Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34156982

ABSTRACT

The stimulator of IFN genes (STING) protein senses cyclic dinucleotides released in response to double-stranded DNA and functions as an adaptor molecule for type I IFN (IFNI) signaling by activating IFNI-stimulated genes (ISG). We found impaired T cell infiltration into the peritoneum in response to TNF-α in global and EC-specific STING-/- mice and discovered that T cell transendothelial migration (TEM) across mouse and human endothelial cells (EC) deficient in STING was strikingly reduced compared with control EC, whereas T cell adhesion was not impaired. STING-/- T cells showed no defect in TEM or adhesion to EC, or immobilized endothelial cell-expressed molecules ICAM1 and VCAM1, compared with WT T cells. Mechanistically, CXCL10, an ISG and a chemoattractant for T cells, was dramatically reduced in TNF-α-stimulated STING-/- EC, and genetic loss or pharmacologic antagonisms of IFNI receptor (IFNAR) pathway reduced T cell TEM. Our data demonstrate a central role for EC-STING during T cell TEM that is dependent on the ISG CXCL10 and on IFNI/IFNAR signaling.


Subject(s)
Interferon Type I , Membrane Proteins/immunology , Receptor, Interferon alpha-beta , T-Lymphocytes , Transendothelial and Transepithelial Migration/immunology , Animals , Immunity, Innate , Intercellular Adhesion Molecule-1/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/immunology
2.
Nat Commun ; 12(1): 86, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397971

ABSTRACT

Inflammation and cell death are closely linked arms of the host immune response to infection, which when carefully balanced ensure host survival. One example of this balance is the tightly regulated transition from TNFR1-associated pro-inflammatory complex I to pro-death complex II. By contrast, here we show that a TRIF-dependent complex containing FADD, RIPK1 and caspase-8 (that we have termed the TRIFosome) mediates cell death in response to Yersinia pseudotuberculosis and LPS. Furthermore, we show that constitutive binding between ZBP1 and RIPK1 is essential for the initiation of TRIFosome interactions, caspase-8-mediated cell death and inflammasome activation, thus positioning ZBP1 as an effector of cell death in the context of bacterial blockade of pro-inflammatory signaling. Additionally, our findings offer an alternative to the TNFR1-dependent model of complex II assembly, by demonstrating pro-death complex formation reliant on TRIF signaling.


Subject(s)
Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caspase 8/metabolism , Cell Death/drug effects , Mice, Inbred C57BL , Protein Binding/drug effects , Receptors, Tumor Necrosis Factor, Type I/metabolism , Yersinia
3.
Cell Rep ; 30(3): 699-713.e4, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968247

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) and 3 (RIPK3) are well known for their capacity to drive necroptosis via mixed-lineage kinase-like domain (MLKL). Recently, RIPK1/3 kinase activity has been shown to drive inflammation via activation of MAPK signaling. However, the regulatory mechanisms underlying this kinase-dependent cytokine production remain poorly understood. In the present study, we establish that the kinase activity of RIPK1/3 regulates cytokine translation in mouse and human macrophages. Furthermore, we show that this inflammatory response is downregulated by type I interferon (IFN) signaling, independent of type I IFN-promoted cell death. Specifically, low-level constitutive IFN signaling attenuates RIPK-driven activation of cap-dependent translation initiation pathway components AKT, mTORC1, 4E-BP and eIF4E, while promoting RIPK-dependent cell death. Altogether, these data characterize constitutive IFN signaling as a regulator of RIPK-dependent inflammation and establish cap-dependent translation as a crucial checkpoint in the regulation of cytokine production.


Subject(s)
Cytokines/metabolism , Interferons/metabolism , Protein Biosynthesis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cytokines/genetics , Down-Regulation , Eukaryotic Initiation Factor-4E/metabolism , Female , Humans , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
4.
Cell Death Differ ; 26(2): 332-347, 2019 01.
Article in English | MEDLINE | ID: mdl-29786074

ABSTRACT

Interferons (IFNs) are critical determinants in immune-competence and autoimmunity, and are endogenously regulated by a low-level constitutive feedback loop. However, little is known about the functions and origins of constitutive IFN. Recently, lipopolysaccharide (LPS)-induced IFN was implicated as a driver of necroptosis, a necrotic form of cell death downstream of receptor-interacting protein (RIP) kinase activation and executed by mixed lineage kinase like-domain (MLKL) protein. We found that the pre-established IFN status of the cell, instead of LPS-induced IFN, is critical for the early initiation of necroptosis in macrophages. This pre-established IFN signature stems from cytosolic DNA sensing via cGAS/STING, and maintains the expression of MLKL and one or more unknown effectors above a critical threshold to allow for MLKL oligomerization and cell death. Finally, we found that elevated IFN-signaling in systemic lupus erythematosus (SLE) augments necroptosis, providing a link between pathological IFN and tissue damage during autoimmunity.


Subject(s)
Interferon-beta/metabolism , Macrophages/metabolism , Necroptosis , Protein Kinases/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Cytosol/metabolism , DNA/metabolism , Gene Knockout Techniques , Humans , Interferon-beta/genetics , Interferon-beta/pharmacology , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
5.
Cell Rep ; 24(1): 155-168.e5, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972777

ABSTRACT

Legionella pneumophila elicits caspase-11-driven macrophage pyroptosis through guanylate-binding proteins (GBPs) encoded on chromosome 3. It has been proposed that microbe-driven IFN upregulates GBPs to facilitate pathogen vacuole rupture and bacteriolysis preceding caspase-11 activation. We show here that macrophage death occurred independently of microbial-induced IFN signaling and that GBPs are dispensable for pathogen vacuole rupture. Instead, the host-intrinsic IFN status sustained sufficient GBP expression levels to drive caspase-1 and caspase-11 activation in response to cytosol-exposed bacteria. In addition, endogenous GBP levels were sufficient for the release of DNA from cytosol-exposed bacteria, preceding the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) pathway for Ifnb induction. Mice deficient for chromosome 3 GBPs were unable to mount a rapid IL-1/chemokine (C-X-C motif) ligand 1 (CXCL1) response during Legionella-induced pneumonia, with defective bacterial clearance. Our results show that rapid GBP activity is controlled by host-intrinsic cytokine signaling and that GBP activities precede immune amplification responses, including IFN induction, inflammasome activation, and cell death.


Subject(s)
DNA, Bacterial/metabolism , GTP-Binding Proteins/metabolism , Interferons/metabolism , Legionella/metabolism , Pyroptosis , Animals , Anti-Infective Agents/pharmacology , Chromosomes, Mammalian/metabolism , Cytosol/metabolism , Female , Humans , Janus Kinases/metabolism , Legionellosis/microbiology , Macrophages/cytology , Male , Mice, Inbred C57BL , Pneumonia/microbiology , Pneumonia/pathology , Receptor, Interferon alpha-beta/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Vacuoles/metabolism
6.
J Immunol ; 199(2): 397-402, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28615418

ABSTRACT

Stimulator of interferon genes (STING) was initially described as a sensor of intracellular bacterial and viral DNA and a promising adjuvant target in innate immune cells; more recently STING has also been shown to detect endogenous DNA and play a role in tumor immunity and autoimmune disease development. Thus far STING has been studied in macrophages and dendritic cells. In this study, to our knowledge we provide the first evidence of STING activation in T cells, in which STING agonists not only provoke type I IFN production and IFN-stimulated gene expression, mirroring the response of innate cells, but are also capable of activating cell stress and death pathways. Our results suggest a re-evaluation of STING agonist-based therapies may be necessary to identify the possible effects on the T cell compartment. Conversely, the effects of STING on T cells could potentially be harnessed for therapeutic applications.


Subject(s)
Cell Death , Interferon Type I/biosynthesis , Interferon Type I/immunology , Membrane Proteins/metabolism , T-Lymphocytes/immunology , Animals , Endoplasmic Reticulum Stress , Immunity, Innate , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Sequence Analysis, RNA , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
7.
Proc Natl Acad Sci U S A ; 113(6): 1606-11, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26798068

ABSTRACT

cFLIP, an inhibitor of apoptosis, is a crucial regulator of cellular death by apoptosis and necroptosis; its importance in development is exemplified by the embryonic lethality in cFLIP-deficient animals. A homolog of caspase 8 (CASP8), cFLIP exists in two main isoforms: cFLIPL (long) and cFLIPR (short). Although both splice variants regulate death receptor (DR)-induced apoptosis by CASP8, the specific role of each isoform is poorly understood. Here, we report a previously unidentified model of resistance to Fas receptor-mediated liver failure in the wild-derived MSM strain, compared with susceptibility in C57BL/6 (B6) mice. Linkage analysis in F2 intercross (B6 x MSM) progeny identified several MSM loci controlling resistance to Fas-mediated death, including the caspase 8- and FADD-like apoptosis regulator (Cflar) locus encoding cFLIP. Furthermore, we identified a 21-bp insertion in the 3' UTR of the fifth exon of Cflar in MSM that influences differential splicing of cFLIP mRNA. Intriguingly, we observed that MSM liver cells predominantly express the FLIPL variant, in contrast to B6 liver cells, which have higher levels of cFLIPR. In keeping with this finding, genome-wide RNA sequencing revealed a relative abundance of FLIPL transcripts in MSM hepatocytes whereas B6 liver cells had significantly more FLIPR mRNA. Importantly, we show that, in the MSM liver, CASP8 is present exclusively as its cleaved p43 product, bound to cFLIPL. Because of partial enzymatic activity of the heterodimer, it might prevent necroptosis. On the other hand, it prevents cleavage of CASP8 to p10/20 necessary for cleavage of caspase 3 and, thus, apoptosis induction. Therefore, MSM hepatocytes are predisposed for protection from DR-mediated cell death.


Subject(s)
Alternative Splicing , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , fas Receptor/metabolism , Alternative Splicing/genetics , Animals , Antibodies , Apoptosis , Base Pairing/genetics , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Disease Susceptibility , Fas Ligand Protein/metabolism , Genetic Linkage , Genetic Loci , Genome , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis, Insertional/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Quantitative Trait, Heritable , Sequence Analysis, DNA , Signal Transduction
8.
J Immunol ; 196(2): 547-52, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26685207

ABSTRACT

With the stimulator of IFN genes (STING) C terminus being extensively studied, the role of the N-terminal domain (NTD) of STING remains an important subject of investigation. In this article, we identify novel mutations in NTD of Sting of the MOLF strain in response to HSV and Listeria monocytogenes both in vitro and in vivo. These mutations are responsible for low levels of IFN-ß caused by failure of MOLF STING to translocate from the endoplasmic reticulum. These data provide evidence that the NTD of STING affects DNA responses via control of trafficking. They also show that the genetic diversity of wild-derived mice resembles the diversity observed in humans. Several human alleles of STING confer attenuated IFN-I production similar to what we observe with the MOLF Sting allele, a crucial functional difference not apparent in classical inbred mice. Thus, understanding the functional significance of polymorphisms in MOLF STING can provide basic mechanistic insights relevant to humans.


Subject(s)
Interferon Type I/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Alleles , Animals , DNA/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Microscopy, Confocal , Mutation , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...