Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Science ; 384(6697): adm9190, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38662913

ABSTRACT

Gasdermins (GSDMs) are pore-forming proteins that execute pyroptosis for immune defense. GSDMs are two-domain proteins activated by proteolytic removal of the inhibitory domain. In this work, we report two types of cleavage-independent GSDM activation. First, TrichoGSDM, a pore-forming domain-only protein from the basal metazoan Trichoplax adhaerens, is a disulfides-linked autoinhibited dimer activated by reduction of the disulfides. The cryo-electron microscopy (cryo-EM) structure illustrates the assembly mechanism for the 44-mer TrichoGSDM pore. Second, RCD-1-1 and RCD-1-2, encoded by the polymorphic regulator of cell death-1 (rcd-1) gene in filamentous fungus Neurospora crassa, are also pore-forming domain-only GSDMs. RCD-1-1 and RCD-1-2, when encountering each other, form pores and cause pyroptosis, underlying allorecognition in Neurospora. The cryo-EM structure reveals a pore of 11 RCD-1-1/RCD-1-2 heterodimers and a heterodimerization-triggered pore assembly mechanism. This study shows mechanistic diversities in GSDM activation and indicates versatile functions of GSDMs.


Subject(s)
Fungal Proteins , Gasdermins , Neurospora crassa , Placozoa , Protein Multimerization , Animals , Cryoelectron Microscopy , Disulfides/chemistry , Fungal Proteins/chemistry , Gasdermins/chemistry , Models, Molecular , Protein Domains , Proteolysis , Pyroptosis
2.
Protein Cell ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38676703

ABSTRACT

Caspase-2, a highly conserved member of the caspase family, is considered an initiator caspase that triggers apoptosis in response to some cellular stresses. Previous studies suggest that an intracellular multi-protein complex PIDDosome, induced by genotoxic stress, serves as a platform for caspase-2 activation. However, due to caspase-2's inability to process effector caspases, the mechanism underlying caspase-2-mediated cell death upon PIDDosome activation remains unclear. Here we conducted an unbiased genome-wide genetic screen and identified that the Bcl2 family protein BID is required for PIDDosome-induced, caspase-2-mediated apoptosis. PIDDosome-activated caspase-2 directly and functionally processes BID to signal the mitochondrial pathway for apoptosis induction. Additionally, a designed chemical screen identified a compound, HUHS015, that specifically activates caspase-2-mediated apoptosis. HUHS015-stimulated apoptosis also requires BID but is independent of the PIDDosome. Through extensive structure-activity relationship efforts, we identified a derivative with a potency of ~ 60 nmol/L in activating caspase-2-mediated apoptosis. The HUHS015-series of compounds act as efficient agonists that directly target the interdomain linker in caspase-2, representing a new mode of initiator caspase activation. Human and mouse caspase-2 differ in two crucial residues in the linker, rendering a selectivity of the agonists for human caspase-2. The caspase-2 agonists are valuable tools to explore the physiological roles of caspase-2-mediated cell death and a base for developing small-molecule drugs for relevant diseases.

3.
Dev Cell ; 58(23): 2761-2775.e5, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37922908

ABSTRACT

Endoplasmic reticulum (ER)-phagy is crucial to regulate the function and homeostasis of the ER via lysosomal degradation, but how it is initiated is unclear. Here we discover that Z-AAT, a disease-causing mutant of α1-antitrypsin, induces noncanonical ER-phagy at ER exit sites (ERESs). Accumulation of misfolded Z-AAT at the ERESs impairs coat protein complex II (COPII)-mediated ER-to-Golgi transport and retains V0 subunits that further assemble V-ATPase at the arrested ERESs. V-ATPase subsequently recruits ATG16L1 onto ERESs to mediate in situ lipidation of LC3C. FAM134B-II is then recruited by LC3C via its LIR motif and elicits ER-phagy leading to efficient lysosomal degradation of Z-AAT. Activation of this ER-phagy mediated by the V-ATPase-ATG16L1-LC3C axis (EVAC) is also triggered by blocking ER export. Our findings identify a pathway which switches COPII-mediated transport to lysosomal degradation for ER quality control.


Subject(s)
Adenosine Triphosphatases , Lysosomes , Adenosine Triphosphatases/metabolism , Lysosomes/metabolism , Protein Transport/physiology , Golgi Apparatus/metabolism , Endoplasmic Reticulum/metabolism , Autophagy
4.
Nature ; 624(7991): 442-450, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37993714

ABSTRACT

The canonical (caspase-1) and noncanonical (comprising caspases 4, 5 and 11; hereafter, caspase-4/5/11) inflammasomes both cleave gasdermin D (GSDMD) to induce pyroptosis1,2. Whereas caspase-1 processes IL-1ß and IL-18 for maturation3-6, no cytokine target has been firmly established for lipopolysaccharide-activated caspase-4/5/117-9. Here we show that activated human caspase-4, but not mouse caspase-11, directly and efficiently processes IL-18 in vitro and during bacterial infections. Caspase-4 cleaves the same tetrapeptide site in pro-IL-18 as caspase-1. The crystal structure of the caspase-4-pro-IL-18 complex reveals a two-site (binary) substrate-recognition mechanism; the catalytic pocket engages the tetrapeptide, and a unique exosite that critically recognizes GSDMD10 similarly binds to a specific structure formed jointly by the propeptide and post-cleavage-site sequences in pro-IL-18. This binary recognition is also used by caspase-5 as well as caspase-1 to process pro-IL-18. In caspase-11, a structural deviation around the exosite underlies its inability to target pro-IL-18, which is restored by rationally designed mutations. The structure of pro-IL-18 features autoinhibitory interactions between the propeptide and the post-cleavage-site region, preventing recognition by the IL-18Rα receptor. Cleavage by caspase-1, -4 or -5 induces substantial conformational changes of IL-18 to generate two critical receptor-binding sites. Our study establishes IL-18 as a target of lipopolysaccharide-activated caspase-4/5. The finding is paradigm shifting in the understanding of noncanonical-inflammasome-mediated defences and also the function of IL-18 in immunity and disease.


Subject(s)
Inflammasomes , Intracellular Signaling Peptides and Proteins , Humans , Inflammasomes/metabolism , Interleukin-18 , Lipopolysaccharides/pharmacology , Caspases/metabolism , Caspase 1/metabolism , Pyroptosis
5.
Methods Mol Biol ; 2696: 149-167, 2023.
Article in English | MEDLINE | ID: mdl-37578722

ABSTRACT

The gasdermin family represents a type of membrane pore-forming proteins. The gasdermin family is extensively characterized as the executioner of pyroptotic cell death in mammals; recent studies suggest that gasdermin-like pore-forming proteins are also present in bacteria and fungi. In humans, gasdermin D (GSDMD) is activated through inter-domain cleavage by caspase-1 in the canonical inflammasome pathway and cytosolic LPS-activated caspase-4 or caspase-5. The cleavage disrupts the autoinhibition of GSDMD and liberates the N-terminal gasdermin-N domain that binds to membrane lipids and forms pores of an inner diameter of ~18 nm on the membrane, responsible for cell pyroptosis. Here, we describe the methods of determining the phospholipid-binding and pore-forming activity of gasdermins in a robust in vitro system. We also introduce a method of specifically detecting the caspase-cleaved form of GSDMD in pyroptotic cells.


Subject(s)
Gasdermins , Intracellular Signaling Peptides and Proteins , Humans , Animals , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Pyroptosis , Caspases/metabolism , Inflammasomes/metabolism , Mammals/metabolism
6.
Nature ; 616(7957): 598-605, 2023 04.
Article in English | MEDLINE | ID: mdl-36991125

ABSTRACT

Cytotoxic lymphocyte-derived granzyme A (GZMA) cleaves GSDMB, a gasdermin-family pore-forming protein1,2, to trigger target cell pyroptosis3. GSDMB and the charter gasdermin family member GSDMD4,5 have been inconsistently reported to be degraded by the Shigella flexneri ubiquitin-ligase virulence factor IpaH7.8 (refs. 6,7). Whether and how IpaH7.8 targets both gasdermins is undefined, and the pyroptosis function of GSDMB has even been questioned recently6,8. Here we report the crystal structure of the IpaH7.8-GSDMB complex, which shows how IpaH7.8 recognizes the GSDMB pore-forming domain. We clarify that IpaH7.8 targets human (but not mouse) GSDMD through a similar mechanism. The structure of full-length GSDMB suggests stronger autoinhibition than in other gasdermins9,10. GSDMB has multiple splicing isoforms that are equally targeted by IpaH7.8 but exhibit contrasting pyroptotic activities. Presence of exon 6 in the isoforms dictates the pore-forming, pyroptotic activity in GSDMB. We determine the cryo-electron microscopy structure of the 27-fold-symmetric GSDMB pore and depict conformational changes that drive pore formation. The structure uncovers an essential role for exon-6-derived elements in pore assembly, explaining pyroptosis deficiency in the non-canonical splicing isoform used in recent studies6,8. Different cancer cell lines have markedly different isoform compositions, correlating with the onset and extent of pyroptosis following GZMA stimulation. Our study illustrates fine regulation of GSDMB pore-forming activity by pathogenic bacteria and mRNA splicing and defines the underlying structural mechanisms.


Subject(s)
Gasdermins , Pore Forming Cytotoxic Proteins , Animals , Humans , Mice , Cell Line, Tumor , Cryoelectron Microscopy , Crystallography, X-Ray , Gasdermins/chemistry , Gasdermins/genetics , Gasdermins/metabolism , Gasdermins/ultrastructure , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/ultrastructure , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/ultrastructure , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Pyroptosis , Shigella flexneri , Species Specificity , Alternative Splicing
7.
Nat Struct Mol Biol ; 30(3): 261-272, 2023 03.
Article in English | MEDLINE | ID: mdl-36624349

ABSTRACT

The caspase-4/11-GSDMD pyroptosis axis recognizes cytosolic lipopolysaccharide for antibacterial defenses. Shigella flexneri employs an OspC3 effector to block pyroptosis by catalyzing NAD+-dependent arginine ADP-riboxanation of caspase-4/11. Here, we identify Ca2+-free calmodulin (CaM) that binds and stimulates OspC3 ADP-riboxanase activity. Crystal structures of OspC3-CaM and OspC3-caspase-4 binary complexes reveal unique CaM binding to an OspC3 N-terminal domain featuring an ADP-ribosyltransferase-like fold and specific recognition of caspase-4 by an OspC3 ankryin repeat domain, respectively. CaM-OspC3-caspase-4 ternary complex structures show that NAD+ binding reorganizes the catalytic pocket, in which D231 and D177 activate the substrate arginine for initial ADP-ribosylation and ribosyl 2'-OH in the ADP-ribosylated arginine, respectively, for subsequent deamination. We also determine structures of unmodified and OspC3-ADP-riboxanated caspase-4. Mechanisms derived from this series of structures covering the entire process of OspC3 action are supported by biochemical analyses in vitro and functional validation in S. flexneri-infected mice.


Subject(s)
Calmodulin , Shigella , Mice , Animals , Calmodulin/metabolism , Pyroptosis , NAD/metabolism , Shigella/metabolism , Arginine
8.
Nat Biomed Eng ; 7(1): 8-23, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36424464

ABSTRACT

The cell-surface glycoprotein CD98-a subunit of the LAT1/CD98 amino acid transporter-is an attractive target for cancer immunotherapies, but its widespread expression has hampered the development of CD98-targeting antibody therapeutics. Here we report that an anti-CD98 antibody, identified via the screening of phage-display libraries of CD98 single-chain variable fragments with mutated complementarity-determining regions, preserves the physiological function of CD98 and elicits broad-spectrum crystallizable-fragment (Fc)-mediated anti-tumour activity (requiring Fcγ receptors for immunoglobulins, macrophages, dendritic cells and CD8+ T cells, as well as other components of the innate and adaptive immune systems) in multiple xenograft and syngeneic tumour models established in CD98-humanized mice. We also show that a variant of the anti-CD98 antibody with pH-dependent binding, generated by solving the structure of the antibody-CD98 complex, displayed enhanced tumour-specific activity and pharmacokinetics. pH-dependent antibody variants targeting widely expressed antigens may lead to superior therapeutic outcomes.


Subject(s)
Neoplasms , Single-Chain Antibodies , Animals , Humans , Mice , CD8-Positive T-Lymphocytes , Hydrogen-Ion Concentration , Fusion Regulatory Protein-1/immunology
9.
mBio ; 13(3): e0069022, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35446120

ABSTRACT

Blocking host cell death is an important virulence strategy employed by many bacterial pathogens. We recently reported that Shigella flexneri inhibits host pyroptosis by delivering a type III secretion system (T3SS) effector OspC3 that catalyzes a novel arginine ADP-riboxanation modification on caspase-4/11. Here, we investigated the OspC3 homologue CopC from Chromobacterium violaceum, an opportunistic but sometimes deadly bacterial pathogen. CopC bears the same arginine ADP-riboxanase activity as OspC3, but with a different substrate specificity. Through proteomic analysis, we first identified host calmodulin (CaM) as a binding partner of CopC. The analyses additionally revealed that CopC preferably modifies apoptotic caspases including caspase-7, -8 and -9. This results in suppression of both extrinsic and intrinsic apoptosis programs in C. violaceum-infected cells. Biochemical reconstitution showed that CopC requires binding to CaM, specifically in the calcium-free state, to achieve efficient ADP-riboxanation of the caspases. We determined crystal structure of the CaM-CopC-CASP7 ternary complex, which illustrates the caspase recognition mechanism and a unique CaM-binding mode in CopC. Structure-directed mutagenesis validated the functional significance of CaM binding for stimulating CopC modification of its caspase substrates. CopC adopts an ADP-ribosyltransferase-like fold with a unique His-Phe-Glu catalytic triad, featuring two acidic residues critical for site-specific arginine ADP-riboxanation. Our study expands and deepens our understanding of the OspC family of ADP-riboxanase effectors. IMPORTANCE Programmed cell death is a suicidal defense mechanism for eukaryotes to combat pathogen infection. In the evolutionary arms race with the host, bacteria are endowed with ingenious tactics to block host cell death to facilitate their replication. Here, we report that the C. violaceum effector CopC ADP-riboxanates caspase-7/8/9, enabled by interacting with the host factor calmodulin, to block host cell apoptosis, illustrating a unique and sophisticated strategy adopted by the pathogen to counteract host defense.


Subject(s)
Calmodulin , Chromobacterium , Adenosine Diphosphate/metabolism , Arginine/metabolism , Calmodulin/metabolism , Caspase 7/metabolism , Caspases/metabolism , Chromobacterium/metabolism , Humans , Proteomics
10.
Autophagy ; 18(11): 2751-2753, 2022 11.
Article in English | MEDLINE | ID: mdl-35410580

ABSTRACT

LC3 lipidation-mediated selective macroautophagy/autophagy helps eukaryotes to defend against endogenous dangers and foreign invaders. However, LC3 activation mechanisms of selective autophagy are still elusive. We previously determined that the V-ATPase-ATG16L1 axis is critical for LC3 recruitment to bacteria-residing vacuoles, whereas the Salmonella effector SopF directly targets V-ATPase to disrupt ATG16L1 interaction. Here we show that host ARF GTPase binding causes SopF-dependent ADP-ribosylation of the Gln124 site of the ATP6V0C/V0C subunit of V-ATPase. Furthermore, LC3 activation by pH perturbation of endolysosomes and the Golgi apparatus is also abolished by SopF or a ATP6V0CQ124A mutation, illustrating that disruption of the proton gradient in acidic compartments is a universal signal that triggers V-ATPase-ATG16L1-induced LC3 lipidation.


Subject(s)
Adenosine Triphosphatases , Microtubule-Associated Proteins , Autophagy-Related Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Autophagy , Macroautophagy
11.
Nat Struct Mol Biol ; 29(1): 67-77, 2022 01.
Article in English | MEDLINE | ID: mdl-35046574

ABSTRACT

Selective autophagy helps eukaryotes to cope with endogenous dangers or foreign invaders; its initiation often involves membrane damage. By studying a Salmonella effector SopF, we recently identified the vacuolar ATPase (V-ATPase)-ATG16L1 axis that initiates bacteria-induced autophagy. Here we show that SopF is an ADP-ribosyltransferase specifically modifying Gln124 of ATP6V0C in V-ATPase. We identify GTP-bound ADP-ribosylation factor (ARF) GTPases as a cofactor required for SopF functioning. Crystal structures of SopF-ARF1 complexes not only reveal structural basis of SopF ADP-ribosyltransferase activity but also a unique effector-binding mode adopted by ARF GTPases. Further, the N terminus of ARF1, although dispensable for high-affinity binding to SopF, is critical for activating SopF to modify ATP6V0C. Moreover, lysosome or Golgi damage-induced autophagic LC3 activation is inhibited by SopF or Q124A mutation of ATP6V0C, thus also mediated by the V-ATPase-ATG16L1 axis. In this process, the V-ATPase functions to sense membrane damages, which can be uncoupled from its proton-pumping activity.


Subject(s)
ADP-Ribosylation Factors/metabolism , Autophagy , Bacterial Proteins/metabolism , Salmonella/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Organelles/metabolism , Protein Binding
12.
Nature ; 599(7884): 290-295, 2021 11.
Article in English | MEDLINE | ID: mdl-34671164

ABSTRACT

Mouse caspase-11 and human caspase-4 and caspase-5 recognize cytosolic lipopolysaccharide (LPS) to induce pyroptosis by cleaving the pore-forming protein GSDMD1-5. This non-canonical inflammasome defends against Gram-negative bacteria6,7. Shigella flexneri, which causes bacillary dysentery, lives freely within the host cytosol where these caspases reside. However, the role of caspase-11-mediated pyroptosis in S. flexneri infection is unknown. Here we show that caspase-11 did not protect mice from S. flexneri infection, in contrast to infection with another cytosolic bacterium, Burkholderia thailandensis8. S. flexneri evaded pyroptosis mediated by caspase-11 or caspase 4 (hereafter referred to as caspase-11/4) using a type III secretion system (T3SS) effector, OspC3. OspC3, but not its paralogues OspC1 and 2, covalently modified caspase-11/4; although it used the NAD+ donor, this modification was not ADP-ribosylation. Biochemical dissections uncovered an ADP-riboxanation modification on Arg314 and Arg310 in caspase-4 and caspase-11, respectively. The enzymatic activity was shared by OspC1 and 2, whose ankyrin-repeat domains, unlike that of OspC3, could not recognize caspase-11/4. ADP-riboxanation of the arginine blocked autoprocessing of caspase-4/11 as well as their recognition and cleavage of GSDMD. ADP-riboxanation of caspase-11 paralysed pyroptosis-mediated defence in Shigella-infected mice and mutation of ospC3 stimulated caspase-11- and GSDMD-dependent anti-Shigella humoral immunity, generating a vaccine-like protective effect. Our study establishes ADP-riboxanation of arginine as a bacterial virulence mechanism that prevents LPS-induced pyroptosis.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Arginine/metabolism , Bacterial Proteins/metabolism , Caspases, Initiator/metabolism , Immune Evasion , Pyroptosis , Shigella flexneri/pathogenicity , Adenosine Diphosphate/metabolism , Animals , Dysentery, Bacillary/immunology , Dysentery, Bacillary/microbiology , Female , Immunity, Humoral , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NAD/metabolism , Pyroptosis/drug effects , Shigella Vaccines , Shigella flexneri/immunology , Virulence
13.
iScience ; 24(9): 102951, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34458700

ABSTRACT

Heat shock factor 1 (HSF1) and 2 (HSF2) play distinct but overlapping regulatory roles in maintaining cellular proteostasis or mediating cell differentiation and development. Upon activation, both HSFs trimerize and bind to heat shock elements (HSEs) present in the promoter region of target genes. Despite structural insights gained from recent studies, structures reflecting the physiological architecture of this transcriptional machinery remains to be determined. Here, we present co-crystal structures of human HSF1 and HSF2 trimers bound to DNA, which reveal a triangular arrangement of the three DNA-binding domains (DBDs) with protein-protein interactions largely mediated by the wing domain. Two structural properties, different flexibility of the wing domain and local DNA conformational changes induced by HSF binding, seem likely to contribute to the subtle differential specificity between HSF1 and HSF2. Besides, two more structures showing DBDs bound to "two-site" head-to-head HSEs were determined as additions to the published tail-to-tail dimer-binding structures.

14.
Science ; 368(6494)2020 05 29.
Article in English | MEDLINE | ID: mdl-32299851

ABSTRACT

Cytotoxic lymphocyte-mediated immunity relies on granzymes. Granzymes are thought to kill target cells by inducing apoptosis, although the underlying mechanisms are not fully understood. Here, we report that natural killer cells and cytotoxic T lymphocytes kill gasdermin B (GSDMB)-positive cells through pyroptosis, a form of proinflammatory cell death executed by the gasdermin family of pore-forming proteins. Killing results from the cleavage of GSDMB by lymphocyte-derived granzyme A (GZMA), which unleashes its pore-forming activity. Interferon-γ (IFN-γ) up-regulates GSDMB expression and promotes pyroptosis. GSDMB is highly expressed in certain tissues, particularly digestive tract epithelia, including derived tumors. Introducing GZMA-cleavable GSDMB into mouse cancer cells promotes tumor clearance in mice. This study establishes gasdermin-mediated pyroptosis as a cytotoxic lymphocyte-killing mechanism, which may enhance antitumor immunity.


Subject(s)
Granzymes/metabolism , Killer Cells, Natural/immunology , Neoplasm Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis/immunology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Granzymes/chemistry , HEK293 Cells , Humans , Interferon-gamma , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/immunology , Neoplasms/pathology , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Protein Domains , Proteolysis
15.
Nature ; 579(7799): 421-426, 2020 03.
Article in English | MEDLINE | ID: mdl-32188939

ABSTRACT

Bioorthogonal chemistry capable of operating in live animals is needed to investigate biological processes such as cell death and immunity. Recent studies have identified a gasdermin family of pore-forming proteins that executes inflammasome-dependent and -independent pyroptosis1-5. Pyroptosis is proinflammatory, but its effect on antitumour immunity is unknown. Here we establish a bioorthogonal chemical system, in which a cancer-imaging probe phenylalanine trifluoroborate (Phe-BF3) that can enter cells desilylates and 'cleaves' a designed linker that contains a silyl ether. This system enabled the controlled release of a drug from an antibody-drug conjugate in mice. When combined with nanoparticle-mediated delivery, desilylation catalysed by Phe-BF3 could release a client protein-including an active gasdermin-from a nanoparticle conjugate, selectively into tumour cells in mice. We applied this bioorthogonal system to gasdermin, which revealed that pyroptosis of less than 15% of tumour cells was sufficient to clear the entire 4T1 mammary tumour graft. The tumour regression was absent in immune-deficient mice or upon T cell depletion, and was correlated with augmented antitumour immune responses. The injection of a reduced, ineffective dose of nanoparticle-conjugated gasdermin along with Phe-BF3 sensitized 4T1 tumours to anti-PD1 therapy. Our bioorthogonal system based on Phe-BF3 desilylation is therefore a powerful tool for chemical biology; our application of this system suggests that pyroptosis-induced inflammation triggers robust antitumour immunity and can synergize with checkpoint blockade.


Subject(s)
Delayed-Action Preparations/administration & dosage , Mammary Neoplasms, Experimental/immunology , Pyroptosis/immunology , Animals , Coumarins/administration & dosage , Coumarins/chemistry , Coumarins/metabolism , Coumarins/pharmacokinetics , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/metabolism , Delayed-Action Preparations/pharmacokinetics , Female , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/pharmacokinetics , HeLa Cells , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Immunoconjugates/pharmacokinetics , Inflammasomes/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacokinetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proteins/administration & dosage , Proteins/chemistry , Proteins/metabolism , Proteins/pharmacokinetics , Silanes/administration & dosage , Silanes/chemistry , Silanes/metabolism , Silanes/pharmacokinetics , T-Lymphocytes/immunology , Trastuzumab/administration & dosage , Trastuzumab/chemistry , Trastuzumab/metabolism , Trastuzumab/pharmacokinetics , Xenograft Model Antitumor Assays
16.
Cell ; 180(5): 941-955.e20, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32109412

ABSTRACT

The pyroptosis execution protein GSDMD is cleaved by inflammasome-activated caspase-1 and LPS-activated caspase-11/4/5. The cleavage unmasks the pore-forming domain from GSDMD-C-terminal domain. How the caspases recognize GSDMD and its connection with caspase activation are unknown. Here, we show site-specific caspase-4/11 autoprocessing, generating a p10 product, is required and sufficient for cleaving GSDMD and inducing pyroptosis. The p10-form autoprocessed caspase-4/11 binds the GSDMD-C domain with a high affinity. Structural comparison of autoprocessed and unprocessed capase-11 identifies a ß sheet induced by the autoprocessing. In caspase-4/11-GSDMD-C complex crystal structures, the ß sheet organizes a hydrophobic GSDMD-binding interface that is only possible for p10-form caspase-4/11. The binding promotes dimerization-mediated caspase activation, rendering a cleavage independently of the cleavage-site tetrapeptide sequence. Crystal structure of caspase-1-GSDMD-C complex shows a similar GSDMD-recognition mode. Our study reveals an unprecedented substrate-targeting mechanism for caspases. The hydrophobic interface suggests an additional space for developing inhibitors specific for pyroptotic caspases.


Subject(s)
Inflammasomes/ultrastructure , Multiprotein Complexes/ultrastructure , Phosphate-Binding Proteins/ultrastructure , Pyroptosis/genetics , Animals , Caspase 1/chemistry , Caspase 1/genetics , Caspase 1/ultrastructure , Caspases, Initiator/chemistry , Caspases, Initiator/genetics , Crystallography, X-Ray , HEK293 Cells , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Inflammasomes/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Phosphate-Binding Proteins/chemistry , Phosphate-Binding Proteins/genetics , Protein Conformation, beta-Strand/genetics , Protein Domains/genetics , Protein Processing, Post-Translational/genetics , Proteolysis
17.
Cell ; 178(3): 552-566.e20, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31327526

ABSTRACT

Antibacterial autophagy (xenophagy) is an important host defense, but how it is initiated is unclear. Here, we performed a bacterial transposon screen and identified a T3SS effector SopF that potently blocked Salmonella autophagy. SopF was a general xenophagy inhibitor without affecting canonical autophagy. S. Typhimurium ΔsopF resembled S. flexneri ΔvirAΔicsB with the majority of intracellular bacteria targeted by autophagy, permitting a CRISPR screen that identified host V-ATPase as an essential factor. Upon bacteria-caused vacuolar damage, the V-ATPase recruited ATG16L1 onto bacteria-containing vacuole, which was blocked by SopF. Mammalian ATG16L1 bears a WD40 domain required for interacting with the V-ATPase. Inhibiting autophagy by SopF promoted S. Typhimurium proliferation in vivo. SopF targeted Gln124 of ATP6V0C in the V-ATPase for ADP-ribosylation. Mutation of Gln124 also blocked xenophagy, but not canonical autophagy. Thus, the discovery of SopF reveals the V-ATPase-ATG16L1 axis that critically mediates autophagic recognition of intracellular pathogen.


Subject(s)
Autophagy-Related Proteins/metabolism , Bacterial Proteins/genetics , Macroautophagy , Salmonella/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Virulence Factors/genetics , ADP-Ribosylation , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism , Protein Binding , Salmonella/pathogenicity , Type III Secretion Systems/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Virulence Factors/metabolism
18.
Mol Cell ; 74(5): 922-935.e6, 2019 06 06.
Article in English | MEDLINE | ID: mdl-30979585

ABSTRACT

Enteropathogenic E. coli NleB and related type III effectors catalyze arginine GlcNAcylation of death domain (DD) proteins to block host defense, but the underlying mechanism is unknown. Here we solve crystal structures of NleB alone and in complex with FADD-DD, UDP, and Mn2+ as well as NleB-GlcNAcylated DDs of TRADD and RIPK1. NleB adopts a GT-A fold with a unique helix-pair insertion to hold FADD-DD; the interface contacts explain the selectivity of NleB for certain DDs. The acceptor arginine is fixed into a cleft, in which Glu253 serves as a base to activate the guanidinium. Analyses of the enzyme-substrate complex and the product structures reveal an inverting sugar-transfer reaction and a detailed catalytic mechanism. These structural insights are validated by mutagenesis analyses of NleB-mediated GlcNAcylation in vitro and its function in mouse infection. Our study builds a structural framework for understanding of NleB-catalyzed arginine GlcNAcylation of host death domain.


Subject(s)
Enteropathogenic Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Host-Pathogen Interactions/genetics , Protein Conformation , Virulence Factors/chemistry , Animals , Apoptosis/genetics , Arginine/chemistry , Arginine/genetics , Coenzyme A Ligases/chemistry , Coenzyme A Ligases/genetics , Crystallography, X-Ray , Death Domain/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Guanidine/chemistry , Humans , Manganese/chemistry , Mice , Mutagenesis , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/genetics , Virulence Factors/genetics
19.
Nature ; 561(7721): 122-126, 2018 09.
Article in English | MEDLINE | ID: mdl-30111836

ABSTRACT

Immune recognition of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors often activates proinflammatory NF-κB signalling1. Recent studies indicate that the bacterial metabolite D-glycero-ß-D-manno-heptose 1,7-bisphosphate (HBP) can activate NF-κB signalling in host cytosol2-4, but it is unclear whether HBP is a genuine PAMP and the cognate pattern recognition receptor has not been identified. Here we combined a transposon screen in Yersinia pseudotuberculosis with biochemical analyses and identified ADP-ß-D-manno-heptose (ADP-Hep), which mediates type III secretion system-dependent NF-κB activation and cytokine expression. ADP-Hep, but not other heptose metabolites, could enter host cytosol to activate NF-κB. A CRISPR-Cas9 screen showed that activation of NF-κB by ADP-Hep involves an ALPK1 (alpha-kinase 1)-TIFA (TRAF-interacting protein with forkhead-associated domain) axis. ADP-Hep directly binds the N-terminal domain of ALPK1, stimulating its kinase domain to phosphorylate and activate TIFA. The crystal structure of the N-terminal domain of ALPK1 and ADP-Hep in complex revealed the atomic mechanism of this ligand-receptor recognition process. HBP was transformed by host adenylyltransferases into ADP-heptose 7-P, which could activate ALPK1 to a lesser extent than ADP-Hep. ADP-Hep (but not HBP) alone or during bacterial infection induced Alpk1-dependent inflammation in mice. Our findings identify ALPK1 and ADP-Hep as a pattern recognition receptor and an effective immunomodulator, respectively.


Subject(s)
Adenosine Diphosphate Sugars/immunology , Burkholderia cenocepacia , Cytosol , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/immunology , Protein Kinases/metabolism , Yersinia pseudotuberculosis , Adenosine Diphosphate Sugars/metabolism , Animals , Burkholderia Infections/enzymology , Burkholderia Infections/immunology , Burkholderia Infections/pathology , Burkholderia cenocepacia/genetics , Burkholderia cenocepacia/immunology , Burkholderia cenocepacia/metabolism , CRISPR-Cas Systems , Crystallography, X-Ray , Cytokines/biosynthesis , Cytosol/enzymology , Cytosol/immunology , Disaccharides/metabolism , Enzyme Activation , Female , Gene Editing , Immunologic Factors/immunology , Immunologic Factors/metabolism , Immunomodulation , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Models, Molecular , NF-kappa B/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis/metabolism
20.
EMBO J ; 37(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29997177

Subject(s)
Pyroptosis , Caspase 1 , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...