Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 19(9): 1061-1070, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759028

ABSTRACT

Cellular senescence is triggered by various distinct stresses and characterized by a permanent cell cycle arrest. Senescent cells secrete a variety of inflammatory factors, collectively referred to as the senescence-associated secretory phenotype (SASP). The mechanism(s) underlying the regulation of the SASP remains incompletely understood. Here we define a role for innate DNA sensing in the regulation of senescence and the SASP. We find that cyclic GMP-AMP synthase (cGAS) recognizes cytosolic chromatin fragments in senescent cells. The activation of cGAS, in turn, triggers the production of SASP factors via stimulator of interferon genes (STING), thereby promoting paracrine senescence. We demonstrate that diverse stimuli of cellular senescence engage the cGAS-STING pathway in vitro and we show cGAS-dependent regulation of senescence following irradiation and oncogene activation in vivo. Our findings provide insights into the mechanisms underlying cellular senescence by establishing the cGAS-STING pathway as a crucial regulator of senescence and the SASP.


Subject(s)
Cellular Senescence , Chromatin/enzymology , Cytosol/enzymology , Immunity, Innate , Nucleotidyltransferases/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cellular Senescence/radiation effects , Chromatin/immunology , Chromatin/radiation effects , Cytosol/immunology , Cytosol/radiation effects , Enzyme Activation , Female , Genotype , Immunity, Innate/radiation effects , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Oxidative Stress , Paracrine Communication , Phenotype , RNA Interference , Signal Transduction , Time Factors , Transfection
2.
Nat Immunol ; 17(5): 583-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26998763

ABSTRACT

Interleukin 1ß (IL-1ß) is critical for the in vivo survival, expansion and effector function of IL-17-producing helper T (T(H)17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1ß during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell-intrinsic inflammasome that drives IL-1ß production during T(H)17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1ß, whereas ATP stimulation triggered T cell production of IL-1ß via ASC-NLRP3-dependent caspase-8 activation. IL-1R was detected on T(H)17 cells but not on type 1 helper T (T(H)1) cells, and ATP-treated T(H)17 cells showed enhanced survival compared with ATP-treated T(H)1 cells, suggesting autocrine action of T(H)17-derived IL-1ß. Together these data reveal a critical role for IL-1ß produced by a T(H)17 cell-intrinsic ASC-NLRP3-caspase-8 inflammasome during inflammation of the central nervous system.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes/immunology , Th17 Cells/immunology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 8/genetics , Caspase 8/immunology , Caspase 8/metabolism , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Gene Expression/immunology , Immunoblotting , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
3.
PLoS One ; 8(5): e64256, 2013.
Article in English | MEDLINE | ID: mdl-23724040

ABSTRACT

Endoplasmic reticulum (ER) stress occurs when unfolded proteins accumulate in the lumen of the organelle, triggering signal transduction events that contribute either to cellular adaptation and recovery or alternatively to cellular dysfunction and death. ER stress has been implicated in numerous diseases. To identify novel modulators of ER stress, we undertook a siRNA library screen of the kinome, revealing Interleukin-1 Receptor-Associated Kinase-2 (IRAK2) as a contributor to unfolded protein response (UPR) signaling and ER stress-induced cell death. Knocking down expression of IRAK2 (but not IRAK1) in cultured mammalian cells suppresses ER stress-induced expression of the pro-apoptotic transcription factor CHOP and activation of stress kinases. Similarly, RNAi-mediated silencing of the IRAK family member Tube (but not Pelle) suppresses activation of stress kinase signaling induced by ER stress in Drosophila cells. The action of IRAK2 maps to the IRE1 pathway, rather than the PERK or ATF6 components of the UPR. Interestingly, ER stress also induces IRAK2 gene expression in an IRE1/XBP1-dependent manner, suggesting a mutually supporting amplification loop involving IRAK2 and IRE1. In vivo, ER stress induces Irak2 expression in mice. Moreover, Irak2 gene knockout mice display defects in ER stress-induced CHOP expression and IRE1 pathway signaling. These findings demonstrate an unexpected linkage of the innate immunity machinery to UPR signaling, revealing IRAK2 as a novel amplifier of the IRE1 pathway.


Subject(s)
Endoplasmic Reticulum Stress , Interleukin-1 Receptor-Associated Kinases/metabolism , Signal Transduction , Animals , Cell Death , Cell Line , Drosophila/cytology , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Sequence Homology, Amino Acid , Unfolded Protein Response
4.
J Neurosci ; 32(24): 8284-92, 2012 Jun 13.
Article in English | MEDLINE | ID: mdl-22699909

ABSTRACT

Cuprizone inhibits mitochondrial function and induces demyelination in the corpus callosum, which resembles pattern III lesions in multiple sclerosis patients. However, the molecular and cellular mechanism by which cuprizone induces demyelination remains unclear. Interleukin-17 (IL-17) secreted by T helper 17 cells and γδT cells are essential in the development of experimental autoimmune encephalomyelitis. In this study, we examined the importance of IL-17 signaling in cuprizone-induced demyelination. We found that mice deficient in IL-17A, IL-17 receptor C (IL-17RC), and adaptor protein Act1 (of IL-17R) all had reduced demyelination accompanied by lessened microglial and polydendrocyte cellular reactivity compared with that in wild-type mice in response to cuprizone feeding, demonstrating the essential role of IL-17-induced Act1-mediated signaling in cuprizone-induced demyelination. Importantly, specific deletion of Act1 in astrocytes reduced the severity of tissue injury in this model, indicating the critical role of CNS resident cells in the pathogenesis of cuprizone-induced demyelination. In cuprizone-fed mice, IL-17 was produced by CNS CD3(+) T cells, suggesting a source of IL-17 in CNS upon cuprizone treatment.


Subject(s)
Connexin 43/physiology , Cuprizone/toxicity , Demyelinating Diseases/physiopathology , Interleukin-17/physiology , Multiple Sclerosis/chemically induced , Multiple Sclerosis/physiopathology , Peptide Fragments/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , CD3 Complex/immunology , Chelating Agents , Connexin 43/genetics , Connexin 43/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Female , Gene Knockdown Techniques , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
5.
Immunity ; 36(5): 821-33, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608496

ABSTRACT

Interleukin-25 (IL-25 or IL-17E), a member of the structurally related IL-17 family, functions as an important mediator of T helper 2 cell-type (type 2) responses. We examined the cell type-specific role of IL-25-induced Act1-mediated signaling in protective immunity against helminth infection. Targeted Act1 deficiency in epithelial cells resulted in a marked delay in worm expulsion and abolished the expansion of the Lin(-)c-Kit(+) innate cell population in the mesenteric lymph node, lung, and liver. Th2 cell-inducing cytokine (IL-25 and IL-33) expression were reduced in the intestinal epithelial cells from the infected and IL-25-injected epithelial-specific Act1-deficient mice. Adoptive transfer of Lin(-)c-Kit(+) cells or combined injection of IL-25 and IL-33 restored the type 2 responses in these mice. Taken together, these results suggest that epithelial-specific Act1 mediates the expansion of the Lin(-)c-Kit(+) innate cell population through the positive-feedback loop of IL-25, initiating the type 2 immunity against helminth infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Epithelial Cells/immunology , Helminthiasis/immunology , Helminths/immunology , Interleukins/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Lineage , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Helminthiasis/metabolism , Helminths/metabolism , Immunity, Innate/immunology , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Liver/cytology , Liver/immunology , Liver/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Th2 Cells/metabolism
6.
Sci Signal ; 4(197): ra72, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22045852

ABSTRACT

Interleukin-17 (IL-17) and IL-25 signaling induce the expression of genes encoding inflammatory factors and are implicated in the pathology of various inflammatory diseases. Nuclear factor κB (NF-κB) activator 1 (Act1) is an adaptor protein and E3 ubiquitin ligase that is critical for signaling by either IL-17 or IL-25, and it is recruited to their receptors (IL-17R and IL-25R) through heterotypic interactions between the SEFIR [SEF (similar expression to fibroblast growth factor genes) and IL-17R] domain of Act1 and that of the receptor. SEFIR domains have structural similarity with the Toll-IL-1 receptor (TIR) domains of Toll-like receptors and IL-1R. Whereas the BB' loop of TIR is required for TIR-TIR interactions, we found that deletion of the BB' loop from Act1 or IL-17RA (a common subunit of both IL-17R and IL-25R) did not affect Act1-IL-17RA interactions; rather, deletion of the CC' loop from Act1 or IL-17RA abolished the interaction between both proteins. Surface plasmon resonance measurements showed that a peptide corresponding to the CC' loop of Act1 bound directly to IL-17RA. A cell-permeable decoy peptide based on the CC' loop sequence inhibited IL-17- or IL-25-mediated signaling in vitro, as well as IL-17- and IL-25-induced pulmonary inflammation in mice. Together, these findings provide the molecular basis for the specificity of SEFIR-SEFIR versus TIR-TIR domain interactions and consequent signaling. Moreover, we suggest that the CC' loop motif of SEFIR domains is a promising target for therapeutic strategies against inflammatory diseases associated with IL-17 or IL-25 signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Peptides/pharmacology , Pneumonia/prevention & control , Receptors, Interleukin-17/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , Cells, Cultured , Female , HEK293 Cells , HeLa Cells , Humans , Interleukin-17/toxicity , Interleukins/toxicity , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Mutation , Peptides/chemistry , Peptides/genetics , Pneumonia/chemically induced , Pneumonia/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, Interleukin-17/chemistry , Receptors, Interleukin-17/genetics , Signal Transduction/drug effects , Surface Plasmon Resonance
7.
J Immunol ; 187(6): 3155-64, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21856933

ABSTRACT

The cellular and molecular mechanisms driven by IL-25 and its cognate receptor IL-17RB necessary for the promotion of Th2-mediating pathogenic pulmonary inflammation remains to be defined. We have previously reported the critical role of the U-box-type E3 ubiquitin ligase Act1 (1) for the downstream signaling of the IL-17 cytokine family including the Th2-promoting cytokine IL-25 (IL-17E) (2). In this study, we report that IL-25-driven but not conventional IL-4-driven Th2 polarization and cytokine production is impaired in Act1-deficient T cells. Also, Act1 deficiency in the T cell compartment results in the abrogation of eosinophilic airway infiltration as well as airway hyperresponsiveness in mouse models of Ag-induced airway inflammation. The in vivo generation of Ag-specific Th2 cytokine-producing cells is defective in the absence of Act1 expression in T cells after OVA/aluminum hydroxide immunization. Notably, the production of OVA-specific IgG(1) but not IgG(2a) or IgE is also impaired. At the molecular level, we report that IL-25-mediated induction of Th2 master regulator GATA-3 and the transcription factor GFI-1 is attenuated in Act1-deficient T cells. Taken together, our findings indicate that Act1 expression in T cells is required for cellular and humoral Th2-mediated allergic responses and the development of airway hyperresponsiveness, in part, through Act1's function in IL-25-induced development of Th2 T cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Hypersensitivity/immunology , Interleukins/immunology , Pneumonia/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hypersensitivity/metabolism , Immunohistochemistry , Interleukins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Pneumonia/metabolism , Signal Transduction/immunology , Th2 Cells/metabolism
8.
Immunity ; 34(1): 3-5, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21272781

ABSTRACT

In this issue of Immunity, Shaw et al. (2011) report that the NOD-RICK signaling axis is required for the activation of dendritic cells infiltrating the central nervous system, leading to reactivation of antigen-specific T cells and autoimmune inflammation.

9.
Immunity ; 32(3): 414-25, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20303295

ABSTRACT

Interleukin-17 (IL-17) secreted by T helper 17 (Th17) cells is essential in the development of experimental autoimmune encephalomyelitis (EAE). However, it remains unclear how IL-17-mediated signaling in different cellular compartments participates in the central nervous system (CNS) inflammatory process. We examined CNS inflammation in mice with specific deletion of Act1, a critical component required for IL-17 signaling, in endothelial cells, macrophages and microglia, and neuroectoderm (neurons, astrocytes, and oligodendrocytes). In Act1-deficient mice, Th17 cells showed normal infiltration into the CNS but failed to recruit lymphocytes, neutrophils, and macrophages. Act1 deficiency in endothelial cells or in macrophages and microglia did not substantially impact the development of EAE. However, targeted Act1 deficiency in neuroectoderm-derived CNS-resident cells resulted in markedly reduced severity in EAE. Specifically, Act1-deficient astrocytes showed impaired IL-17-mediated inflammatory gene induction. Thus, astroctyes are critical in IL-17-Act1-mediated leukocyte recruitment during autoimmune-induced inflammation of the CNS.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Astrocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/metabolism , Cell Survival , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
10.
Immunity ; 26(4): 461-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17398123

ABSTRACT

Despite constant contact with the large population of commensal bacteria, the colonic mucosa is normally hyporesponsive to these potentially proinflammatory signals. Here we report that the single immunoglobulin IL-1 receptor-related molecule (SIGIRR), a negative regulator for Toll-IL-1R signaling, plays a critical role in gut homeostasis, intestinal inflammation, and colitis-associated tumorigenesis by maintaining the microbial tolerance of the colonic epithelium. SIGIRR-deficient (Sigirr(-/-)) colonic epithelial cells displayed commensal bacteria-dependent homeostatic defects, as shown by constitutive upregulation of inflammatory genes, increased inflammatory responses to dextran sulfate sodium (DSS) challenge, and increased Azoxymethane (AOM)+DSS-induced colitis-associated tumorigenesis. Gut epithelium-specific expression of the SIGIRR transgene in the SIGIRR-deficient background reduced the cell survival of the SIGIRR-deficient colon epithelium, abrogated the hypersensitivity of the Sigirr(-/-) mice to DSS-induced colitis, and reduced AOM+DSS-induced tumorigenesis. Taken together, our results indicate that epithelium-derived SIGIRR is critical in controlling the homeostasis and innate immune responses of the colon to enteric microflora.


Subject(s)
Cell Transformation, Neoplastic/immunology , Colitis/immunology , Colon/immunology , Colonic Neoplasms/immunology , Immunity, Innate/genetics , Receptors, Interleukin-1/physiology , Animals , Bacteria/immunology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Chemokines/genetics , Colitis/genetics , Colitis/pathology , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation , Homeostasis/genetics , Homeostasis/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , NF-kappa B/metabolism , Receptors, Interleukin-1/genetics , STAT3 Transcription Factor/metabolism
11.
Nat Immunol ; 8(3): 247-56, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17277779

ABSTRACT

T helper cells that produce interleukin 17 (IL-17) are associated with inflammation and the control of certain bacteria. We report here the essential involvement of the adaptor protein Act1 in IL-17 receptor (IL-17R) signaling and IL-17-dependent immune responses. After stimulation with IL-17, recruitment of Act1 to IL-17R required the IL-17R conserved cytoplasmic 'SEFIR' domain, followed by recruitment of the kinase TAK1 and E3 ubiquitin ligase TRAF6, which mediate 'downstream' activation of transcription factor NF-kappaB. IL-17-induced expression of inflammation-related genes was abolished in Act1-deficient primary astroglial and gut epithelial cells. This reduction was associated with much less inflammatory disease in vivo in both autoimmune encephalomyelitis and dextran sodium sulfate-induced colitis. Our data show that Act1 is essential in IL-17-dependent signaling in autoimmune and inflammatory disease.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Autoimmune Diseases , Inflammation/immunology , Interleukin-17/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adoptive Transfer , Animals , Autoimmunity , B-Cell Activation Factor Receptor/immunology , B-Cell Activation Factor Receptor/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation/immunology , HeLa Cells , Humans , Interleukin-17/metabolism , Mice , Receptors, Interleukin-17/immunology , Receptors, Interleukin-17/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Transfection , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins
12.
J Biol Chem ; 282(9): 6075-89, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17197697

ABSTRACT

Interleukin-1 (IL-1) receptor-associated kinase (IRAK) is phosphorylated after it is recruited to the receptor, subsequently ubiquitinated, and eventually degraded upon IL-1 stimulation. Although a point mutation changing lysine 134 to arginine (K134R) in IRAK abolished IL-1-induced IRAK ubiquitination and degradation, mutations of serines and threonines adjacent to lysine 134 to alanines ((S/T)A (131-144)) reduced IL-1-induced IRAK phosphorylation and abolished IRAK ubiquitination. Through the study of these IRAK modification mutants, we uncovered two parallel IL-1-mediated signaling pathways for NFkappaB activation, TAK1-dependent and MEKK3-dependent, respectively. These two pathways bifurcate at the level of IRAK modification. The TAK1-dependent pathway leads to IKKalpha/beta phosphorylation and IKKbeta activation, resulting in classical NFkappaB activation through IkappaBalpha phosphorylation and degradation. The TAK1-independent MEKK3-dependent pathway involves IKKgamma phosphorylation and IKKalpha activation, resulting in NFkappaB activation through IkappaBalpha phosphorylation and subsequent dissociation from NFkappaB but without IkappaBalpha degradation. These results provide significant insight to our further understanding of NFkappaB activation pathways.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1/pharmacology , MAP Kinase Kinase Kinase 3/physiology , MAP Kinase Kinase Kinases/physiology , NF-kappa B/metabolism , Animals , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Point Mutation , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...