrated that it activates gasdermin D but has reduced ability to activate IL-1ß. Our reconstruction suggests that caspase-1 was lost in a Carnivora ancestor, perhaps upon a selective pressure for which the generation of biologically active IL-1ß by caspase-1 was detrimental. We speculate that later, a Carnivora encountered selective pressures that required the production of IL-1ß, and caspase-4 subsequently gained this activity. This hypothesis would explain why extant Carnivora possess an inflammatory caspase with caspase-1 catalytic function placed on a caspase-4 scaffold.


Subject(s)
Caspases , Animals , Carnivora/genetics , Carnivora/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Caspases/genetics , Caspases/metabolism , Escherichia coli/genetics , Inflammation/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pyroptosis/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Selection, Genetic
6.
Mol Cell ; 77(5): 927-929, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142688
7.
Biochem J ; 479(14): 1533-1542, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35789254

ABSTRACT

A patient diagnosed with multiple myeloma, bicuspid aortic valve, and Von Hippel-Lindau syndrome underwent whole-exome sequencing seeking a unified genetic cause for these three pathologies. The patient possessed a single-point mutation of arginine to cysteine (R24C) in the N-terminal region(pro-domain) of matrix metalloproteinase 9 (MMP-9). The pro-domain interacts with the catalytic site of this enzyme rendering it inactive. MMP-9 has previously been associated with all three pathologies suffered by the patient. We hypothesized that the observed mutation in the pro-domain would influence the activity of this enzyme. We expressed recombinant versions of MMP-9 and an investigation of their biochemical properties revealed that MMP-9 R24C is a constitutively active zymogen. To our knowledge, this is the first example of a mutation that discloses catalytic activity in the pro-form in any of the 24 human MMPs.


Subject(s)
Bicuspid Aortic Valve Disease , Multiple Myeloma , von Hippel-Lindau Disease , Gain of Function Mutation , Humans , Matrix Metalloproteinase 9/genetics , Multiple Myeloma/complications , Multiple Myeloma/genetics , von Hippel-Lindau Disease/complications , von Hippel-Lindau Disease/genetics
8.
Proc Natl Acad Sci U S A ; 117(18): 9952-9963, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32345717

ABSTRACT

Genetic polymorphisms in the region of the trimeric serine hydrolase high-temperature requirement 1 (HTRA1) are associated with increased risk of age-related macular degeneration (AMD) and disease progression, but the precise biological function of HtrA1 in the eye and its contribution to disease etiologies remain undefined. In this study, we have developed an HtrA1-blocking Fab fragment to test the therapeutic hypothesis that HtrA1 protease activity is involved in the progression of AMD. Next, we generated an activity-based small-molecule probe (ABP) to track target engagement in vivo. In addition, we used N-terminomic proteomic profiling in preclinical models to elucidate the in vivo repertoire of HtrA1-specific substrates, and identified substrates that can serve as robust pharmacodynamic biomarkers of HtrA1 activity. One of these HtrA1 substrates, Dickkopf-related protein 3 (DKK3), was successfully used as a biomarker to demonstrate the inhibition of HtrA1 activity in patients with AMD who were treated with the HtrA1-blocking Fab fragment. This pharmacodynamic biomarker provides important information on HtrA1 activity and pharmacological inhibition within the ocular compartment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antibodies, Anti-Idiotypic/pharmacology , Geographic Atrophy/drug therapy , High-Temperature Requirement A Serine Peptidase 1/genetics , Macular Degeneration/drug therapy , Adaptor Proteins, Signal Transducing/isolation & purification , Aged , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Biomarkers/blood , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Geographic Atrophy/blood , Geographic Atrophy/genetics , Geographic Atrophy/immunology , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Macular Degeneration/blood , Macular Degeneration/genetics , Macular Degeneration/immunology , Male , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Proteome/immunology , Rats , Retina/drug effects , Retina/immunology , Retina/pathology , Small Molecule Libraries/pharmacology
9.
Trends Biochem Sci ; 43(10): 829-844, 2018 10.
Article in English | MEDLINE | ID: mdl-30097385

ABSTRACT

Proteases are considered of major importance in biomedical research because of their crucial roles in health and disease. Their ability to hydrolyze their protein and peptide substrates at single or multiple sites, depending on their specificity, makes them unique among the enzymes. Understanding protease specificity is therefore crucial to understand their biology as well as to develop tools and drugs. Recent advancements in the fields of proteomics and chemical biology have improved our understanding of protease biology through extensive specificity profiling and identification of physiological protease substrates. There are growing efforts to transfer this knowledge into clinical modalities, but their success is often limited because of overlapping protease features, protease redundancy, and chemical tools lacking specificity. Herein, we discuss the current trends and challenges in protease research and how to exploit the growing information on protease specificities for understanding protease biology, as well as for development of selective substrates, cleavable linkers, and activity-based probes and for biomarker discovery.


Subject(s)
Peptide Hydrolases/metabolism , Biomarkers/metabolism , Datasets as Topic , Humans , Proteomics , Substrate Specificity
10.
Biochem J ; 478(12): 2233-2245, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34037204

ABSTRACT

Caspases are a family of enzymes that play roles in cell death and inflammation. It has been suggested that in the execution phase of the apoptotic pathway, caspase-3, -6 and -7 are involved. The substrate specificities of two proteases (caspases 3 and 7) are highly similar, which complicates the design of compounds that selectively interact with a single enzyme exclusively. The recognition of residues other than Asp in the P1 position of the substrate by caspase-3/-7 has been reported, promoting interest in the effects of phosphorylation of amino acids in the direct vicinity of the scissile bond. To evaluate conflicting reports on this subject, we synthesized a series of known caspase-3 and -7 substrates and phosphorylated analogs, performed enzyme kinetic assays and mapped the peptide cleavage sites using internally quenched fluorescent peptide substrates. Caspases 3 and 7 will tolerate pSer at the P1 position but only poorly at the P2' position. Our investigation demonstrates the importance of peptide length and composition in interpreting sequence/activity relationships. Based on the results, we conclude that the relationship between caspase-3/-7 and their substrates containing phosphorylated amino acids might depend on the steric conditions and not be directly connected with ionic interactions. Thus, the precise effect of phospho-amino acid residues located in the vicinity of the cleaved bond on the regulation of the substrate specificity of caspases remains difficult to predict. Our observations allow to predict that natural phosphorylated proteins may be cleaved by caspases, but only when extended substrate binding site interactions are satisfied.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Peptide Fragments/metabolism , Proteolysis , Serine/metabolism , Transcription Factors/chemistry , Vimentin/chemistry , Binding Sites , Caspase 3/genetics , Caspase 7/genetics , Humans , Kinetics , Models, Molecular , Phosphorylation , Serine/chemistry , Substrate Specificity , YAP-Signaling Proteins
11.
J Biol Chem ; 295(32): 11292-11302, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32554464

ABSTRACT

Pyroptosis is the caspase-dependent inflammatory cell death mechanism that underpins the innate immune response against pathogens and is dysregulated in inflammatory disorders. Pyroptosis occurs via two pathways: the canonical pathway, signaled by caspase-1, and the noncanonical pathway, regulated by mouse caspase-11 and human caspase-4/5. All inflammatory caspases activate the pyroptosis effector protein gasdermin D, but caspase-1 mostly activates the inflammatory cytokine precursors prointerleukin-18 and prointerleukin-1ß (pro-IL18/pro-IL1ß). Here, in vitro cleavage assays with recombinant proteins confirmed that caspase-11 prefers cleaving gasdermin D over the pro-ILs. However, we found that caspase-11 recognizes protein substrates through a mechanism that is different from that of most caspases. Results of kinetics analysis with synthetic fluorogenic peptides indicated that P1'-P4', the C-terminal gasdermin D region adjacent to the cleavage site, influences gasdermin D recognition by caspase-11. Furthermore, introducing the gasdermin D P1'-P4' region into pro-IL18 enhanced catalysis by caspase-11 to levels comparable with that of gasdermin D cleavage. Pro-IL1ß cleavage was only moderately enhanced by similar substitutions. We conclude that caspase-11 specificity is mediated by the P1'-P4' region in its substrate gasdermin D, and similar experiments confirmed that the substrate specificities of the human orthologs of caspase-11, i.e. caspase-4 and caspase-5, are ruled by the same mechanism. We propose that P1'-P4'-based inhibitors could be exploited to specifically target inflammatory caspases.


Subject(s)
Caspases/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Phosphate-Binding Proteins/physiology , Pyroptosis , Animals , Catalysis , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Phosphate-Binding Proteins/chemistry , Phosphate-Binding Proteins/metabolism , Proteolysis , Substrate Specificity
12.
J Biol Chem ; 295(8): 2464-2472, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31953328

ABSTRACT

Since their discovery, the matrix metalloproteinase (MMP) family proteases have been considered as therapeutic targets in numerous diseases and disorders. Unfortunately, clinical trials with MMP inhibitors have failed to yield any clinical benefits of these inhibitors. These failures were largely due to a lack of MMP-selective agents; accordingly, it has become important to identify a platform with which high selectivity can be achieved. To this end, we propose using MMP-targeting antibodies that can achieve high specificity in interactions with their targets. Using a scaffold of single-domain antibodies, here we raised a panel of MMP10-selective antibodies through immunization of llamas, a member of the camelid family, whose members generate conventional heavy/light-chain antibodies and also smaller antibodies lacking light-chain and CH1 domains. We report the generation of a highly selective and tightly binding MMP10 inhibitor (Ki < 2 nm). Using bio-layer interferometry-based binding assays, we found that this antibody interacts with the MMP10 active site. Activity assays demonstrated that the antibody selectively inhibits MMP10 over its closest relative, MMP3. The ability of a single-domain antibody to discriminate between the most conserved MMP pair via an active site-directed mechanism of inhibition reported here supports the potential of this antibody as a broadly applicable scaffold for the development of selective, tightly binding MMP inhibitors.


Subject(s)
Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Single-Domain Antibodies/pharmacology , Animals , Camelids, New World , Humans , Immunization , Kinetics , Peptide Library , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Substrate Specificity/drug effects , alpha 1-Antitrypsin/metabolism
13.
J Biol Chem ; 295(51): 17624-17631, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33454002

ABSTRACT

Neutrophils are primary host innate immune cells defending against pathogens. One proposed mechanism by which neutrophils prevent the spread of pathogens is NETosis, the extrusion of cellular DNA resulting in neutrophil extracellular traps (NETs). The protease neutrophil elastase (NE) has been implicated in the formation of NETs through proteolysis of nuclear proteins leading to chromatin decondensation. In addition to NE, neutrophils contain three other serine proteases that could compensate if the activity of NE was neutralized. However, whether they do play such a role is unknown. Thus, we deployed recently described specific inhibitors against all four of the neutrophil serine proteases (NSPs). Using specific antibodies to the NSPs along with our labeled inhibitors, we show that catalytic activity of these enzymes is not required for the formation of NETs. Moreover, the NSPs that decorate NETs are in an inactive conformation and thus cannot participate in further catalytic events. These results indicate that NSPs play no role in either NETosis or arming NETs with proteolytic activity.


Subject(s)
Extracellular Traps/metabolism , Neutrophils/enzymology , Serine Proteases/metabolism , Animals , Antibodies/chemistry , Antibodies/immunology , Candida albicans/physiology , DNA/metabolism , Escherichia coli/physiology , Extracellular Traps/drug effects , Humans , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/immunology , Leukocyte Elastase/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Confocal , Neutrophils/drug effects , Pyroptosis/drug effects , RAW 264.7 Cells , Serine Proteases/chemistry , Serine Proteases/immunology , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Tetradecanoylphorbol Acetate/pharmacology
14.
J Biol Chem ; 295(28): 9567-9582, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32439802

ABSTRACT

Natural killer (NK) cells are key innate immunity effectors that combat viral infections and control several cancer types. For their immune function, human NK cells rely largely on five different cytotoxic proteases, called granzymes (A/B/H/K/M). Granzyme B (GrB) initiates at least three distinct cell death pathways, but key aspects of its function remain unexplored because selective probes that detect its activity are currently lacking. In this study, we used a set of unnatural amino acids to fully map the substrate preferences of GrB, demonstrating previously unknown GrB substrate preferences. We then used these preferences to design substrate-based inhibitors and a GrB-activatable activity-based fluorogenic probe. We show that our GrB probes do not significantly react with caspases, making them ideal for in-depth analyses of GrB localization and function in cells. Using our quenched fluorescence substrate, we observed GrB within the cytotoxic granules of human YT cells. When used as cytotoxic effectors, YT cells loaded with GrB attacked MDA-MB-231 target cells, and active GrB influenced its target cell-killing efficiency. In summary, we have developed a set of molecular tools for investigating GrB function in NK cells and demonstrate noninvasive visual detection of GrB with an enzyme-activated fluorescent substrate.


Subject(s)
Fluorescent Dyes/chemistry , Granzymes , Optical Imaging , Peptides/chemistry , T-Lymphocytes/enzymology , Cell Line, Tumor , Granzymes/chemistry , Granzymes/metabolism , Humans
15.
J Immunol ; 203(3): 736-748, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31209100

ABSTRACT

The pyroptotic cell death effector gasdermin D (GSDMD) is required for murine models of hereditary inflammasome-driven, IL-1ß-dependent, autoinflammatory disease, making it an attractive therapeutic target. However, the importance of GSDMD for more common conditions mediated by pathological IL-1ß activation, such as gout, remain unclear. In this study, we address whether GSDMD and the recently described GSDMD inhibitor necrosulfonamide (NSA) contribute to monosodium urate (MSU) crystal-induced cell death, IL-1ß release, and autoinflammation. We demonstrate that MSU crystals, the etiological agent of gout, rapidly activate GSDMD in murine macrophages. Despite this, the genetic deletion of GSDMD or the other lytic effector implicated in MSU crystal killing, mixed lineage kinase domain-like (MLKL), did not prevent MSU crystal-induced cell death. Consequently, GSDMD or MLKL loss did not hinder MSU crystal-mediated release of bioactive IL-1ß. Consistent with in vitro findings, IL-1ß induction and autoinflammation in MSU crystal-induced peritonitis was not reduced in GSDMD-deficient mice. Moreover, we show that the reported GSDMD inhibitor, NSA, blocks inflammasome priming and caspase-1 activation, thereby preventing pyroptosis independent of GSDMD targeting. The inhibition of cathepsins, widely implicated in particle-induced macrophage killing, also failed to prevent MSU crystal-mediated cell death. These findings 1) demonstrate that not all IL-1ß-driven autoinflammatory conditions will benefit from the therapeutic targeting of GSDMD, 2) document a unique mechanism of MSU crystal-induced macrophage cell death not rescued by pan-cathepsin inhibition, and 3) show that NSA inhibits inflammasomes upstream of GSDMD to prevent pyroptotic cell death and IL-1ß release.


Subject(s)
Gout/pathology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis/physiology , Uric Acid/metabolism , Acrylamides/pharmacology , Animals , Caspase 1/metabolism , Cathepsins/antagonists & inhibitors , Female , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrofurans/pharmacology , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/pathology , Phosphate-Binding Proteins/genetics , Protein Kinases/genetics , Styrenes/pharmacology , Sulfonamides/pharmacology
16.
Cell ; 147(2): 476-476.e1, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-22000022
17.
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
18.
Semin Cell Dev Biol ; 82: 79-85, 2018 10.
Article in English | MEDLINE | ID: mdl-29329946

ABSTRACT

Caspases belong to a diverse clan of proteolytic enzymes known as clan CD with highly disparate functions in cell signaling. The caspase members of this clan are only found in animals, and most of them orchestrate the demise of cells by the highly distinct regulated cell death phenotypes known as apoptosis and pyroptosis. This review looks at the mechanistic distinctions between the activity and activation mechanisms of mammalian caspases compared to other members of clan CD. We also compare and contrast the role of different caspase family members that program anti-inflammatory and pro-inflammatory cell death pathways.


Subject(s)
Caspases/metabolism , Humans
19.
J Am Chem Soc ; 142(39): 16704-16715, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32870676

ABSTRACT

The subset of the proteome that contains enzymes in their catalytically active form can be interrogated by using probes targeted toward individual specific enzymes. A subset of such enzymes are proteases that are frequently studied with activity-based probes, small inhibitors equipped with a detectable tag, commonly a fluorophore. Due to the spectral overlap of these commonly used fluorophores, multiplex analysis becomes limited. To overcome this, we developed a series of protease-selective lanthanide-labeled probes compatible with mass cytometry giving us the ability to monitor the activity of multiple proteases in parallel. Using these probes, we were able to identify the distribution of four proteases with different active site geometries in three cell lines and peripheral blood mononuclear cells. This provides a framework for the use of mass cytometry for multiplexed enzyme activity detection.


Subject(s)
Coordination Complexes/chemistry , Lanthanoid Series Elements/chemistry , Molecular Probes/chemistry , Peptide Hydrolases/analysis , Cell Line , Coordination Complexes/chemical synthesis , Humans , Molecular Probes/chemical synthesis , Molecular Structure , Peptide Hydrolases/metabolism
20.
J Enzyme Inhib Med Chem ; 35(1): 1387-1402, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32633155

ABSTRACT

Aza-peptide aldehydes and ketones are a new class of reversible protease inhibitors that are specific for the proteasome and clan CD cysteine proteases. We designed and synthesised aza-Leu derivatives that were specific for the chymotrypsin-like active site of the proteasome, aza-Asp derivatives that were effective inhibitors of caspases-3 and -6, and aza-Asn derivatives that inhibited S. mansoni and I. ricinus legumains. The crystal structure of caspase-3 in complex with our caspase-specific aza-peptide methyl ketone inhibitor with an aza-Asp residue at P1 revealed a covalent linkage between the inhibitor carbonyl carbon and the active site cysteinyl sulphur. Aza-peptide aldehydes and ketones showed no cross-reactivity towards cathepsin B or chymotrypsin. The initial in vitro selectivity of these inhibitors makes them suitable candidates for further development into therapeutic agents to potentially treat multiple myeloma, neurodegenerative diseases, and parasitic infections.


Subject(s)
Aldehydes/pharmacology , Aza Compounds/pharmacology , Drug Design , Ketones/pharmacology , Peptides/pharmacology , Protease Inhibitors/pharmacology , Aldehydes/chemistry , Animals , Aza Compounds/chemistry , Cattle , Crystallography, X-Ray , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Humans , Ketones/chemistry , Models, Molecular , Molecular Structure , Peptides/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL