Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters

Publication year range
1.
Immunity ; 49(1): 66-79.e5, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29980436

ABSTRACT

Genetic mutations of CARD14 (encoding CARMA2) are observed in psoriasis patients. Here we showed that Card14E138A/+ and Card14ΔQ136/+ mice developed spontaneous psoriasis-like skin inflammation, which resulted from constitutively activated CARMA2 via self-aggregation leading to the enhanced activation of the IL-23-IL-17A cytokine axis. Card14-/- mice displayed attenuated skin inflammation in the imiquimod-induced psoriasis model due to impaired IL-17A signaling in keratinocytes. CARMA2, mainly expressed in keratinocytes, associates with the ACT1-TRAF6 signaling complex and mediates IL-17A-induced NF-κB and MAPK signaling pathway activation, which leads to expression of pro-inflammatory factors. Thus, CARMA2 serves as a key mediator of IL-17A signaling and its constitutive activation in keratinocytes leads to the onset of psoriasis, which indicates an important role of NF-κB activation in keratinocytes in psoriatic initiation.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Dermatitis/genetics , Gain of Function Mutation , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Interleukin-17/metabolism , Keratinocytes/metabolism , Psoriasis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/deficiency , Cell Line , Cytokines/genetics , Cytokines/metabolism , Dermatitis/physiopathology , Gene Expression Regulation/drug effects , Guanylate Kinases/chemistry , Guanylate Kinases/deficiency , HEK293 Cells , Humans , Imiquimod , Keratinocytes/pathology , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Psoriasis/chemically induced , Psoriasis/physiopathology , Signal Transduction , T-Lymphocyte Subsets/metabolism , TNF Receptor-Associated Factor 6/metabolism
2.
Mol Cell ; 67(5): 733-743.e4, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28844863

ABSTRACT

Nuclear receptors recruit multiple coactivators sequentially to activate transcription. This "ordered" recruitment allows different coactivator activities to engage the nuclear receptor complex at different steps of transcription. Estrogen receptor (ER) recruits steroid receptor coactivator-3 (SRC-3) primary coactivator and secondary coactivators, p300/CBP and CARM1. CARM1 recruitment lags behind the binding of SRC-3 and p300 to ER. Combining cryo-electron microscopy (cryo-EM) structure analysis and biochemical approaches, we demonstrate that there is a close crosstalk between early- and late-recruited coactivators. The sequential recruitment of CARM1 not only adds a protein arginine methyltransferase activity to the ER-coactivator complex, it also alters the structural organization of the pre-existing ERE/ERα/SRC-3/p300 complex. It induces a p300 conformational change and significantly increases p300 HAT activity on histone H3K18 residues, which, in turn, promotes CARM1 methylation activity on H3R17 residues to enhance transcriptional activity. This study reveals a structural role for a coactivator sequential recruitment and biochemical process in ER-mediated transcription.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Estrogen Receptor alpha/metabolism , Guanylate Cyclase/metabolism , Nuclear Receptor Coactivator 3/metabolism , Transcription, Genetic , Acetylation , Binding Sites , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/chemistry , E1A-Associated p300 Protein/genetics , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Guanylate Cyclase/chemistry , Guanylate Cyclase/genetics , HEK293 Cells , HeLa Cells , Histones/chemistry , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Models, Molecular , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Receptor Coactivator 3/chemistry , Nuclear Receptor Coactivator 3/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship , Time Factors , Transcription Factors , Transcriptional Activation , Transfection
3.
Immunity ; 43(4): 715-26, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26488816

ABSTRACT

CARD9 is a central component of anti-fungal innate immune signaling via C-type lectin receptors, and several immune-related disorders are associated with CARD9 alterations. Here, we used a rare CARD9 variant that confers protection against inflammatory bowel disease as an entry point to investigating CARD9 regulation. We showed that the protective variant of CARD9, which is C-terminally truncated, acted in a dominant-negative manner for CARD9-mediated cytokine production, indicating an important role for the C terminus in CARD9 signaling. We identified TRIM62 as a CARD9 binding partner and showed that TRIM62 facilitated K27-linked poly-ubiquitination of CARD9. We identified K125 as the ubiquitinated residue on CARD9 and demonstrated that this ubiquitination was essential for CARD9 activity. Furthermore, we showed that similar to Card9-deficient mice, Trim62-deficient mice had increased susceptibility to fungal infection. In this study, we utilized a rare protective allele to uncover a TRIM62-mediated mechanism for regulation of CARD9 activation.


Subject(s)
CARD Signaling Adaptor Proteins/physiology , Candidiasis, Invasive/immunology , Receptors, Angiotensin/physiology , Receptors, Endothelin/physiology , Ubiquitin-Protein Ligases/physiology , Adjuvants, Immunologic/pharmacology , Animals , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Candidiasis, Invasive/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/prevention & control , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genes, Dominant , Genetic Predisposition to Disease , HEK293 Cells , HeLa Cells , Humans , Inflammatory Bowel Diseases/genetics , Mice , Mice, 129 Strain , Mice, Knockout , Protein Interaction Mapping , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptors, Angiotensin/chemistry , Receptors, Angiotensin/deficiency , Receptors, Endothelin/chemistry , Receptors, Endothelin/deficiency , Recombinant Fusion Proteins/metabolism , Signal Transduction , Specific Pathogen-Free Organisms , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
4.
Nature ; 552(7685): 355-361, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29293211

ABSTRACT

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , CARD Signaling Adaptor Proteins/metabolism , Microglia/metabolism , Protein Aggregation, Pathological , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antibodies/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/immunology , Female , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Presenilin-1/deficiency , Presenilin-1/genetics , Protein Domains , Spatial Memory/physiology
5.
Int J Mol Sci ; 24(3)2023 Jan 22.
Article in English | MEDLINE | ID: mdl-36768499

ABSTRACT

ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)) is the only adaptor involved in the formation of multiple types of inflammasomes. Accumulating evidence demonstrates that ASC plays a critical role in the protection of the host against pathogen infection. In this study, we identified an ASC gene in the large yellow croaker (Larimichthys crocea), namely LcASC, and then investigated the expression characteristics and related signal pathways. On one hand, LcASC has several conserved protein modules, i.e., an N-terminal PYD region, a C-terminal CARD region, and twelve α-helix structures. On the other hand, it has a high variable linker between PYD and CARD domains. Moreover, LcASC has varying degrees of expression in different tissues, among which the highest expression is observed in the spleen followed by the gills and skin. It also shows induced expressions in the head kidney, liver, and spleen following immune stimulation, especially Vibrio Parahaemolyticus infection. Further subcellular localization analysis showed that LcASC formed a clear aggregated speck in the cytoplasm close to the nucleus. In addition, we found 46 DEGs in a comparative transcriptome analysis between the LcASC overexpression group and the control vector group. Notedly, the up-regulated gene Fos and down-regulated gene DOK3 in LcASC overexpressed cells play important roles in the immune system. How ASC contacts these two genes needs to be clarified in upcoming studies. These findings collectively provide new insights into finfish ASC and its potential regulatory signaling pathway as well.


Subject(s)
Inflammasomes , Perciformes , Animals , Inflammasomes/metabolism , Caspase Activation and Recruitment Domain , Apoptosis , CARD Signaling Adaptor Proteins/chemistry , Perciformes/genetics , Perciformes/metabolism , Signal Transduction
6.
J Biol Chem ; 296: 100597, 2021.
Article in English | MEDLINE | ID: mdl-33781745

ABSTRACT

Inflammasomes are macromolecular complexes involved in the host response to external and endogenous danger signals. Inflammasome-mediated sterile inflammation plays a central role in several human conditions such as autoimmune diseases, type-2 diabetes, and neurodegenerative disorders, indicating inflammasomes could be appealing therapeutic targets. Previous work has demonstrated that inhibiting the ATPase activity of the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3), disrupts inflammasome assembly and function. However, there is a necessity to find new potent compounds with therapeutic potential. Here we combine computational modeling of the target and virtual screening to discover a group of novel compounds predicted to inhibit NLRP3. We characterized the best compounds and determined their potency, specificity, and ability to inhibit processes downstream from NLRP3 activation. Moreover, we analyzed in mice the competence of a lead candidate to reduce lipopolysaccharide-induced inflammation. We also validated the active pharmacophore shared among all the NLRP3 inhibitors, and through computational docking, we clarify key structural features for compound positioning within the inflammasome ATP-binding site. Our study sets the basis for rational design and optimization of inflammasome-targeting probes and drugs.


Subject(s)
CARD Signaling Adaptor Proteins/antagonists & inhibitors , Calcium-Binding Proteins/antagonists & inhibitors , Drug Discovery , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , CARD Signaling Adaptor Proteins/chemistry , Calcium-Binding Proteins/chemistry , Drug Evaluation, Preclinical , Humans , Inflammasomes/chemistry , Mice , Models, Molecular , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , Protein Domains , User-Computer Interface
7.
Mol Cell ; 51(6): 766-79, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074955

ABSTRACT

The CARMA1/Bcl10/MALT1 (CBM) signalosome mediates antigen receptor-induced NF-κB signaling to regulate multiple lymphocyte functions. While CARMA1 and Bcl10 contain caspase recruitment domains (CARDs), MALT1 is a paracaspase with structural similarity to caspases. Here we show that the reconstituted CBM signalosome is a helical filamentous assembly in which substoichiometric CARMA1 nucleates Bcl10 filaments. Bcl10 filament formation is a highly cooperative process whose threshold is sensitized by oligomerized CARMA1 upon receptor activation. In cells, both cotransfected CARMA1/Bcl10 complex and the endogenous CBM signalosome are filamentous morphologically. Combining crystallography, nuclear magnetic resonance, and electron microscopy, we reveal the structure of the Bcl10 CARD filament and the mode of interaction between CARMA1 and Bcl10. Structure-guided mutagenesis confirmed the observed interfaces in Bcl10 filament assembly and MALT1 activation in vitro and NF-κB activation in cells. These data support a paradigm of nucleation-induced signal transduction with threshold response due to cooperativity and signal amplification by polymerization.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CARD Signaling Adaptor Proteins/genetics , Caspases/genetics , Guanylate Cyclase/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , Binding Sites , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Caspases/chemistry , Caspases/metabolism , Crystallography, X-Ray , Gene Expression Regulation , Guanylate Cyclase/chemistry , Guanylate Cyclase/metabolism , Humans , Jurkat Cells , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , NF-kappa B/chemistry , NF-kappa B/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Protein Binding , Protein Structure, Secondary
8.
Proc Natl Acad Sci U S A ; 115(43): 10845-10852, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30279182

ABSTRACT

Canonical inflammasomes are cytosolic supramolecular complexes that activate caspase-1 upon sensing extrinsic microbial invasions and intrinsic sterile stress signals. During inflammasome assembly, adaptor proteins ASC and NLRC4 recruit caspase-1 through homotypic caspase recruitment domain (CARD) interactions, leading to caspase-1 dimerization and activation. Activated caspase-1 processes proinflammatory cytokines and Gasdermin D to induce cytokine maturation and pyroptotic cell death. Here, we present cryo-electron microscopy (cryo-EM) structures of NLRC4 CARD and ASC CARD filaments mediated by conserved three types of asymmetric interactions (types I, II, and III). We find that the CARDs of these two adaptor proteins share a similar assembly pattern, which matches that of the caspase-1 CARD filament whose structure we defined previously. These data indicate a unified mechanism for downstream caspase-1 recruitment through CARD-CARD interactions by both adaptors. Using structure modeling, we further show that full-length NLRC4 assembles via two separate symmetries at its CARD and its nucleotide-binding domain (NBD), respectively.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , Calcium-Binding Proteins/chemistry , Caspase 1/chemistry , Cryoelectron Microscopy , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Enzyme Activation , Humans , Inflammasomes , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism
9.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33467177

ABSTRACT

The inflammasome is a three-component (sensor, adaptor, and effector) filamentous signaling platform that shields from multiple pathogenic infections by stimulating the proteolytical maturation of proinflammatory cytokines and pyroptotic cell death. The signaling process initiates with the detection of endogenous and/or external danger signals by specific sensors, followed by the nucleation and polymerization from sensor to downstream adaptor and then to the effector, caspase-1. Aberrant activation of inflammasomes promotes autoinflammatory diseases, cancer, neurodegeneration, and cardiometabolic disorders. Therefore, an equitable level of regulation is required to maintain the equilibrium between inflammasome activation and inhibition. Recent advancement in the structural and mechanistic understanding of inflammasome assembly potentiates the emergence of novel therapeutics against inflammasome-regulated diseases. In this review, we have comprehensively discussed the recent and updated insights into the structure of inflammasome components, their activation, interaction, mechanism of regulation, and finally, the formation of densely packed filamentous inflammasome complex that exists as micron-sized punctum in the cells and mediates the immune responses.


Subject(s)
DNA-Binding Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/chemistry , Caspase 1/metabolism , DNA-Binding Proteins/chemistry , Humans , Inflammasomes/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , Protein Domains , Protein Multimerization
10.
J Biol Chem ; 294(2): 439-452, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30459235

ABSTRACT

The inflammasome is a multiprotein complex necessary for the onset of inflammation. The adapter protein ASC assembles inflammasome components by acting as a molecular glue between danger-signal sensors and procaspase-1. The assembly is mediated by ASC self-association and protein interactions via its two Death Domains, PYD and CARD. Truncated versions of ASC have been shown to form filaments, but information on the filaments formed by full-length ASC is needed to construct a meaningful model of inflammasome assembly. To gain insights into this system, we used a combination of transmission EM, NMR, and computational analysis to investigate intact ASC structures. We show that ASC forms ∼6-7-nm-wide filaments that stack laterally to form bundles. The structural characteristics and dimensions of the bundles indicate that both PYD and CARD are integral parts of the filament. A truncated version of ASC with only the CARD domain (ASCCARD) forms different filaments (∼3-4-nm width), providing further evidence that both domains work in concert in filament assembly. Ring-shaped protein particles bound to pre-existing filaments match the size of ASC dimer structures generated by NMR-based protein docking, suggesting that the ASC dimer could be a basic building block for filament formation. Solution NMR binding studies identified the protein surfaces involved in the ASCCARD-ASCCARD interaction. These data provide new insights into the structural underpinnings of the inflammasome and should inform future efforts to interrogate this important biological system.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/ultrastructure , Caspase Activation and Recruitment Domain , Pyrin Domain , Apoptosis , CARD Signaling Adaptor Proteins/immunology , Humans , Hydrogen-Ion Concentration , Inflammasomes/immunology , Models, Molecular , Protein Conformation , Protein Conformation, alpha-Helical , Protein Multimerization
11.
J Biol Chem ; 294(40): 14648-14660, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31391255

ABSTRACT

The activation of key signaling pathways downstream of antigen receptor engagement is critically required for normal lymphocyte activation during the adaptive immune response. CARD11 is a multidomain signaling scaffold protein required for antigen receptor signaling to NF-κB, c-Jun N-terminal kinase, and mTOR. Germline mutations in the CARD11 gene result in at least four types of primary immunodeficiency, and somatic CARD11 gain-of-function mutations drive constitutive NF-κB activity in diffuse large B cell lymphoma and other lymphoid cancers. In response to antigen receptor triggering, CARD11 transitions from a closed, inactive state to an open, active scaffold that recruits multiple signaling partners into a complex to relay downstream signaling. However, how this signal-induced CARD11 conversion occurs remains poorly understood. Here we investigate the role of Inducible Element 1 (IE1), a short regulatory element in the CARD11 Inhibitory Domain, in the CARD11 signaling cycle. We find that IE1 controls the signal-dependent Opening Step that makes CARD11 accessible to the binding of cofactors, including Bcl10, MALT1, and the HOIP catalytic subunit of the linear ubiquitin chain assembly complex. Surprisingly, we find that IE1 is also required at an independent step for the maximal activation of HOIP and MALT1 enzymatic activity after cofactor recruitment to CARD11. This role of IE1 reveals that there is an Enzymatic Activation Step in the CARD11 signaling cycle that is distinct from the Cofactor Association Step. Our results indicate that CARD11 has evolved to actively coordinate scaffold opening and the induction of enzymatic activity among recruited cofactors during antigen receptor signaling.


Subject(s)
Adaptive Immunity/genetics , CARD Signaling Adaptor Proteins/chemistry , Guanylate Cyclase/chemistry , Multiprotein Complexes/chemistry , Receptors, Antigen/genetics , B-Cell CLL-Lymphoma 10 Protein/genetics , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/ultrastructure , Germ-Line Mutation/genetics , Guanylate Cyclase/genetics , Guanylate Cyclase/ultrastructure , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Jurkat Cells , Lymphocyte Activation/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , NF-kappa B/genetics , Protein Binding/genetics , Protein Conformation , Receptors, Antigen/chemistry , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics
12.
J Biol Chem ; 293(52): 20240-20248, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30385506

ABSTRACT

Inflammasomes are supramolecular signaling platforms integral to innate immune defense against invading pathogens. The NOD-like receptor (NLR) family apoptosis inhibitory protein (NAIP)·NLR family caspase-recruiting domain (CARD) domain-containing 4 (NLRC4) inflammasome recognizes intracellular bacteria and induces the polymerization of the caspase-1 protease, which in turn executes maturation of interleukin-1ß (IL-1ß) and pyroptosis. Several high-resolution structures of the fully assembled NAIP·NLRC4 complex are available, but these structures do not resolve the architecture of the CARD filament in atomic detail. Here, we present the cryo-EM structure of the filament assembled by the CARD of human NLRC4 (NLRC4CARD) at 3.4 Å resolution. The structure revealed that the helical architecture of the NLRC4CARD filament is essentially identical to that of the downstream filament assembled by the CARD of caspase-1 (casp1CARD), but deviates from the split washer-like assembly of the NAIP·NLRC4 oligomer. Our results suggest that architectural complementarity is a major driver for the recognition between upstream and downstream CARD assemblies in inflammasomes. Furthermore, a Monte Carlo simulation of the NLRC4CARD filament assembly rationalized why an (un)decameric NLRC4 oligomer is optimal for assembling the helical base of the NLRC4CARD filament. Together, our results explain how symmetric and asymmetric supramolecular assemblies enable high-fidelity signaling in inflammasomes.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , Calcium-Binding Proteins/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Neuronal Apoptosis-Inhibitory Protein/chemistry , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cryoelectron Microscopy , Humans , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Neuronal Apoptosis-Inhibitory Protein/metabolism , Protein Structure, Quaternary
13.
J Biol Chem ; 293(43): 16803-16817, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30206119

ABSTRACT

The caspase recruitment domain-containing protein 9 (CARD9)-B-cell lymphoma/leukemia 10 (Bcl10) signaling axis is activated in myeloid cells during the innate immune response to a variety of diverse pathogens. This signaling pathway requires a critical caspase recruitment domain (CARD)-CARD interaction between CARD9 and Bcl10 that promotes downstream activation of factors, including NF-κB and the mitogen-activated protein kinase (MAPK) p38. Despite these insights, CARD9 remains structurally uncharacterized, and little mechanistic understanding of its regulation exists. We unexpectedly found here that the CARD in CARD9 binds to Zn2+ with picomolar affinity-a concentration comparable with the levels of readily accessible Zn2+ in the cytosol. NMR solution structures of the CARD9-CARD in the apo and Zn2+-bound states revealed that Zn2+ has little effect on the ground-state structure of the CARD; yet the stability of the domain increased considerably upon Zn2+ binding, with a concomitant reduction in conformational flexibility. Moreover, Zn2+ binding inhibited polymerization of the CARD9-CARD into helical assemblies. Here, we also present a 20-Å resolution negative-stain EM (NS-EM) structure of these filamentous assemblies and show that they adopt a similar helical symmetry as reported previously for filaments of the Bcl10 CARD. Using both bulk assays and direct NS-EM visualization, we further show that the CARD9-CARD assemblies can directly template and thereby nucleate Bcl10 polymerization, a capacity considered critical to propagation of the CARD9-Bcl10 signaling cascade. Our findings indicate that CARD9 is a potential target of Zn2+-mediated signaling that affects Bcl10 polymerization in innate immune responses.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/metabolism , CARD Signaling Adaptor Proteins/metabolism , Zinc/metabolism , B-Cell CLL-Lymphoma 10 Protein/chemistry , B-Cell CLL-Lymphoma 10 Protein/genetics , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Crystallography, X-Ray , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Polymerization , Protein Binding , Protein Domains , Signal Transduction , Zinc/chemistry , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Int J Sports Med ; 40(10): 670-677, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31342477

ABSTRACT

Apoptosis-associated, speck-like protein containing a caspase recruitment domain (ASC) plays an important role in inflammatory cytokine synthesis in peripheral blood mononuclear cells (PBMCs), and the expression of ASC is suppressed by increased methylation of its CpG sites. The current study investigated the longitudinal association of replacing sedentary time with light-intensity physical activity (LPA) or moderate to vigorous-intensity physical activity (MVPA) on the ASC methylation in middle-aged people. We investigated 1 238 individuals who participated in baseline and 5-year follow-up surveys of a population-based cohort study. Sedentary, LPA and MVPA time were objectively measured using accelerometers. ASC methylation in PBMCs was measured by pyrosequencing. Using a multiple linear regression and employing an isotemporal substitution model, the longitudinal associations of changes in the sedentary time, LPA and MVPA on the changes in the ASC methylation were analyzed after adjusting for potential confounders. Substituting 60 min per day of LPA for sedentary time was associated with 1.17 times (95% confidence interval 1.07, 1.27) higher ASC methylation levels (mean of 7 CpG sites, P<0.001). However, such effects were not seen for MVPA. These results suggest that substituting LPA for sedentary time may be linked with increased (favorable) ASC methylation as a potential biomarker of systemic inflammation.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , DNA Methylation , Exercise , Accelerometry , Aged , Anthropometry , Cohort Studies , CpG Islands , Cytokines/blood , Female , Fitness Trackers , Humans , Leukocytes, Mononuclear , Male , Middle Aged , Sedentary Behavior
15.
J Allergy Clin Immunol ; 142(6): 1956-1967.e6, 2018 12.
Article in English | MEDLINE | ID: mdl-29778503

ABSTRACT

BACKGROUND: Monogenic autoinflammatory disorders are characterized by dysregulation of the innate immune system, for example by gain-of-function mutations in inflammasome-forming proteins, such as NOD-like receptor family CARD-containing 4 protein (NLRC4). OBJECTIVE: Here we investigate the mechanism by which a novel mutation in the leucine-rich repeat (LRR) domain of NLRC4 (c.G1965C, p.W655C) contributes to autoinflammatory disease. METHODS: We studied 2 unrelated patients with early-onset macrophage activation syndrome harboring the same de novo mutation in NLRC4. In vitro inflammasome complex formation was quantified by using flow cytometric analysis of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) specks. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 techniques and lentiviral transduction were used to generate THP-1 cells with either wild-type or mutant NLRC4 cDNA. Cell death and release of IL-1ß/IL-18 were quantified by using flow cytometry and ELISA, respectively. RESULTS: The p.W655C NLRC4 mutation caused increased ASC speck formation, caspase-1-dependent cell death, and IL-1ß/IL-18 production. ASC contributed to p.W655C NLRC4-mediated cytokine release but not cell death. Mutation of p.W655 activated the NLRC4 inflammasome complex by engaging with 2 interfaces on the opposing LRR domain of the oligomer. One key set of residues (p.D1010, p.D1011, p.L1012, and p.I1015) participated in LRR-LRR oligomerization when triggered by mutant NLRC4 or type 3 secretion system effector (PrgI) stimulation of the NLRC4 inflammasome complex. CONCLUSION: This is the first report of a mutation in the LRR domain of NLRC4 causing autoinflammatory disease. c.G1965C/p.W655C NLRC4 increased inflammasome activation in vitro. Data generated from various NLRC4 mutations provides evidence that the LRR-LRR interface has an important and previously unrecognized role in oligomerization of the NLRC4 inflammasome complex.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/genetics , Inflammasomes/genetics , Leucine , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/immunology , Female , HEK293 Cells , Humans , Infant , Infant, Newborn , Inflammasomes/chemistry , Inflammasomes/immunology , Macrophage Activation , Male , Protein Domains , Syndrome , THP-1 Cells
16.
Nature ; 490(7421): 539-42, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22885697

ABSTRACT

NLRC4 is a cytosolic member of the NOD-like receptor family that is expressed in innate immune cells. It senses indirectly bacterial flagellin and type III secretion systems, and responds by assembling an inflammasome complex that promotes caspase-1 activation and pyroptosis. Here we use knock-in mice expressing NLRC4 with a carboxy-terminal 3×Flag tag to identify phosphorylation of NLRC4 on a single, evolutionarily conserved residue, Ser 533, following infection of macrophages with Salmonella enterica serovar Typhimurium (also known as Salmonella typhimurium). Western blotting with a NLRC4 phospho-Ser 533 antibody confirmed that this post-translational modification occurs only in the presence of stimuli known to engage NLRC4 and not the related protein NLRP3 or AIM2. Nlrc4(-/-) macrophages reconstituted with NLRC4 mutant S533A, unlike those reconstituted with wild-type NLRC4, did not activate caspase-1 and pyroptosis in response to S. typhimurium, indicating that S533 phosphorylation is critical for NLRC4 inflammasome function. Conversely, phosphomimetic NLRC4 S533D caused rapid macrophage pyroptosis without infection. Biochemical purification of the NLRC4-phosphorylating activity and a screen of kinase inhibitors identified PRKCD (PKCδ) as a candidate NLRC4 kinase. Recombinant PKCδ phosphorylated NLRC4 S533 in vitro, immunodepletion of PKCδ from macrophage lysates blocked NLRC4 S533 phosphorylation in vitro, and Prkcd(-/-) macrophages exhibited greatly attenuated caspase-1 activation and IL-1ß secretion specifically in response to S. typhimurium. Phosphorylation-defective NLRC4 S533A failed to recruit procaspase-1 and did not assemble inflammasome specks during S. typhimurium infection, so phosphorylation of NLRC4 S533 probably drives conformational changes necessary for NLRC4 inflammasome activity and host innate immunity.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Inflammasomes/metabolism , Amino Acid Sequence , Animals , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Caspase 1/metabolism , Enzyme Activation , Gene Knock-In Techniques , Humans , Immunity, Innate/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Mice , Molecular Sequence Data , Phosphorylation , Protein Conformation , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Salmonella typhimurium/immunology , Sequence Alignment
17.
Curr Top Microbiol Immunol ; 397: 23-42, 2016.
Article in English | MEDLINE | ID: mdl-27460803

ABSTRACT

Inflammasomes are multimeric protein complexes that mediate the activation of inflammatory caspases. One central component of inflammasomes is nucleotide-binding domain (NBD)- and leucine-rich repeat (LRR)-containing proteins (NLRs) that can function as pattern recognition receptors (PRRs). In resting cells, NLR proteins exist in an auto-inhibited, monomeric, and ADP-bound state. Perception of microbial or damage-associated signals results in NLR oligomerization, thus recruiting inflammatory caspases directly or through the adaptor molecule apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). The assembled NLR inflammasomes serve as dedicated machinery to facilitate the activation of the inflammatory caspases. Here, we review current understanding of the structures of NLR inflammasomes with an emphasis on the molecular mechanisms of their assembly and activation. We also discuss implications of the self-propagation model derived from the NAIP-NLRC4 inflammasomes for the activation of other NLR inflammasomes and a potential role of the C-terminal LRR domain in the activation of an NLR protein.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/immunology , Inflammasomes/chemistry , Inflammasomes/immunology , Animals , CARD Signaling Adaptor Proteins/genetics , Humans , Inflammasomes/genetics , Multigene Family , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction
18.
Int J Mol Sci ; 18(12)2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29194363

ABSTRACT

The three CARD-containing MAGUK (CARMA) proteins function as scaffolding molecules that regulate activation of the pro-inflammatory transcription factor NF-κB. Recently, mutations in CARMA2 have been linked to psoriasis susceptibility due to their acquired altered capacity to activate NF-κB. By means of two-hybrid screening with yeast, we identified RING finger protein 7 (RNF7) as an interactor of CARMA2. We present evidence that RNF7 functions as a negative regulator of the NF-κB-activating capacity of CARMA2. Mechanistically, RNF7 influences CARMA2 signaling by regulating the ubiquitination state of MALT1 and the NF-κB-regulatory molecule NEMO. Interestingly, CARMA2short (CARMA2sh) mutants associated with psoriasis susceptibility escape the negative control exerted by RNF7. In conclusion, our findings identify a new mechanism through which the ability of CARMA2 to activate NF-κB is regulated, which could have significant implications for our understanding of why mutations of this protein trigger human psoriasis.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , CARD Signaling Adaptor Proteins/chemistry , Cell Line , Gene Expression Regulation , Guanylate Cyclase/chemistry , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , Membrane Proteins/chemistry , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Mutation , NF-kappa B/metabolism , Protein Binding , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
19.
Wei Sheng Wu Xue Bao ; 56(9): 1406-14, 2016 Sep.
Article in Zh | MEDLINE | ID: mdl-29738213

ABSTRACT

Under the effects of a series of microbial infection and endogenous or exogenous stimuli, inflammasomes are assembled as multiple protein complexes in the cytoplasm, which mainly contain pattern recognition receptors (PRRs), apoptosis-associated speck-like protein containing a CARD (ASC), and pro-caspase-1. The inflammasomes are the platform for caspase-1 activation and subsequent proinflammatory cytokines secretion, and caspase-1 dependent cell death as well. As a key adaptor protein, ASC concatenates PRRs and pro-caspase-1 in the cytoplasm. During inflammasome activation, ASC molecules assemble into large molecule dimers, which is called ASC-speck. The formation of ASC-speck is critical for caspase-1 activation, and ASC-speck becomes a target for the therapy and prevention of inflammatory diseases. In this review, advances in molecular mechanism of ASC-speck formation and the regulation systems for ASC-speck are summarized from the aspects of phosphorylation, ubiquitination and iron channels etc. Finally, the research prospects in this field are discussed.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Inflammasomes/immunology , Animals , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Humans , Inflammasomes/chemistry , Inflammasomes/genetics , Phosphorylation , Ubiquitination
20.
Apoptosis ; 20(2): 124-35, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25398537

ABSTRACT

Apoptosis is an important process to maintain cellular homeostasis. Deregulated apoptosis has linked to a number of diseases, such as inflammatory diseases, neurodegenerative disorder, and cancers. A major signaling complex in the death receptor signaling pathway leading to apoptosis is death-induced signaling complex (DISC), which is regulated mainly by death effector domain (DED)-containing proteins. There are seven DED-containing proteins in human, including FADD, c-FLIP, caspase-8, caspase-10, DEDD, DEDD2, and PEA-15. The main players in DISC formation employ tandem DEDs for regulating signaling complex formation. The regulatory mechanism of signaling complex formation is important and yet remains unclear. Interestingly, three caspase recruitment domain (CARD)-containing members, which belong to the same DD superfamily as DED-containing proteins, also contains similar tandem CARDs. Recent structural studies have shown that tandem CARDs are essential for the formation of a helical signaling complex. This review summarizes recent structural studies on DED-containing proteins and especially discusses the studies on tandem DEDs and tandem CARDs, which suggest new mechanisms of signaling complex assembly.


Subject(s)
Apoptosis , CARD Signaling Adaptor Proteins/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , CARD Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/physiology , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Signal Transduction , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL