Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 210
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 24(12): 2021-2031, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37903858

ABSTRACT

S100A8/S100A9 is a proinflammatory mediator released by myeloid cells during many acute and chronic inflammatory disorders. However, the precise mechanism of its release from the cytosolic compartment of neutrophils is unclear. Here, we show that E-selectin-induced rapid S100A8/S100A9 release during inflammation occurs in an NLRP3 inflammasome-dependent fashion. Mechanistically, E-selectin engagement triggers Bruton's tyrosine kinase-dependent tyrosine phosphorylation of NLRP3. Concomitant potassium efflux via the voltage-gated potassium channel KV1.3 mediates ASC oligomerization. This is followed by caspase 1 cleavage and downstream activation of pore-forming gasdermin D, enabling cytosolic release of S100A8/S100A9. Strikingly, E-selectin-mediated gasdermin D pore formation does not result in cell death but is a transient process involving activation of the ESCRT III membrane repair machinery. These data clarify molecular mechanisms of controlled S100A8/S100A9 release from neutrophils and identify the NLRP3/gasdermin D axis as a rapid and reversible activation system in neutrophils during inflammation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Gasdermins , Neutrophils/metabolism , E-Selectin/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Inflammation/metabolism
2.
Cell ; 179(6): 1264-1275.e13, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31778653

ABSTRACT

TLR8 is among the highest-expressed pattern-recognition receptors in the human myeloid compartment, yet its mode of action is poorly understood. TLR8 engages two distinct ligand binding sites to sense RNA degradation products, although it remains unclear how these ligands are formed in cellulo in the context of complex RNA molecule sensing. Here, we identified the lysosomal endoribonuclease RNase T2 as a non-redundant upstream component of TLR8-dependent RNA recognition. RNase T2 activity is required for rendering complex single-stranded, exogenous RNA molecules detectable for TLR8. This is due to RNase T2's preferential cleavage of single-stranded RNA molecules between purine and uridine residues, which critically contributes to the supply of catabolic uridine and the generation of purine-2',3'-cyclophosphate-terminated oligoribonucleotides. Thus-generated molecules constitute agonistic ligands for the first and second binding pocket of TLR8. Together, these results establish the identity and origin of the RNA-derived molecular pattern sensed by TLR8.


Subject(s)
Endoribonucleases/metabolism , Proteolysis , Toll-Like Receptor 8/metabolism , Amino Acid Motifs , Base Sequence , Cell Line , Endoribonucleases/deficiency , Humans , Models, Molecular , Monocytes/metabolism , Myeloid Cells/metabolism , Nitrogen Isotopes , Oligonucleotides/metabolism , Purines/metabolism , RNA/metabolism , Staphylococcus aureus/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/chemistry , Uridine/metabolism
3.
Immunity ; 57(7): 1482-1496.e8, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38697119

ABSTRACT

Toll-like receptor 7 (TLR7) is essential for recognition of RNA viruses and initiation of antiviral immunity. TLR7 contains two ligand-binding pockets that recognize different RNA degradation products: pocket 1 recognizes guanosine, while pocket 2 coordinates pyrimidine-rich RNA fragments. We found that the endonuclease RNase T2, along with 5' exonucleases PLD3 and PLD4, collaboratively generate the ligands for TLR7. Specifically, RNase T2 generated guanosine 2',3'-cyclic monophosphate-terminated RNA fragments. PLD exonuclease activity further released the terminal 2',3'-cyclic guanosine monophosphate (2',3'-cGMP) to engage pocket 1 and was also needed to generate RNA fragments for pocket 2. Loss-of-function studies in cell lines and primary cells confirmed the critical requirement for PLD activity. Biochemical and structural studies showed that PLD enzymes form homodimers with two ligand-binding sites important for activity. Previously identified disease-associated PLD mutants failed to form stable dimers. Together, our data provide a mechanistic basis for the detection of RNA fragments by TLR7.


Subject(s)
Endoribonucleases , Toll-Like Receptor 7 , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Humans , Endoribonucleases/metabolism , Ligands , Phospholipase D/metabolism , Phospholipase D/genetics , RNA/metabolism , HEK293 Cells , Lysosomes/metabolism , Animals , Exonucleases/metabolism , Mice , Binding Sites
4.
Immunity ; 57(8): 1812-1827.e7, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38955184

ABSTRACT

An important property of the host innate immune response during microbial infection is its ability to control the expression of antimicrobial effector proteins, but how this occurs post-transcriptionally is not well defined. Here, we describe a critical antibacterial role for the classic antiviral gene 2'-5'-oligoadenylate synthetase 1 (OAS1). Human OAS1 and its mouse ortholog, Oas1b, are induced by interferon-γ and protect against cytosolic bacterial pathogens such as Francisella novicida and Listeria monocytogenes in vitro and in vivo. Proteomic and transcriptomic analysis showed reduced IRF1 protein expression in OAS1-deficient cells. Mechanistically, OAS1 binds and localizes IRF1 mRNA to the rough endoplasmic reticulum (ER)-Golgi endomembranes, licensing effective translation of IRF1 mRNA without affecting its transcription or decay. OAS1-dependent translation of IRF1 leads to the enhanced expression of antibacterial effectors, such as GBPs, which restrict intracellular bacteria. These findings uncover a noncanonical function of OAS1 in antibacterial innate immunity.


Subject(s)
2',5'-Oligoadenylate Synthetase , Immunity, Innate , Interferon Regulatory Factor-1 , 2',5'-Oligoadenylate Synthetase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Animals , Humans , Mice , Protein Biosynthesis/immunology , Listeria monocytogenes/immunology , Mice, Knockout , Mice, Inbred C57BL , Listeriosis/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology
5.
Immunity ; 57(3): 446-461.e7, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38423012

ABSTRACT

In response to viral infection, how cells balance translational shutdown to limit viral replication and the induction of antiviral components like interferons (IFNs) is not well understood. Moreover, how distinct isoforms of IFN-induced oligoadenylate synthetase 1 (OAS1) contribute to this antiviral response also requires further elucidation. Here, we show that human, but not mouse, OAS1 inhibits SARS-CoV-2 replication through its canonical enzyme activity via RNase L. In contrast, both mouse and human OAS1 protect against West Nile virus infection by a mechanism distinct from canonical RNase L activation. OAS1 binds AU-rich elements (AREs) of specific mRNAs, including IFNß. This binding leads to the sequestration of IFNß mRNA to the endomembrane regions, resulting in prolonged half-life and continued translation. Thus, OAS1 is an ARE-binding protein with two mechanisms of antiviral activity: driving inhibition of translation but also a broader, non-canonical function of protecting IFN expression from translational shutdown.


Subject(s)
2',5'-Oligoadenylate Synthetase , Interferons , Oligoribonucleotides , Virus Diseases , West Nile Fever , Animals , Humans , Mice , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Adenine Nucleotides , Antiviral Agents/pharmacology , West Nile Fever/genetics , West Nile Fever/metabolism , West Nile virus/metabolism , West Nile virus/pathogenicity
6.
Nat Rev Mol Cell Biol ; 21(9): 501-521, 2020 09.
Article in English | MEDLINE | ID: mdl-32424334

ABSTRACT

The cGAS-STING signalling axis, comprising the synthase for the second messenger cyclic GMP-AMP (cGAS) and the cyclic GMP-AMP receptor stimulator of interferon genes (STING), detects pathogenic DNA to trigger an innate immune reaction involving a strong type I interferon response against microbial infections. Notably however, besides sensing microbial DNA, the DNA sensor cGAS can also be activated by endogenous DNA, including extranuclear chromatin resulting from genotoxic stress and DNA released from mitochondria, placing cGAS-STING as an important axis in autoimmunity, sterile inflammatory responses and cellular senescence. Initial models assumed that co-localization of cGAS and DNA in the cytosol defines the specificity of the pathway for non-self, but recent work revealed that cGAS is also present in the nucleus and at the plasma membrane, and such subcellular compartmentalization was linked to signalling specificity of cGAS. Further confounding the simple view of cGAS-STING signalling as a response mechanism to infectious agents, both cGAS and STING were shown to have additional functions, independent of interferon response. These involve non-catalytic roles of cGAS in regulating DNA repair and signalling via STING to NF-κB and MAPK as well as STING-mediated induction of autophagy and lysosome-dependent cell death. We have also learnt that cGAS dimers can multimerize and undergo liquid-liquid phase separation to form biomolecular condensates that could importantly regulate cGAS activation. Here, we review the molecular mechanisms and cellular functions underlying cGAS-STING activation and signalling, particularly highlighting the newly emerging diversity of this signalling pathway and discussing how the specificity towards normal, damage-induced and infection-associated DNA could be achieved.


Subject(s)
Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Animals , Autophagy , Cyclic AMP/metabolism , Cyclic AMP/physiology , Cyclic GMP/metabolism , Cyclic GMP/physiology , Cytosol/metabolism , DNA/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/physiology , Nucleotides, Cyclic , Nucleotidyltransferases/genetics , Signal Transduction
7.
Cell ; 171(5): 1110-1124.e18, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29033128

ABSTRACT

Detection of cytosolic DNA constitutes a central event in the context of numerous infectious and sterile inflammatory conditions. Recent studies have uncovered a bipartite mode of cytosolic DNA recognition, in which the cGAS-STING axis triggers antiviral immunity, whereas AIM2 triggers inflammasome activation. Here, we show that AIM2 is dispensable for DNA-mediated inflammasome activation in human myeloid cells. Instead, detection of cytosolic DNA by the cGAS-STING axis induces a cell death program initiating potassium efflux upstream of NLRP3. Forward genetics identified regulators of lysosomal trafficking to modulate this cell death program, and subsequent studies revealed that activated STING traffics to the lysosome, where it triggers membrane permeabilization and thus lysosomal cell death (LCD). Importantly, the cGAS-STING-NLRP3 pathway constitutes the default inflammasome response during viral and bacterial infections in human myeloid cells. We conclude that targeting the cGAS-STING-LCD-NLRP3 pathway will ameliorate pathology in inflammatory conditions that are associated with cytosolic DNA sensing.


Subject(s)
Cell Death , Inflammasomes/metabolism , Monocytes/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , DNA/metabolism , Humans , Membrane Proteins/metabolism , Monocytes/metabolism , Signal Transduction
8.
Mol Cell ; 84(12): 2218-2220, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906113

ABSTRACT

Three recent publications by Du et al.,1 Balasubramanian et al.,2 and Zhang et al.3 identified palmitoylation on cysteine 191/192 in gasdermin D as a key determinant of gasdermin D membrane translocation and oligomerization, ensuring efficient plasma membrane permeabilization during pyroptosis.


Subject(s)
Lipoylation , Phosphate-Binding Proteins , Pyroptosis , Humans , Animals , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cell Membrane/metabolism , Cysteine/metabolism , Protein Transport , Gasdermins
9.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36384135

ABSTRACT

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , I-kappa B Kinase , Inflammasomes/metabolism , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , trans-Golgi Network/metabolism
10.
Immunity ; 54(4): 648-659.e8, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33667383

ABSTRACT

Loss of lymphocytes, particularly T cell apoptosis, is a central pathological event after severe tissue injury that is associated with increased susceptibility for life-threatening infections. The precise immunological mechanisms leading to T cell death after acute injury are largely unknown. Here, we identified a monocyte-T cell interaction driving bystander cell death of T cells in ischemic stroke and burn injury. Specifically, we found that stroke induced a FasL-expressing monocyte population, which led to extrinsic T cell apoptosis. This phenomenon was driven by AIM2 inflammasome-dependent interleukin-1ß (IL-1ß) secretion after sensing cell-free DNA. Pharmacological inhibition of this pathway improved T cell survival and reduced post-stroke bacterial infections. As such, this study describes inflammasome-dependent monocyte activation as a previously unstudied cause of T cell death after injury and challenges the current paradigms of post-injury lymphopenia.


Subject(s)
Coinfection/immunology , DNA-Binding Proteins/immunology , Immune Tolerance/immunology , Inflammasomes/immunology , Signal Transduction/immunology , Animals , Apoptosis/immunology , Bacterial Infections/immunology , Burns/immunology , Burns/microbiology , Coinfection/microbiology , Humans , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Stroke/immunology , Stroke/microbiology , T-Lymphocytes/immunology
11.
Nature ; 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112714

ABSTRACT

The risk of early recurrent events after stroke remains high despite currently established secondary prevention strategies1. Risk is particularly high in patients with atherosclerosis, with more than 10% of patients experiencing early recurrent events1,2. However, despite the enormous medical burden of this clinical phenomenon, the underlying mechanisms leading to increased vascular risk and recurrent stroke are largely unknown. Here, using a novel mouse model of stroke-induced recurrent ischaemia, we show that stroke leads to activation of the AIM2 inflammasome in vulnerable atherosclerotic plaques via an increase of circulating cell-free DNA. Enhanced plaque inflammation post-stroke results in plaque destabilization and atherothrombosis, finally leading to arterioarterial embolism and recurrent stroke within days after the index stroke. We confirm key steps of plaque destabilization also after experimental myocardial infarction and in carotid artery plaque samples from patients with acute stroke. Rapid neutrophil NETosis was identified as the main source of cell-free DNA after stroke and NET-DNA as the causative agent leading to AIM2 inflammasome activation. Neutralization of cell-free DNA by DNase treatment or inhibition of inflammasome activation reduced the rate of stroke recurrence after experimental stroke. Our findings present an explanation for the high recurrence rate after incident ischaemic events in patients with atherosclerosis. The detailed mechanisms uncovered here provide clinically uncharted therapeutic targets for which we show high efficacy to prevent recurrent events. Targeting DNA-mediated inflammasome activation after remote tissue injury represents a promising avenue for further clinical development in the prevention of early recurrent events.

12.
Nature ; 609(7927): 590-596, 2022 09.
Article in English | MEDLINE | ID: mdl-36002575

ABSTRACT

Bacterial cell wall components provide various unique molecular structures that are detected by pattern recognition receptors (PRRs) of the innate immune system as non-self. Most bacterial species form a cell wall that consists of peptidoglycan (PGN), a polymeric structure comprising alternating amino sugars that form strands cross-linked by short peptides. Muramyl dipeptide (MDP) has been well documented as a minimal immunogenic component of peptidoglycan1-3. MDP is sensed by the cytosolic nucleotide-binding oligomerization domain-containing protein 24 (NOD2). Upon engagement, it triggers pro-inflammatory gene expression, and this functionality is of critical importance in maintaining a healthy intestinal barrier function5. Here, using a forward genetic screen to identify factors required for MDP detection, we identified N-acetylglucosamine kinase (NAGK) as being essential for the immunostimulatory activity of MDP. NAGK is broadly expressed in immune cells and has previously been described to contribute to the hexosamine biosynthetic salvage pathway6. Mechanistically, NAGK functions upstream of NOD2 by directly phosphorylating the N-acetylmuramic acid moiety of MDP at the hydroxyl group of its C6 position, yielding 6-O-phospho-MDP. NAGK-phosphorylated MDP-but not unmodified MDP-constitutes an agonist for NOD2. Macrophages from mice deficient in NAGK are completely deficient in MDP sensing. These results reveal a link between amino sugar metabolism and innate immunity to bacterial cell walls.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine , Nod2 Signaling Adaptor Protein , Phosphotransferases (Alcohol Group Acceptor) , Acetylmuramyl-Alanyl-Isoglutamine/chemistry , Acetylmuramyl-Alanyl-Isoglutamine/immunology , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Bacteria/chemistry , Bacteria/immunology , Cell Wall/chemistry , Hexosamines/biosynthesis , Immunity, Innate , Macrophages/enzymology , Macrophages/immunology , Mice , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , Peptidoglycan/chemistry , Peptidoglycan/immunology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism
13.
EMBO J ; 42(23): e113279, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37881155

ABSTRACT

The immune system is in place to assist in ensuring tissue homeostasis, which can be easily perturbed by invading pathogens or nonpathogenic stressors causing tissue damage. Extracellular nucleotides are well known to contribute to innate immune signaling specificity and strength, but how their signaling is relayed downstream of cell surface receptors and how this translates into antiviral immunity is only partially understood. Here, we systematically investigated the responses of human macrophages to extracellular nucleotides, focusing on the nucleotide-sensing GPRC receptors of the P2Y family. Time-resolved transcriptomic analysis showed that adenine- and uridine-based nucleotides induce a specific, immediate, and transient cytokine response through the MAPK signaling pathway that regulates transcriptional activation by AP-1. Using receptor trans-complementation, we identified a subset of P2Ys (P2Y1, P2Y2, P2Y6, and P2Y11) that govern inflammatory responses via cytokine induction, while others (P2Y4, P2Y11, P2Y12, P2Y13, and P2Y14) directly induce antiviral responses. Notably, P2Y11 combined both activities, and depletion or inhibition of this receptor in macrophages impaired both inflammatory and antiviral responses. Collectively, these results highlight the underappreciated functions of P2Y receptors in innate immune processes.


Subject(s)
Nucleotides , Signal Transduction , Humans , Cytokines , Immunity , Macrophages/metabolism , Nucleotides/metabolism , Virus Replication
14.
Nat Immunol ; 16(10): 1025-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343537

ABSTRACT

Cytosolic DNA that emerges during infection with a retrovirus or DNA virus triggers antiviral type I interferon responses. So far, only double-stranded DNA (dsDNA) over 40 base pairs (bp) in length has been considered immunostimulatory. Here we found that unpaired DNA nucleotides flanking short base-paired DNA stretches, as in stem-loop structures of single-stranded DNA (ssDNA) derived from human immunodeficiency virus type 1 (HIV-1), activated the type I interferon-inducing DNA sensor cGAS in a sequence-dependent manner. DNA structures containing unpaired guanosines flanking short (12- to 20-bp) dsDNA (Y-form DNA) were highly stimulatory and specifically enhanced the enzymatic activity of cGAS. Furthermore, we found that primary HIV-1 reverse transcripts represented the predominant viral cytosolic DNA species during early infection of macrophages and that these ssDNAs were highly immunostimulatory. Collectively, our study identifies unpaired guanosines in Y-form DNA as a highly active, minimal cGAS recognition motif that enables detection of HIV-1 ssDNA.


Subject(s)
DNA, Complementary/chemistry , DNA, Viral/chemistry , DNA, Viral/immunology , HIV-1/genetics , HIV-1/immunology , Interferon-alpha/immunology , Nucleotidyltransferases/genetics , Animals , Cell Line , Cells, Cultured , DNA, Complementary/genetics , DNA, Complementary/immunology , DNA, Viral/genetics , HEK293 Cells , Humans , Immunization , Mice
15.
Nature ; 587(7835): 678-682, 2020 11.
Article in English | MEDLINE | ID: mdl-32911480

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is an innate immune sensor for cytosolic microbial DNA1. After binding DNA, cGAS synthesizes the messenger 2'3'-cyclic GMP-AMP (cGAMP)2-4, which triggers cell-autonomous defence and the production of type I interferons and pro-inflammatory cytokines via the activation of STING5. In addition to responding to cytosolic microbial DNA, cGAS also recognizes mislocalized cytosolic self-DNA and has been implicated in autoimmunity and sterile inflammation6,7. Specificity towards pathogen- or damage-associated DNA was thought to be caused by cytosolic confinement. However, recent findings place cGAS robustly in the nucleus8-10, where tight tethering of chromatin is important to prevent autoreactivity to self-DNA8. Here we show how cGAS is sequestered and inhibited by chromatin. We provide a cryo-electron microscopy structure of the cGAS catalytic domain bound to a nucleosome, which shows that cGAS does not interact with the nucleosomal DNA, but instead interacts with histone 2A-histone 2B, and is tightly anchored to the 'acidic patch'. The interaction buries the cGAS DNA-binding site B, and blocks the formation of active cGAS dimers. The acidic patch robustly outcompetes agonistic DNA for binding to cGAS, which suggests that nucleosome sequestration can efficiently inhibit cGAS, even when accessible DNA is nearby, such as in actively transcribed genomic regions. Our results show how nuclear cGAS is sequestered by chromatin and provides a mechanism for preventing autoreactivity to nuclear self-DNA.


Subject(s)
Catalytic Domain , Chromatin/chemistry , Chromatin/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/chemistry , Amino Acid Sequence , Animals , Autoantigens/chemistry , Autoantigens/immunology , Autoantigens/metabolism , Autoantigens/ultrastructure , Binding Sites , Binding, Competitive , Chromatin/genetics , Chromatin/ultrastructure , Cryoelectron Microscopy , DNA/chemistry , DNA/immunology , DNA/metabolism , DNA/ultrastructure , Enzyme Activation , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Hydrophobic and Hydrophilic Interactions , Immunity, Innate , Mice , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/ultrastructure , Protein Multimerization , THP-1 Cells
16.
Eur J Immunol ; : e2451135, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39086059

ABSTRACT

Inflammasomes are essential for host defense, recognizing foreign or stress signals to trigger immune responses, including maturation of IL-1 family cytokines and pyroptosis. Here, NLRP1 is emerging as an important sensor of viral infection in barrier tissues. NLRP1 is activated by various stimuli, including viral double-stranded (ds) RNA, ribotoxic stress, and inhibition of dipeptidyl peptidases 8 and 9 (DPP8/9). However, certain viruses, most notably the vaccinia virus, have evolved strategies to subvert inflammasome activation or effector functions. Using the modified vaccinia virus Ankara (MVA) as a model, we investigated how the vaccinia virus inhibits inflammasome activation. We confirmed that the early gene F1L plays a critical role in inhibiting NLRP1 inflammasome activation. Interestingly, it blocks dsRNA and ribotoxic stress-dependent NLRP1 activation without affecting its DPP9-inhibition-mediated activation. Complementation and loss-of-function experiments demonstrated the sufficiency and necessity of F1L in blocking NLRP1 activation. Furthermore, we found that F1L-deficient, but not wild-type MVA, induced ZAKα activation. Indeed, an F1L-deficient virus was found to disrupt protein translation more prominently than an unmodified virus, suggesting that F1L acts in part upstream of ZAKα. These findings underscore the inhibitory role of F1L on NLRP1 inflammasome activation and provide insight into viral evasion of host defenses and the intricate mechanisms of inflammasome activation.

17.
Nat Methods ; 19(1): 81-89, 2022 01.
Article in English | MEDLINE | ID: mdl-34949807

ABSTRACT

CD4+ T cells are central mediators of adaptive and innate immune responses and constitute a major reservoir for human immunodeficiency virus (HIV) in vivo. Detailed investigations of resting human CD4+ T cells have been precluded by the absence of efficient approaches for genetic manipulation limiting our understanding of HIV replication and restricting efforts to find a cure. Here we report a method for rapid, efficient, activation-neutral gene editing of resting, polyclonal human CD4+ T cells using optimized cell cultivation and nucleofection conditions of Cas9-guide RNA ribonucleoprotein complexes. Up to six genes, including HIV dependency and restriction factors, were knocked out individually or simultaneously and functionally characterized. Moreover, we demonstrate the knock in of double-stranded DNA donor templates into different endogenous loci, enabling the study of the physiological interplay of cellular and viral components at single-cell resolution. Together, this technique allows improved molecular and functional characterizations of HIV biology and general immune functions in resting CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CRISPR-Cas Systems/genetics , Gene Editing/methods , HIV Infections/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , CRISPR-Associated Protein 9/genetics , Cell Movement/genetics , Cells, Cultured , DNA , Gene Knockout Techniques , HIV Infections/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , RNA, Guide, Kinetoplastida , SAM Domain and HD Domain-Containing Protein 1/genetics , Transgenes , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
18.
Immunity ; 44(4): 739-54, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096317

ABSTRACT

Recognition of DNA and RNA by endosomal and cytosolic sensors constitutes a central element in the detection of microbial invaders by the innate immune system. However, the capacity of these sensors to discriminate between microbial and endogenous nucleic acids is limited. Over the past few years, evidence has accumulated to suggest that endogenous DNA or RNA species can engage nucleic-acid-sensing pattern-recognition receptors that can trigger or sustain detrimental pathology. Here, we review principles of how the activation of innate sensors by host nucleic acids is prevented in the steady state and discuss four important determinants of whether a nucleic-acid-driven innate response is mounted. These include structural features of the ligand being sensed, the subcellular location and quantity of pathogen-derived or endogenous nucleic acids, and the regulation of sensor-activation thresholds. Furthermore, we emphasize disease mechanisms initiated by failure to discriminate self from non-self in nucleic acid detection.


Subject(s)
DNA/immunology , RNA/immunology , Receptors, Pattern Recognition/immunology , Humans , Immunity, Innate/immunology , Signal Transduction/immunology
19.
Immunity ; 44(6): 1406-21, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27287410

ABSTRACT

Monobenzone is a pro-hapten that is exclusively metabolized by melanocytes, thereby haptenizing melanocyte-specific antigens, which results in cytotoxic autoimmunity specifically against pigmented cells. Studying monobenzone in a setting of contact hypersensitivity (CHS), we observed that monobenzone induced a long-lasting, melanocyte-specific immune response that was dependent on NK cells, yet fully intact in the absence of T- and B cells. Consistent with the concept of "memory NK cells," monobenzone-induced NK cells resided in the liver and transfer of these cells conferred melanocyte-specific immunity to naive animals. Monobenzone-exposed skin displayed macrophage infiltration and cutaneous lymph nodes showed an inflammasome-dependent influx of macrophages with a tissue-resident phenotype, coinciding with local NK cell activation. Indeed, macrophage depletion or the absence of the NLRP3 inflammasome, the adaptor protein ASC or interleukin-18 (IL-18) abolished monobenzone CHS, thereby establishing a non-redundant role for the NLRP3 inflammasome as a critical proinflammatory checkpoint in the induction of hapten-dependent memory NK cells.


Subject(s)
Dermatitis, Contact/immunology , Immunologic Memory , Inflammasomes/immunology , Killer Cells, Natural/immunology , Macrophages/physiology , Melanocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adaptive Immunity , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Cells, Cultured , Hydroquinones , Interleukin-18/genetics , Interleukin-18/metabolism , Liver/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
20.
Immunity ; 44(4): 833-46, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27037191

ABSTRACT

Interleukin-1ß (IL-1ß) is a cytokine whose bioactivity is controlled by activation of the inflammasome. However, in response to lipopolysaccharide, human monocytes secrete IL-1ß independently of classical inflammasome stimuli. Here, we report that this constituted a species-specific response that is not observed in the murine system. Indeed, in human monocytes, lipopolysaccharide triggered an "alternative inflammasome" that relied on NLRP3-ASC-caspase-1 signaling, yet was devoid of any classical inflammasome characteristics including pyroptosome formation, pyroptosis induction, and K(+) efflux dependency. Genetic dissection of the underlying signaling pathway in a monocyte transdifferentiation system revealed that alternative inflammasome activation was propagated by TLR4-TRIF-RIPK1-FADD-CASP8 signaling upstream of NLRP3. Importantly, involvement of this signaling cascade was limited to alternative inflammasome activation and did not extend to classical NLRP3 activation. Because alternative inflammasome activation embraces both sensitivity and promiscuity of TLR4, we propose a pivotal role for this signaling cascade in TLR4-driven, IL-1ß-mediated immune responses and immunopathology in humans.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Monocytes/immunology , Toll-Like Receptor 4/immunology , Animals , Caspase 1/immunology , Cell Line , Cell Transdifferentiation/immunology , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism , Potassium Channels/immunology , Pyroptosis/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL