Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021065

ABSTRACT

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Base Sequence , Case-Control Studies , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Methotrexate/pharmacology , Mutation/genetics , Phosphorylation/drug effects , Polymorphism, Single Nucleotide/genetics , Pyroptosis/drug effects , Pyroptosis/genetics , Reproducibility of Results , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Wound Healing/drug effects , Wound Healing/genetics
3.
Nat Immunol ; 20(10): 1299-1310, 2019 10.
Article in English | MEDLINE | ID: mdl-31534238

ABSTRACT

Resisting and tolerating microbes are alternative strategies to survive infection, but little is known about the evolutionary mechanisms controlling this balance. Here genomic analyses of anatomically modern humans, extinct Denisovan hominins and mice revealed a TNFAIP3 allelic series with alterations in the encoded immune response inhibitor A20. Each TNFAIP3 allele encoded substitutions at non-catalytic residues of the ubiquitin protease OTU domain that diminished IκB kinase-dependent phosphorylation and activation of A20. Two TNFAIP3 alleles encoding A20 proteins with partial phosphorylation deficits seemed to be beneficial by increasing immunity without causing spontaneous inflammatory disease: A20 T108A;I207L, originating in Denisovans and introgressed in modern humans throughout Oceania, and A20 I325N, from an N-ethyl-N-nitrosourea (ENU)-mutagenized mouse strain. By contrast, a rare human TNFAIP3 allele encoding an A20 protein with 95% loss of phosphorylation, C243Y, caused spontaneous inflammatory disease in humans and mice. Analysis of the partial-phosphorylation A20 I325N allele in mice revealed diminished tolerance of bacterial lipopolysaccharide and poxvirus inoculation as tradeoffs for enhanced immunity.


Subject(s)
Poxviridae Infections/immunology , Poxviridae/physiology , Protein Domains/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Alleles , Animals , Extinction, Biological , Humans , Immunity , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense/genetics , Phosphorylation
4.
Immunity ; 53(1): 106-114.e5, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32553275

ABSTRACT

The recognition and cleavage of gasdermin D (GSDMD) by inflammatory caspases-1, 4, 5, and 11 are essential steps in initiating pyroptosis after inflammasome activation. Previous work has identified cleavage site signatures in substrates such as GSDMD, but it is unclear whether these are the sole determinants for caspase engagement. Here we report the crystal structure of a complex between human caspase-1 and the full-length murine GSDMD. In addition to engagement of the GSDMD N- and C-domain linker by the caspase-1 active site, an anti-parallel ß sheet at the caspase-1 L2 and L2' loops bound a hydrophobic pocket within the GSDMD C-terminal domain distal to its N-terminal domain. This "exosite" interface endows an additional function for the GSDMD C-terminal domain as a caspase-recruitment module besides its role in autoinhibition. Our study thus reveals dual-interface engagement of GSDMD by caspase-1, which may be applicable to other physiological substrates of caspases.


Subject(s)
Caspase 1/metabolism , Catalytic Domain/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis/immunology , Animals , Cell Line , Crystallography, X-Ray , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Inflammasomes/immunology , Mice , Protein Binding/physiology , Protein Conformation, beta-Strand/physiology , THP-1 Cells
5.
Immunity ; 51(1): 43-49.e4, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31097341

ABSTRACT

Gasdermin D (GSDMD) is an effector molecule for pyroptosis downstream of canonical and noncanonical inflammasome signaling pathways. Cleavage of GSDMD by inflammatory caspases triggers the oligomerization and lipid binding by its N-terminal domain, which assembles membrane pores, whereas its C-terminal domain binds the N-terminal domain to inhibit pyroptosis. Despite recent progress in our understanding of the structure and function of the murine gasdermin A3 (mGSDMA3), the molecular mechanisms of GSDMD activation and regulation remain poorly characterized. Here, we report the crystal structures of the full-length murine and human GSDMDs, which reveal the architecture of the GSDMD N-terminal domains and demonstrate distinct and common features of autoinhibition among gasdermin family members utilizing their ß1-ß2 loops. Disruption of the intramolecular domain interface enhanced pyroptosis, whereas mutations at the predicted lipid-binding or oligomerization surface reduced cytolysis. Our study provides a framework for understanding the autoinhibition, lipid binding, and oligomerization of GSDMD by using overlapping interfaces.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Crystallization/methods , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Lipid Metabolism , Lipids/chemistry , Mice , Mutagenesis, Site-Directed , Mutation/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Phosphate-Binding Proteins , Protein Conformation , Protein Domains/genetics , Protein Multimerization , Pyroptosis/genetics , Structure-Activity Relationship
6.
Semin Immunol ; 70: 101845, 2023 11.
Article in English | MEDLINE | ID: mdl-37806032

ABSTRACT

The gasdermin family of proteins are central effectors of the inflammatory, lytic cell death modality known as pyroptosis. Characterized in 2015, the most well-studied member gasdermin D can be proteolyzed, typically by caspases, to generate an active pore-forming N-terminal domain. At least well-studied three pharmacological inhibitors (necrosulfonamide, disulfiram, dimethyl fumarate) since 2018 have been shown to affect gasdermin D activity either through modulation of processing or interference with pore formation. A multitude of murine in vivo studies have since followed. Here, we discuss the current state of research surrounding these three inhibitors, caveats to their use, and a set of guiding principles that researchers should consider when pursuing further studies of gasdermin D inhibition.


Subject(s)
Gasdermins , Animals , Humans , Mice , Caspases/metabolism , Gasdermins/chemistry , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Pyroptosis
7.
J Biol Chem ; 300(6): 107386, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38763335

ABSTRACT

Inflammasomes serve as critical sensors for disruptions to cellular homeostasis, with inflammasome assembly leading to inflammatory caspase activation, gasdermin cleavage, and cytokine release. While the canonical pathways leading to priming, assembly, and pyroptosis are well characterized, recent work has begun to focus on the role of post-translational modifications (PTMs) in regulating inflammasome activity. A diverse array of PTMs, including phosphorylation, ubiquitination, SUMOylation, acetylation, and glycosylation, exert both activating and inhibitory influences on members of the inflammasome cascade through effects on protein-protein interactions, stability, and localization. Dysregulation of inflammasome activation is associated with a number of inflammatory diseases, and evidence is emerging that aberrant modification of inflammasome components contributes to this dysregulation. This review provides insight into PTMs within the NLRP3 inflammasome pathway and their functional consequences on the signaling cascade and highlights outstanding questions that remain regarding the complex web of signals at play.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Processing, Post-Translational , Signal Transduction , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Animals , Acetylation
8.
J Biol Chem ; 299(2): 102908, 2023 02.
Article in English | MEDLINE | ID: mdl-36642180

ABSTRACT

When activated, gasdermin family members are thought to be pore-forming proteins that cause lytic cell death. Despite this, numerous studies have suggested that the threshold for lytic cell death is dependent on which gasdermin family member is activated. Determination of the propensity of various gasdermin family members to cause pyroptosis has been handicapped by the fact that for many of them, the mechanisms and timing of their activation are uncertain. In this article, we exploit the recently discovered exosite-mediated recognition of gasdermin D (GSDMD) by the inflammatory caspases to develop a system that activates gasdermin family members in an efficient and equivalent manner. We leverage this system to show that upon activation, GSDMD and gasdermin A (GSDMA) exhibit differential subcellular localization, differential plasma membrane permeabilization, and differential lytic cell death. While GSDMD localizes rapidly to both the plasma membrane and organelle membranes, GSDMA preferentially localizes to the mitochondria with delayed and diminished accumulation at the plasma membrane. As a consequence of this differential kinetics of subcellular localization, N-terminal GSDMA results in early mitochondrial dysfunction relative to plasma membrane permeabilization. This study thus challenges the assumption that gasdermin family members effect cell death through identical mechanisms and establishes that their activation in their respective tissues of expression likely results in different immunological outcomes.


Subject(s)
Gasdermins , Pyroptosis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Membranes/metabolism , Phosphate-Binding Proteins/metabolism , Cell Membrane/metabolism , Inflammasomes/metabolism , Protein Engineering
9.
Nat Immunol ; 12(9): 834-43, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21765415

ABSTRACT

In response to stimulation with proinflammatory cytokines, the deubiquitinase A20 inducibly interacts with the regulatory molecules TAX1BP1, Itch and RNF11 to form the A20 ubiquitin-editing complex. However, the molecular signal that coordinates the assembly of this complex has remained elusive. Here we demonstrate that TAX1BP1 was inducibly phosphorylated on Ser593 and Ser624 in response to proinflammatory stimuli. The kinase IKKα, but not IKKß, was required for phosphorylation of TAX1BP1 and directly phosphorylated TAX1BP1 in response to stimulation with tumor necrosis factor (TNF) or interleukin 1 (IL-1). TAX1BP1 phosphorylation was pivotal for cytokine-dependent interactions among TAX1BP1, A20, Itch and RNF11 and downregulation of signaling by the transcription factor NF-κB. IKKα therefore serves a key role in the negative feedback of NF-κB canonical signaling by orchestrating assembly of the A20 ubiquitin-editing complex to limit inflammatory gene activation.


Subject(s)
Carrier Proteins/immunology , Cysteine Endopeptidases/immunology , I-kappa B Kinase/immunology , Immunity, Innate , Intracellular Signaling Peptides and Proteins/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Phosphorylation/drug effects , Recombinant Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Antibodies, Phospho-Specific/immunology , Antibodies, Phospho-Specific/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cloning, Molecular , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins , Escherichia coli , Fibroblasts/drug effects , Fibroblasts/immunology , Gene Deletion , Gene Expression Regulation , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Kinase/pharmacology , Interleukin-1/immunology , Interleukin-1/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
10.
J Biol Chem ; 295(10): 3228-3238, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31988247

ABSTRACT

Exomic studies have demonstrated that innate immune genes exhibit an even higher degree of variation than the majority of other gene families. However, the phenotypic implications of this genetic variation are not well understood, with effects ranging from hypomorphic to silent to hyperfunctioning. In this work, we study the functional consequences of this variation by investigating polymorphisms in gasdermin D, the key pyroptotic effector protein. We find that, although SNPs affecting potential posttranslational modifications did not affect gasdermin D function or pyroptosis, polymorphisms disrupting sites predicted to be structurally important dramatically alter gasdermin D function. The manner in which these polymorphisms alter function varies from conserving normal pyroptotic function to inhibiting caspase cleavage to disrupting oligomerization and pore formation. Further, downstream of inflammasome activation, polymorphisms that cause loss of gasdermin D function convert inflammatory pyroptotic cell death into immunologically silent apoptotic cell death. These findings suggest that human genetic variation can alter mechanisms of cell death in inflammation.


Subject(s)
Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Phosphate-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Apoptosis/drug effects , Caspase 3/metabolism , HEK293 Cells , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Phosphorylation , Propidium/pharmacology , Protein Multimerization , Protein Processing, Post-Translational , Ubiquitination
11.
Proc Natl Acad Sci U S A ; 115(26): 6792-6797, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891674

ABSTRACT

The inflammasomes are signaling platforms that promote the activation of inflammatory caspases such as caspases-1, -4, -5, and -11. Recent studies identified gasdermin D (GSDMD) as an effector for pyroptosis downstream of the inflammasome signaling pathways. Cleavage of GSDMD by inflammatory caspases allows its N-terminal domain to associate with membrane lipids and form pores that induce pyroptotic cell death. Despite the important role of GSDMD in pyroptosis, the molecular mechanisms of GSDMD recognition and cleavage by inflammatory caspases that trigger pyroptosis are poorly understood. Here, we demonstrate that the catalytic domains of inflammatory caspases can directly bind to both the full-length GSDMD and its cleavage site peptide, FLTD. A GSDMD-derived inhibitor, N-acetyl-Phe-Leu-Thr-Asp-chloromethylketone (Ac-FLTD-CMK), inhibits GSDMD cleavage by caspases-1, -4, -5, and -11 in vitro, suppresses pyroptosis downstream of both canonical and noncanonical inflammasomes, as well as reduces IL-1ß release following activation of the NLRP3 inflammasome in macrophages. By contrast, the inhibitor does not target caspase-3 or apoptotic cell death, suggesting that Ac-FLTD-CMK is a specific inhibitor for inflammatory caspases. Crystal structure of caspase-1 in complex with Ac-FLTD-CMK reveals extensive enzyme-inhibitor interactions involving both hydrogen bonds and hydrophobic contacts. Comparison with other caspase-1 structures demonstrates drastic conformational changes at the four active-site loops that assemble the catalytic groove. The present study not only contributes to our understanding of GSDMD recognition by inflammatory caspases but also reports a specific inhibitor for these caspases that can serve as a tool for investigating inflammasome signaling.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Caspase Inhibitors/chemistry , Neoplasm Proteins/chemistry , Peptides/chemistry , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 3/chemistry , Caspase 3/metabolism , Caspase Inhibitors/metabolism , Catalytic Domain , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Mice , Neoplasm Proteins/metabolism , Peptides/metabolism , Phosphate-Binding Proteins , Protein Structure, Secondary , RAW 264.7 Cells , THP-1 Cells
12.
Proc Natl Acad Sci U S A ; 115(40): E9362-E9370, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30224451

ABSTRACT

Defective and/or delayed wound healing has been implicated in the pathogenesis of several chronic inflammatory disorders, including inflammatory bowel disease (IBD). The resolution of inflammation is particularly important in mucosal organs, such as the gut, where restoration of epithelial barrier function is critical to reestablish homeostasis with the interfacing microenvironment. Although IL-33 and its receptor ST2/ILRL1 are known to be increased and associated with IBD, studies using animal models of colitis to address the mechanism have yielded ambiguous results, suggesting both pathogenic and protective functions. Unlike those previously published studies, we focused on the functional role of IL-33/ST2 during an extended (2-wk) recovery period after initial challenge in dextran sodium sulfate (DSS)-induced colitic mice. Our results show that during acute, resolving colitis the normal function of endogenous IL-33 is protection, and the lack of either IL-33 or ST2 impedes the overall recovery process, while exogenous IL-33 administration during recovery dramatically accelerates epithelial restitution and repair, with concomitant improvement of colonic inflammation. Mechanistically, we show that IL-33 stimulates the expression of a network of microRNAs (miRs) in the Caco2 colonic intestinal epithelial cell (IEC) line, especially miR-320, which is increased by >16-fold in IECs isolated from IL-33-treated vs. vehicle-treated DSS colitic mice. Finally, IL-33-dependent in vitro proliferation and wound closure of Caco-2 IECs is significantly abrogated after specific inhibition of miR-320A. Together, our data indicate that during acute, resolving colitis, IL-33/ST2 plays a crucial role in gut mucosal healing by inducing epithelial-derived miR-320 that promotes epithelial repair/restitution and the resolution of inflammation.


Subject(s)
Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Interleukin-33/metabolism , Intestinal Mucosa/physiology , MicroRNAs/metabolism , Regeneration , Acute Disease , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Mice , Mice, Knockout , MicroRNAs/genetics
13.
J Biol Chem ; 293(3): 1100-1105, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29212706

ABSTRACT

Heightened and extended inflammation underlies the pathogenesis of many disorders, including inflammatory bowel disease, sepsis, and inflammatory arthritis. Ubiquitin networks help dictate the strength and duration of inflammatory signaling. In innate immunity, the itchy E3 ubiquitin protein ligase (ITCH)-A20 ubiquitin-editing complex inhibits receptor-interacting Ser/Thr kinase (RIPK) activation by removing Lys-63-linked polyubiquitinated chains from key proteins in the nuclear factor kappa B (NF-κB) signaling pathway. The complex then attaches polyubiquitinated chains to these proteins to target them for lysosomal or proteasomal destruction. ITCH is phosphorylated and thereby inhibited by inhibitor of nuclear factor kappa B kinase subunit beta (IKKß) to fine-tune the inflammatory response to the strength of the offending signal. However, the biochemical mechanism by which E3 ubiquitination is impaired by IKK-driven phosphorylation remains unclear. Here, we report that this phosphorylation impedes ITCH binding to its cognate E2 ubiquitin ligase, UbcH7. Using CRISPR-Cas9 genetic knockout to mimic the ITCH-UbcH7-inhibited state, we further show that genetic UbcH7 deficiency phenocopies ITCH phosphorylation in regulating RIPK2 ubiquitination. We conclude that phosphorylation can disrupt the binding of an E3 ubiquitin ligase to an E2-conjugating enzyme, leading to prolonged inflammatory signaling. To our knowledge, this is the first report of E3 ubiquitin ligase phosphorylation inhibiting E3 ligase activity by impairing E2-E3 complex formation.


Subject(s)
Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Inflammation/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Ubiquitination/physiology
14.
J Biol Chem ; 292(23): 9666-9679, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28404814

ABSTRACT

The X-linked inhibitor of apoptosis (XIAP) protein has been identified as a key genetic driver of two distinct inflammatory disorders, X-linked lymphoproliferative syndrome 2 (XLP-2) and very-early-onset inflammatory bowel disease (VEO-IBD). Molecularly, the role of XIAP mutations in the pathogenesis of these disorders is unclear. Recent work has consistently shown XIAP to be critical for signaling downstream of the Crohn's disease susceptibility protein nucleotide-binding oligomerization domain-containing 2 (NOD2); however, the reported effects of XLP-2 and VEO-IBD XIAP mutations on cell death have been inconsistent. In this manuscript, we describe a CRISPR-mediated genetic system for cells of the myeloid lineage in which XIAP alleles can be replaced with disease-associated XIAP variants expressed at endogenous levels to simultaneously study inflammation-related cell death and NOD2 signaling. We show that, consistent with previous studies, NOD2 signaling is critically dependent on the BIR2 domain of XIAP. We further used this system to reconcile the aforementioned inconsistent XIAP cell death data to show that XLP-2 and VEO-IBD XIAP mutations that exhibit a loss-of-function NOD2 phenotype also lower the threshold for inflammatory cell death. Last, we identified and studied three novel patient XIAP mutations and used this system to characterize NOD2 and cell death phenotypes driven by XIAP. The results of this work support the role of XIAP in mediating NOD2 signaling while reconciling the role of XLP-2 and VEO-IBD XIAP mutations in inflammatory cell death and provide a set of tools and framework to rapidly test newly discovered XIAP variants.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Lymphoproliferative Disorders/metabolism , Mutation , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , X-Linked Inhibitor of Apoptosis Protein/metabolism , Cell Line , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Nod2 Signaling Adaptor Protein/genetics , Protein Domains , X-Linked Inhibitor of Apoptosis Protein/genetics
15.
J Biol Chem ; 292(35): 14649-14658, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28726636

ABSTRACT

Pyroptosis is a form of cell death important in defenses against pathogens that can also result in a potent and sometimes pathological inflammatory response. During pyroptosis, GSDMD (gasdermin D), the pore-forming effector protein, is cleaved, forms oligomers, and inserts into the membranes of the cell, resulting in rapid cell death. However, the potent cell death induction caused by GSDMD has complicated our ability to understand the biology of this protein. Studies aimed at visualizing GSDMD have relied on expression of GSDMD fragments in epithelial cell lines that naturally lack GSDMD expression and also lack the proteases necessary to cleave GSDMD. In this work, we performed mutagenesis and molecular modeling to strategically place tags and fluorescent proteins within GSDMD that support native pyroptosis and facilitate live-cell imaging of pyroptotic cell death. Here, we demonstrate that these fusion proteins are cleaved by caspases-1 and -11 at Asp-276. Mutations that disrupted the predicted p30-p20 autoinhibitory interface resulted in GSDMD aggregation, supporting the oligomerizing activity of these mutations. Furthermore, we show that these novel GSDMD fusions execute inflammasome-dependent pyroptotic cell death in response to multiple stimuli and allow for visualization of the morphological changes associated with pyroptotic cell death in real time. This work therefore provides new tools that not only expand the molecular understanding of pyroptosis but also enable its direct visualization.


Subject(s)
Caspase 1/metabolism , Caspases, Initiator/metabolism , Caspases/metabolism , Inflammasomes/metabolism , Macrophages/cytology , Models, Biological , Neoplasm Proteins/metabolism , Pyroptosis , Amino Acid Substitution , Animals , Cell Line, Transformed , HEK293 Cells , Humans , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Microscopy, Video , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphate-Binding Proteins , Point Mutation , Protein Multimerization , Protein Transport , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
16.
J Immunol ; 196(10): 4291-7, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27045108

ABSTRACT

The RIP kinases (RIPKs) play an essential role in inflammatory signaling and inflammatory cell death. However, the function of their kinase activity has been enigmatic, and only recently has kinase domain activity been shown to be crucial for their signal transduction capacity. Despite this uncertainty, the RIPKs have been the subject of intense pharmaceutical development with a number of compounds currently in preclinical testing. In this work, we seek to determine the functional redundancy between the kinase domains of the four major RIPK family members. We find that although RIPK1, RIPK2, and RIPK4 are similar in that they can all activate NF-κB and induce NF-κB essential modulator ubiquitination, only RIPK2 is a dual-specificity kinase. Domain swapping experiments showed that the RIPK4 kinase domain could be converted to a dual-specificity kinase and is essentially indistinct from RIPK2 in biochemical and molecular activity. Surprisingly, however, replacement of RIPK2's kinase domain with RIPK4's did not complement a nucleotide-binding oligomerization domain 2 signaling or gene expression induction defect in RIPK2(-/-) macrophages. These findings suggest that RIPK2's kinase domain is functionally unique compared with other RIPK family members and that pharmacologic targeting of RIPK2 can be separated from the other RIPKs.


Subject(s)
Cell Death , Immunity, Innate , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , Synthetic Biology , Gene Expression , HEK293 Cells , Humans , Inflammation , Macrophages/metabolism , NF-kappa B p50 Subunit/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Protein Domains , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitination
17.
J Immunol ; 197(4): 1353-67, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27385778

ABSTRACT

Canonical inflammasome activation induces a caspase-1/gasdermin D (Gsdmd)-dependent lytic cell death called pyroptosis that promotes antimicrobial host defense but may contribute to sepsis. The nature of the caspase-1-dependent change in plasma membrane (PM) permeability during pyroptotic progression remains incompletely defined. We assayed propidium(2+) (Pro(2+)) influx kinetics during NLRP3 or Pyrin inflammasome activation in murine bone marrow-derived macrophages (BMDMs) as an indicator of this PM permeabilization. BMDMs were characterized by rapid Pro(2+) influx after initiation of NLRP3 or Pyrin inflammasomes by nigericin (NG) or Clostridium difficile toxin B (TcdB), respectively. No Pro(2+) uptake in response to NG or TcdB was observed in Casp1(-/-) or Asc(-/-) BMDMs. The cytoprotectant glycine profoundly suppressed NG and TcdB-induced lysis but not Pro(2+) influx. The absence of Gsdmd expression resulted in suppression of NG-stimulated Pro(2+) influx and pyroptotic lysis. Extracellular La(3+) and Gd(3+) rapidly and reversibly blocked the induced Pro(2+) influx and markedly delayed pyroptotic lysis without limiting upstream inflammasome assembly and caspase-1 activation. Thus, caspase-1-driven pyroptosis requires induction of initial prelytic pores in the PM that are dependent on Gsdmd expression. These PM pores also facilitated the efflux of cytosolic ATP and influx of extracellular Ca(2+) Although lanthanides and Gsdmd deletion both suppressed PM pore activity and pyroptotic lysis, robust IL-1ß release was observed in lanthanide-treated BMDMs but not in Gsdmd-deficient cells. This suggests roles for Gsdmd in both passive IL-1ß release secondary to pyroptotic lysis and in nonlytic/nonclassical IL-1ß export.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Pyroptosis/physiology , Animals , Cell Membrane/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Lanthanoid Series Elements/pharmacology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins
18.
Genes Dev ; 24(23): 2666-77, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123652

ABSTRACT

Upon intracellular bacterial exposure, the Crohn's disease and sarcoidosis susceptibility protein NOD2 (nucleotide oligomerization domain protein 2) binds to the protein kinase RIP2 (receptor-interacting protein 2) to coordinate NF-κB (nuclear factor κ B)-mediated cytokine responses. While RIP2 clearly has kinase activity, the function of its kinase domain has been enigmatic. Although originally classified as a serine-threonine kinase based on homology scans, we find that RIP2 also has tyrosine kinase activity. RIP2 undergoes autophosphorylation on Tyr 474 (Y474). This phosphorylation event is necessary for effective NOD2 signaling and does not occur in the presence of the most common Crohn's disease-associated NOD2 allele. Given this tyrosine kinase activity, a small-molecule inhibitor screen designed to identify pharmacologic agents that inhibit RIP2's tyrosine kinase activity was performed. At nanomolar concentrations, the EGFR (epidermal growth factor receptor) tyrosine kinase inhibitors gefitinib (Iressa) and erlotinib (Tarceva) were found to inhibit both RIP2 tyrosine phosphorylation and MDP (muramyl dipeptide)-induced cytokine release in a variety of NOD2 hyperactivation states. This effect is specific for RIP2 and does not depend on EGFR. The finding that RIP2 has tyrosine kinase activity and the finding that gefitinib and erlotinib, two agents already used clinically for cancer chemotherapy, can inhibit this activity suggest that RIP2's tyrosine kinase activity could be targeted specifically in the treatment of inflammatory diseases.


Subject(s)
Cytokines/metabolism , Enzyme Activation/drug effects , Nod2 Signaling Adaptor Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Animals , Cells, Cultured , Erlotinib Hydrochloride , Gefitinib , Gene Knockout Techniques , HEK293 Cells , HT29 Cells , Humans , Mice , Phosphorylation/drug effects , Quinazolines/pharmacology , Tyrosine/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
19.
Am J Pathol ; 186(4): 885-98, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26908008

ABSTRACT

Although a clear association has been established between IL-33 and inflammatory bowel disease, mechanistic studies to date, primarily using acute murine models of colitis, have yielded contradicting results, demonstrating both pathogenic and protective roles. We used a well-characterized, spontaneous model of inflammatory bowel disease [ie, SAMP1/YitFc (SAMP) mice] to investigate the role of IL-33 during chronic intestinal inflammation. Our results showed marked eosinophil infiltration into the gut mucosa with increased levels of eotaxins and type 2 helper T-cell (Th2) cytokines as disease progressed and became more severe, which could be reversed upon either eosinophil depletion or blockade of IL-33 signaling. Exogenous IL-33 administration recapitulated these effects in ilea of uninflamed (parental) control AKR/J mice. Human data supported these findings, showing colocalization and up-regulation of IL-33 and eosinophils in the colonic mucosa of inflammatory bowel disease patients versus noninflamed controls. Finally, colonization of commensal flora by fecal material transplantation into germ-free SAMP and the presence of the gut microbiome induced IL-33, subsequent eosinophil infiltration, and mounting of Th2 immune responses, leading to exacerbation of chronic intestinal inflammation characteristic of SAMP mice. These data demonstrate a pathogenic role for IL-33-mediated eosinophilia and activation of Th2 immunity in chronic intestinal inflammation that is dependent on the gut microbiome. Targeting IL-33 may represent a novel therapeutic approach to treat patients with inflammatory bowel disease.


Subject(s)
Eosinophils/cytology , Ileitis/pathology , Interleukin-33/metabolism , Th2 Cells/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Ileitis/immunology , Inflammation/immunology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Mice , Up-Regulation
20.
Mol Cell ; 34(4): 461-72, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19481526

ABSTRACT

The noncanonical IKK family member IKKepsilon is essential for regulating antiviral signaling pathways and is a recently discovered breast cancer oncoprotein. Although several IKKepsilon targets have been described, direct IKKepsilon substrates necessary for regulating cell transformation have not been identified. Here, we performed a screen for putative IKKepsilon substrates using an unbiased proteomic and bioinformatic approach. Using a positional scanning peptide library assay, we determined the optimal phosphorylation motif for IKKepsilon and used bioinformatic approaches to predict IKKepsilon substrates. Of these potential substrates, serine 418 of the tumor suppressor CYLD was identified as a likely site of IKKepsilon phosphorylation. We confirmed that CYLD is directly phosphorylated by IKKepsilon and that IKKepsilon phosphorylates serine 418 in vivo. Phosphorylation of CYLD at serine 418 decreases its deubiquitinase activity and is necessary for IKKepsilon-driven transformation. Together, these observations define IKKepsilon and CYLD as an oncogene-tumor suppressor network that participates in tumorigenesis.


Subject(s)
Breast Neoplasms , Cell Transformation, Neoplastic , I-kappa B Kinase/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Deubiquitinating Enzyme CYLD , Female , Genes, Reporter , Humans , I-kappa B Kinase/genetics , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Sequence Alignment , Serine/metabolism , Substrate Specificity , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL