Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 277
Filter
1.
J Biol Chem ; 300(5): 107249, 2024 May.
Article in English | MEDLINE | ID: mdl-38556084

ABSTRACT

Tripartite-motif protein-56 (TRIM56) positively regulates the induction of type I interferon response via the TLR3 pathway by enhancing IRF3 activation and depends on its C-terminal residues 621-750 for interacting with the adaptor TRIF. However, the precise underlying mechanism and detailed TRIM56 determinants remain unclear. Herein, we show ectopic expression of murine TRIM56 also enhances TLR3-dependent interferon-ß promoter activation, suggesting functional conservation. We found that endogenous TRIM56 and TRIF formed a complex early (0.5-2 h) after poly-I:C stimulation and that TRIM56 overexpression also promoted activation of NF-κB by poly-I:C but not that by TNF-α or IL-1ß, consistent with a specific effect on TRIF prior to the bifurcation of NF-κB and IRF3. Using transient transfection and Tet-regulated cell lines expressing various TRIM56 mutants, we demonstrated the Coiled-coil domain and a segment spanning residues ∼434-610, but not the B-box or residues 355-433, were required for TRIM56 augmentation of TLR3 signaling. Moreover, alanine substitution at each putative phosphorylation site, Ser471, Ser475, and Ser710, abrogated TRIM56 function. Concordantly, mutants bearing Ser471Ala, Ser475Ala, or Ser710Ala, or lacking the Coiled-coil domain, all lost the capacity to enhance poly-I:C-induced establishment of an antiviral state. Furthermore, the Ser710Ala mutation disrupted the TRIM56-TRIF association. Using phospho-specific antibodies, we detected biphasic phosphorylation of TRIM56 at Ser471 and Ser475 following TLR3 stimulation, with the early phase occurring at ∼0.5 to 1 h, prior to IRF3 phosphorylation. Together, these data reveal novel molecular details critical for the TRIM56 augmentation of TLR3-dependent antiviral response and highlight important roles for TRIM56 scaffolding and phosphorylation.


Subject(s)
Adaptor Proteins, Vesicular Transport , Immunity, Innate , Poly I-C , Toll-Like Receptor 3 , Tripartite Motif Proteins , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Phosphorylation , Animals , Humans , Mice , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Poly I-C/pharmacology , Protein Domains , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , HEK293 Cells , NF-kappa B/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
2.
J Virol ; 96(18): e0121222, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36069553

ABSTRACT

The mitochondrial apoptosis pathway has the function to kill the cell, but recent work shows that this pathway can also be activated to a sublethal level, where signal transduction can be observed but the cell survives. Intriguingly, this signaling has been shown to contribute to inflammatory activity of epithelial cells upon infection with numerous agents. This suggests that microbial recognition can generate sublethal activity in the mitochondrial apoptosis pathway. Because this recognition is achieved by pattern recognition receptors (PRRs), it also implies that PRR signals are linked to the mitochondrial apoptosis apparatus. We here test this hypothesis during infection of epithelial cells with modified vaccinia virus Ankara (MVA). MVA recognition is achieved through receptors specific for nucleic acids, and we present evidence that the three receptors, Toll-like receptor 3 (TLR3), RIG-I/MDA5, and cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING), are involved in this signaling. When stimulated directly by specific ligands, all three receptors could trigger sublethal apoptosis signals. During infection with MVA, sublethal apoptosis signals were unmasked in X-linked IAP (XIAP)-deficient cells, where apoptosis induction was observed. Deletion of any of the three signaling adapters, TRIF, MAVS, and STING, reduced the DNA damage response, a sensitive measure of sublethal apoptosis signals. Our results suggest that PRRs signal via mitochondria, where they generate sublethal signals through the BCL-2-family, which may contribute to the response to infectious agents. IMPORTANCE A contribution of the mitochondrial apoptosis apparatus, in the absence of cell death, to the reaction of nonprofessional immune cells to viruses is suggested to play a role as a broad alert system of an infected cell: the apoptosis system can be activated by many upstream signals and could therefore act as a central coordinator of viral recognition. The proapoptotic activity of PRRs has been documented in multiple situations, but this activity seems too low to be meaningful, and a physiological significance of such activity is not immediately obvious. This work suggests the alternative interpretation that PRRs do not have the primary function to induce apoptosis but to trigger sublethal signals in the apoptosis system. A number of lines of recent research suggest that mitochondria contribute to cellular reactions, and this pathway may be a way of triggering an early host response.


Subject(s)
Apoptosis , Mitochondria , Nucleic Acids , Receptors, Pattern Recognition , Virus Diseases , Adaptor Proteins, Vesicular Transport/immunology , Humans , Immunity, Innate , Mitochondria/immunology , Nucleotidyltransferases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Pattern Recognition/immunology , Toll-Like Receptor 3/metabolism , Vaccinia virus , Virus Diseases/immunology
3.
Mol Immunol ; 143: 7-16, 2022 03.
Article in English | MEDLINE | ID: mdl-34990938

ABSTRACT

DDX43 is one of the members of the DExD/H-box protein family, and emerging data suggest that it may play an important role in antiviral immunity across mammals. However, little is known about DDX43 in the fish immune response. In this study, we isolated the cDNA sequence of ddx43 in Nile tilapia (Oreochromis niloticus). The ddx43 gene was 2338 bp in length, contained an open reading frame (ORF) of 2064 bp and encoded a polypeptide of 687 amino acids. The predicted protein of OnDDX43 has three conserved domains, including the RNA binding domain KH, DEAD-like helicase superfamily DEXDc and C-terminal HELICc domain. In healthy Nile tilapia, the Onddx43 transcript was broadly expressed in all examined tissues, with the highest expression levels in the muscle and brain and the lowest in the liver. After challenge with Streptococcus agalactiae, lipopolysaccharides (LPS) and polyinosinic polycytidylic acid (Poly I:C), the expression level of Onddx43 mRNA was upregulated or downregulated in all of the tissues tested. Overexpression of OnDDX43 in 293 T cells showed that it has a positive regulatory effect on IFN-ß. The subcellular localization showed that OnDDX43 was expressed in the cytoplasm. We performed further pull-down assays and found that OnDDX43 interacted with both interferon-ß promoter stimulator1 (IPS-1) and TIR domain-containing adaptor inducing interferon-ß (TRIF).


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/immunology , Cichlids/immunology , DEAD-box RNA Helicases/immunology , Fish Diseases/immunology , Fish Proteins/immunology , Interferon-beta/immunology , Signal Transduction/immunology , Animals , Cichlids/microbiology
4.
Neurosci Lett ; 768: 136374, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34852285

ABSTRACT

Macrophage-dominated inflammation by the activation of Toll-like receptor (TLR) pathway leads to neurological disruption after spinal cord injury (SCI). Regulator of G-protein signaling 1 (RGS1) is reported to be a regulator in inflammation. The present study thus purposes to identify the unknown role of RGS1 mediating TLR on inflammation post SCI. A mouse model of traumatic SCI was established by a mechanical trauma at T10. The mice underwent SCI and a macrophage line activated by lipopolysaccharide (LPS) were treated with shRNA-RGS1 to elucidate the role of RGS1 in inflammatory progression. The inflammatory factors were measured, and the degree of histology and function protection were determined. The expression levels of RGS1, myeloid differentiation primary response protein 88 (Myd88), (TIR-domain-containing adaptor inducing interferon-ß (TRIF), p38, metalloproteinase (MMP)-2, and MMP-9 were determined. RGS1 was robustly increased both in LPS-activated macrophage and SCI mice. The TLR signaling pathway-induced inflammation was suppressed by RGS1 knockdown. shRNA-mediated silence of RGS1 was exhibited a prominent decrease in TNF-α, IL-1ß and IL-6 via TLR/TRIF/ nuclear factor kappa-B (NF-κB) axis. Depletion of RGS1 also inhibited MMP-induced tissue degradation via MAPK-p38 pathway in SCI mice. Moreover, suppression of RGS1 improved spinal cord histology and function recovery. These findings suggest that RGS1 regulates inflammation and tissue disruption via TLR/TRIF/NF-κB signaling pathway in mice with SCI, thereby explaining a novel target that regulates macrophage inflammation post SCI.


Subject(s)
Inflammation/immunology , Macrophages/immunology , RGS Proteins/immunology , Signal Transduction/immunology , Spinal Cord Injuries/immunology , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , RAW 264.7 Cells , RGS Proteins/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
5.
Front Immunol ; 12: 734520, 2021.
Article in English | MEDLINE | ID: mdl-34956174

ABSTRACT

Upon recognition of bacterial or viral components by Toll-like receptors (TLRs), cells could be activated to induce a series of reactions to produce inflammatory cytokines, type I interferon (IFN), and IFN stimulating genes (ISG). MicroRNAs (miRNAs) are an important regulatory molecules that are widely involved in the regulatory networks of mammalian inflammation and immune responses; however, in lower vertebrates, the regulatory network of miRNA-mediated immune responses is poorly understood. Here, we report two miRNAs form Miichthys miiuy, namely, miR-181b-2 and miR-21-1, that play a negative role in host antiviral and antibacterial immunity. We found that miR-181b-2 and miR-21-1 are abundantly expressed in gram-negative bacteria, as well as RNA rhabdovirus infection. Inducible miR-181b-2 and miR-21-1 suppress the production of inflammatory cytokines and type I IFN by targeting TRIF, thereby avoiding excessive inflammation. We further revealed that miR-181b-2 and miR-21-1 modulate antibacterial and antiviral immunity through the TRIF-mediated NF-κB and IRF3 signaling pathways. The overall results indicate that miR-181b-2 and miR-21-1 act as negative feedback regulators and participate in host antibacterial and antiviral immune responses; this finding could provide information for a deeper understanding of the resistance of lower vertebrates to the invasion of pathogens and to avoidance of excessive immunity.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Interferon Regulatory Factor-3/immunology , MicroRNAs/immunology , NF-kappa B/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/immunology , MicroRNAs/genetics , Perciformes , RNA, Messenger/genetics , RNA, Messenger/immunology
6.
Front Immunol ; 12: 752898, 2021.
Article in English | MEDLINE | ID: mdl-34630431

ABSTRACT

The Toll/interleukin-1 receptor (TIR) domain is the signature signalling motif of innate immunity, with essential roles in innate immune signalling in bacteria, plants, and animals. TIR domains canonically function as scaffolds, with stimulus-dependent multimerization generating binding sites for signalling molecules such as kinases and ligases that activate downstream immune mechanisms. Recent studies have dramatically expanded our understanding of the TIR domain, demonstrating that the primordial function of the TIR domain is to metabolize NAD+. Mammalian SARM1, the central executioner of pathological axon degeneration, is the founding member of the TIR-domain class of NAD+ hydrolases. This unexpected NADase activity of TIR domains is evolutionarily conserved, with archaeal, bacterial, and plant TIR domains all sharing this catalytic function. Moreover, this enzymatic activity is essential for the innate immune function of these proteins. These evolutionary relationships suggest a link between SARM1 and ancient self-defense mechanisms that has only been strengthened by the recent discovery of the SARM1 activation mechanism which, we will argue, is strikingly similar to bacterial toxin-antitoxin systems. In this brief review we will describe the regulation and function of SARM1 in programmed axon self-destruction, and highlight the parallels between the SARM1 axon degeneration pathway and bacterial innate immune mechanisms.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Armadillo Domain Proteins/immunology , Cytoskeletal Proteins/immunology , Immunity, Innate/immunology , NAD+ Nucleosidase/immunology , Animals , Bacteriophages/immunology , Biological Evolution , Humans , Toxin-Antitoxin Systems/immunology
7.
Front Immunol ; 12: 725150, 2021.
Article in English | MEDLINE | ID: mdl-34504499

ABSTRACT

TRIF, an important adaptor downstream of Toll-like receptor signaling, plays a critical role in the innate immune response. In this study, the full-length coding sequence of TRIF from common carp (Cyprinus carpio L.) was cloned and characterized. Bioinformatics analysis showed that common carp TRIF exhibited a conserved TIR domain and had the closest relationship with grass carp TRIF. Expression analysis revealed that TRIF was constitutively expressed in the examined tissues of common carp, with the highest expression in the spleen and the lowest expression in the head kidney, and could be upregulated under Aeromonas hydrophila and poly(I:C) stimulation in vivo and under poly(I:C), LPS, PGN, flagellin, and Pam3CSK4 stimulation in vitro. Laser confocal microscopy showed that common carp TRIF colocalized with the Golgi apparatus. A luciferase reporter assay showed that carp TRIF elicited the activity of ifn-1 and nf-κb through the C-terminal domain. Additionally, crystal violet staining and qPCR assays revealed that carp TRIF inhibited the replication of SVCV in epithelioma papulosum cyprini (EPC) cells. Then, the signaling downstream of carp TRIF was investigated. Coimmunoprecipitation and Western blotting analysis demonstrated that carp TRIF interacted with TBK1 and augmented the expression of TRAF6 and phosphorylation of TBK1. Overexpression of carp TRIF significantly enhanced the expression of interferon-stimulated genes and inflammatory cytokines. Furthermore, flow cytometric (FCM) analysis suggested that carp TRIF induced apoptosis through the activation of caspase-8. In summary, our study indicated that TRIF plays an essential role in the innate immune responses of common carp against bacterial and viral infection.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Carps/immunology , Immunity, Innate , Interferons/immunology , NF-kappa B/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Apoptosis , Carps/genetics , Signal Transduction
8.
J Zhejiang Univ Sci B ; 22(8): 609-632, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34414698

ABSTRACT

Toll-like receptor 3 (TLR3) is a member of the TLR family, mediating the transcriptional induction of type I interferons (IFNs), proinflammatory cytokines, and chemokines, thereby collectively establishing an antiviral host response. Studies have shown that unlike other TLR family members, TLR3 is the only RNA sensor that is utterly dependent on the Toll-interleukin-1 receptor (TIR)|-domain-containing adaptor-inducing IFN-|ß (TRIF). However, the details of how the TLR3-TRIF signaling pathway works in an antiviral response and how it is regulated are unclear. In this review, we focus on recent advances in understanding the antiviral mechanism of the TRIF pathway and describe the essential characteristics of TLR3 and its antiviral effects. Advancing our understanding of TLR3 may contribute to disease diagnosis and could foster the development of novel treatments for viral diseases.


Subject(s)
Antiviral Restriction Factors/immunology , Immunity, Innate , Toll-Like Receptor 3/immunology , Adaptor Proteins, Vesicular Transport/immunology , Humans , Signal Transduction
9.
J Immunol ; 206(11): 2682-2691, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34021047

ABSTRACT

Type I/III IFNs induce expression of hundreds of IFN-stimulated genes through the JAK/STAT pathway to combat viral infections. Although JAK/STAT signaling is seemingly straightforward, it is nevertheless subjected to complex cellular regulation. In this study, we show that an ubiquitination regulatory X (UBX) domain-containing protein, UBXN6, positively regulates JAK-STAT1/2 signaling. Overexpression of UBXN6 enhanced type I/III IFNs-induced expression of IFN-stimulated genes, whereas deletion of UBXN6 inhibited their expression. RNA viral replication was increased in human UBXN6-deficient cells, accompanied by a reduction in both type I/III IFN expression, when compared with UBXN6-sufficient cells. Mechanistically, UBXN6 interacted with tyrosine kinase 2 (TYK2) and inhibited IFN-ß-induced degradation of both TYK2 and type I IFNR. These results suggest that UBXN6 maintains normal JAK-STAT1/2 signaling by stabilizing key signaling components during viral infection.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Autophagy-Related Proteins/immunology , Janus Kinases/immunology , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/immunology , Animals , Cells, Cultured , Chlorocebus aethiops , Humans , Signal Transduction/immunology
10.
J Med Virol ; 93(9): 5376-5389, 2021 09.
Article in English | MEDLINE | ID: mdl-33913550

ABSTRACT

The suppression of types I and III interferon (IFN) responses by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contributes to the pathogenesis of coronavirus disease 2019 (COVID-19). The strategy used by SARS-CoV-2 to evade antiviral immunity needs further investigation. Here, we reported that SARS-CoV-2 ORF9b inhibited types I and III IFN production by targeting multiple molecules of innate antiviral signaling pathways. SARS-CoV-2 ORF9b impaired the induction of types I and III IFNs by Sendai virus and poly (I:C). SARS-CoV-2 ORF9b inhibited the activation of types I and III IFNs induced by the components of cytosolic dsRNA-sensing pathways of RIG-I/MDA5-MAVS signaling, including RIG-I, MDA-5, MAVS, TBK1, and IKKε, rather than IRF3-5D, which is the active form of IRF3. SARS-CoV-2 ORF9b also suppressed the induction of types I and III IFNs by TRIF and STING, which are the adaptor protein of the endosome RNA-sensing pathway of TLR3-TRIF signaling and the adaptor protein of the cytosolic DNA-sensing pathway of cGAS-STING signaling, respectively. A mechanistic analysis revealed that the SARS-CoV-2 ORF9b protein interacted with RIG-I, MDA-5, MAVS, TRIF, STING, and TBK1 and impeded the phosphorylation and nuclear translocation of IRF3. In addition, SARS-CoV-2 ORF9b facilitated the replication of the vesicular stomatitis virus. Therefore, the results showed that SARS-CoV-2 ORF9b negatively regulates antiviral immunity and thus facilitates viral replication. This study contributes to our understanding of the molecular mechanism through which SARS-CoV-2 impairs antiviral immunity and provides an essential clue to the pathogenesis of COVID-19.


Subject(s)
DEAD Box Protein 58/immunology , Immune Evasion/genetics , Interferons/immunology , Nucleotidyltransferases/immunology , Receptors, Immunologic/immunology , SARS-CoV-2/immunology , Toll-Like Receptor 3/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/immunology , DEAD Box Protein 58/genetics , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferons/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Nucleotidyltransferases/genetics , Phosphoproteins/genetics , Phosphoproteins/immunology , Plasmids/chemistry , Plasmids/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptors, Immunologic/genetics , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/genetics , Transfection , Vero Cells , Virus Replication/immunology
11.
Dev Comp Immunol ; 117: 103991, 2021 04.
Article in English | MEDLINE | ID: mdl-33387560

ABSTRACT

Toll/interleukin 1 receptor domain-containing adaptor protein (TIRAP) and toll/interleukin 1 receptor-domain-containing adapter-inducing interferon-ß (TRIF) are crucial adaptors of signal transduction for the signaling pathways of toll-like receptors (TLRs). TIRAP and TRIF perform an essential function in an antimicrobial immune response; however, their function in Nile tilapia remains unknown. Herein, TIRAP and TRIF from Nile tilapia were identified and functionally characterized. Phylogenetic analysis showed that OnTIRAP and OnTRIF clustered with corresponding homologs from other fish species, with comparable gene structures to those of select vertebrate TIRAP and TRIF genes, respectively. The expression profiles of OnTIRAP and OnTRIF were broadly distributed in the ten tissues investigated, with high transcript levels noticed in immune organs. The transcription levels of OnTIRAP and OnTRIF were upregulated in response to bacterial and poly (I:C) challenges. GFP signals were only detected in the cytoplasmic region of fish cells transfected with OnTIRAP-GFP and OnTRIF-GFP expression plasmids. Moreover, overexpression of OnTIRAP and OnTRIF activated interferon-ß (IFN-ß) and activator protein 1 (AP1) reporters in HEK 293 cells. Activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) reporter was only observed in OnTRIF-overexpressing HEK 293 cells. Furthermore, the results of the co-immunoprecipitation analysis showed that OnTRIF, but not OnTIRAP, was recruited as an adaptor protein by OnTLR25. This study provides the first evidence on the functions of OnTIRAP and OnTRIF in the immune system of Nile tilapia against pathogens and may serve as the basis for further investigations on TLR signaling in fish.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/immunology , Cichlids/immunology , Fish Proteins/immunology , Gene Expression/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/classification , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/classification , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Sequence , Animals , Bacteria/immunology , Bacteria/pathogenicity , Cell Line , Cichlids/genetics , Cichlids/microbiology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Profiling/methods , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Phylogeny , Sequence Homology, Amino Acid , Signal Transduction/genetics , Virulence/immunology
12.
Blood ; 135(14): 1087-1100, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32016282

ABSTRACT

Bacterial infection not only stimulates innate immune responses but also activates coagulation cascades. Overactivation of the coagulation system in bacterial sepsis leads to disseminated intravascular coagulation (DIC), a life-threatening condition. However, the mechanisms by which bacterial infection activates the coagulation cascade are not fully understood. Here we show that type 1 interferons (IFNs), a widely expressed family of cytokines that orchestrate innate antiviral and antibacterial immunity, mediate bacterial infection-induced DIC by amplifying the release of high-mobility group box 1 (HMGB1) into the bloodstream. Inhibition of the expression of type 1 IFNs and disruption of their receptor IFN-α/ßR or downstream effector (eg, HMGB1) uniformly decreased gram-negative bacteria-induced DIC. Mechanistically, extracellular HMGB1 markedly increased the procoagulant activity of tissue factor by promoting the externalization of phosphatidylserine to the outer cell surface, where phosphatidylserine assembles a complex of cofactor-proteases of the coagulation cascades. These findings not only provide novel insights into the link between innate immune responses and coagulation, but they also open a new avenue for developing novel therapeutic strategies to prevent DIC in sepsis.


Subject(s)
Disseminated Intravascular Coagulation/immunology , Endotoxemia/immunology , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Adaptor Proteins, Vesicular Transport/immunology , Animals , Blood Coagulation , Disseminated Intravascular Coagulation/blood , Disseminated Intravascular Coagulation/etiology , Endotoxemia/blood , Endotoxemia/complications , Gram-Negative Bacterial Infections/blood , Gram-Negative Bacterial Infections/complications , HMGB1 Protein/blood , HMGB1 Protein/immunology , Humans , Immunity, Innate , Mice, Inbred C57BL
13.
Intern Emerg Med ; 15(3): 381-393, 2020 04.
Article in English | MEDLINE | ID: mdl-31919781

ABSTRACT

Experimental and clinical studies aimed at investigating the mechanism(s) underlying vascular complications of diabetes indicate that a great number of molecules are involved in the pathogenesis of these complications. Most of these molecules are inflammatory mediators or markers generated by immune or adipose tissue. Some of them, i.e. resistin and sortilin, have been shown to be involved in the cross talk between adipocytes and inflammatory cells. This interaction is an attractive area of research, particularly in type 2 diabetes and obesity. Other proteins, such as adiponectin and visfatin, appear to be more promising as possible vascular markers. In addition, some molecules involved in calcium/phosphorus metabolism, such as klotho and FGF23, have an involvement in the pathogenesis of diabetic vasculopathy, which appears to be dependent on the degree of vascular impairment. Inflammatory markers are a promising tool for treatment decisions while measuring plasma levels of adipokines, sortilin, Klotho and FGF23 in adequately sized longitudinal studies is expected to allow a more precise characterization of diabetic vascular disease and the optimal use of personalized treatment strategies.


Subject(s)
Adipose Tissue/immunology , Biomarkers/analysis , Cardiovascular Diseases/diagnosis , Immune System/immunology , Signal Transduction/immunology , Adaptor Proteins, Vesicular Transport/analysis , Adaptor Proteins, Vesicular Transport/blood , Adaptor Proteins, Vesicular Transport/immunology , Adipokines/analysis , Adipokines/blood , Adipokines/immunology , Adipose Tissue/physiopathology , Biomarkers/blood , C-Reactive Protein/analysis , C-Reactive Protein/immunology , Cardiovascular Diseases/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Exosomes/immunology , Fibroblast Growth Factor-23 , Glucuronidase/analysis , Glucuronidase/blood , Glucuronidase/immunology , HMGB Proteins/analysis , HMGB Proteins/blood , HMGB Proteins/immunology , Humans , Immune System/physiopathology , Interleukin-1/analysis , Interleukin-1/blood , Interleukin-1/immunology , Klotho Proteins , Osteoprotegerin/analysis , Osteoprotegerin/blood , Osteoprotegerin/immunology , Prevalence , Serum Amyloid P-Component/analysis , Serum Amyloid P-Component/immunology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
14.
Front Immunol ; 11: 581111, 2020.
Article in English | MEDLINE | ID: mdl-33519804

ABSTRACT

Postoperative ileus (POI) is triggered by an innate immune response in the muscularis externa (ME) and is accompanied by bacterial translocation. Bacteria can trigger an innate immune response via toll-like receptor (TLR) activation, but the latter's contribution to POI has been disproved for several TLRs, including TLR2 and TLR4. Herein we investigated the role of double-stranded RNA detection via TLR3 and TIR-domain-containing adapter-inducing interferon-ß (TRIF) signaling pathway in POI. POI was induced by small bowel intestinal manipulation in wt, TRIF-/-, TLR3-/-, type I interferon receptor-/- and interferon-ß reporter mice, all on C57BL/6 background, and POI severity was quantified by gene expression analysis, gastrointestinal transit and leukocyte extravasation into the ME. TRIF/TLR3 deficiency reduced postoperative ME inflammation and prevented POI. With bone marrow transplantation, RNA-sequencing, flow cytometry and immunohistochemistry we revealed a distinct TLR3-expressing radio-resistant MHCIIhiCX3CR1- IBA-1+ resident macrophage population within the deep myenteric plexus. TLR3 deficiency in these cells, but not in MHCIIhiCX3CR1+ macrophages, reduced cytokine expression in POI. While this might not be an exclusive macrophage-privileged pathway, the TLR3/TRIF axis contributes to proinflammatory cytokine production in MHCIIhiCX3CR1- IBA-1+ macrophages during POI. Deficiency in TLR3/TRIF protects mice from POI. These data suggest that TLR3 antagonism may prevent POI in humans.


Subject(s)
Ileus/etiology , Macrophages/immunology , Postoperative Complications/etiology , Toll-Like Receptor 3/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Disease Models, Animal , Female , Gene Expression , Ileus/immunology , Ileus/pathology , Immunity, Innate , Macrophages/classification , Macrophages/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myenteric Plexus/immunology , Postoperative Complications/immunology , Postoperative Complications/pathology , Radiation Tolerance/immunology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Transplantation Chimera/immunology
15.
Fish Shellfish Immunol ; 97: 114-124, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31841694

ABSTRACT

As a member of tumor necrosis factor receptor (TNFR)-associated factor (TRAF) family, TRAF3 is an important regulator of NF-κB and type I interferon (IFN) activation, especially in Toll-like receptors (TLRs)- and retinoic acid inducible gene I (RIG-I)-like receptors (RLRs)-mediated signaling pathway. In the present study, a TRAF3 homologue named Lc-TRAF3 was characterized in large yellow croaker (Larimichthys crocea). The open reading frame (ORF) of Lc-TRAF3 contains 1788 bp encoding a protein of 595 amino acids (aa). Sequence analysis indicated that Lc-TRAF3 is conserved in vertebrates, constituted with a N-terminal RING finger, two TRAF-type zinc fingers, and a C-terminal TRAF-MATH domain. The genome organization of Lc-TRAF3 is conserved in fish, with 13 exons and 12 introns, but different from that in birds or mammals, which contains 10 exons and 9 introns. Lc-TRAF3 was identified as cytosolic protein base on fluorescence microscopy analysis. Expression analysis revealed that Lc-TRAF3 was broadly distributed in examined organs/tissues, with the highest expression level in gill and weakest in brain, and could be up-regulated under poly I:C, LPS, PGN, and Pseudomonas plecoglossicida stimulation in vivo. Interestingly, overexpression Lc-TRAF3 could induce the activation of NF-κB, and Lc-TRAF3 co-transfected with Lc-TRIF induced a significantly higher level of NF-κB and IRF3 promoter activity, implying that Lc-TRAF3 may function as an enhancer in Lc-TRIF-mediated signaling pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Interferon Regulatory Factors/genetics , NF-kappa B/metabolism , Perciformes/immunology , Signal Transduction , TNF Receptor-Associated Factor 3/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Bacteria/immunology , Interferon Regulatory Factors/immunology , NF-kappa B/immunology , Perciformes/genetics , Perciformes/microbiology , TNF Receptor-Associated Factor 3/immunology
16.
Immun Inflamm Dis ; 7(4): 318-325, 2019 12.
Article in English | MEDLINE | ID: mdl-31691534

ABSTRACT

INTRODUCTION: Complexins (CPLXs), initially identified in neuronal presynaptic terminals, are cytoplasmic proteins that interact with the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) complex to regulate the fusion of vesicles to the plasma membrane. Although much is known about CPLX function in neuronal synaptic vesicle exocytosis, their distribution and role in immune cells are still unclear. In this study, we investigated CPLX2 knockout (KO) mice to reveal the role of CPLXs in exocytosis of lymphocytes. METHODS: We examined the expression of CPLXs and SNAREs in lymphocytes. To study the effect of CPLXs on the immune system in vivo, we analyzed the immune phenotype of CPLX2 KO mice. Furthermore, antibodies secretion from the peritoneal cavity, spleen, and bone marrow cells of wild-type (WT) and CPLX2 KO mice were determined. RESULTS: CPLX2 was detected in B cells but not in T cells, while other CPLXs and SNAREs were expressed at a similar level in both B and T cells. To clarify the function of CPLX2 in B lymphocytes, serum concentrations of immunoglobulin G (IgG), IgA, IgM, and IgE were measured in WT and CPLX2 KO mice using enzyme-linked immunosorbent assay. The level of IgM, which mainly consists of natural antibodies, was higher in KO mice than that in WT mice, while the levels of other antibodies were similar in both types of mice. Additionally, we found that spontaneous secretion of IgM and IgG1 was enhanced from the splenic antibody-secreting cells (ASCs) of CPLX2 KO mice. CONCLUSION: Our data suggest that CPLX2 inhibits spontaneous secretion of IgM and IgG1 from splenic ASCs. This study provides new insight into the mechanism of antibody secretion of ASCs.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , B-Lymphocytes/immunology , Immunoglobulins/immunology , Nerve Tissue Proteins/immunology , Spleen/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , B-Lymphocytes/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , SNARE Proteins/genetics , SNARE Proteins/immunology , Spleen/cytology
17.
Front Immunol ; 10: 2202, 2019.
Article in English | MEDLINE | ID: mdl-31620127

ABSTRACT

DExD/H-box helicases play essential roles in RNA metabolism, and emerging data suggests that they have additional functions in antiviral immunity across species. However, little is known about this evolutionarily conserved family in antiviral responses in lower species. Here, through isolation of poly(I:C)-binding proteins in amphioxus, an extant basal chordate, we found that DExD/H-box helicases DHX9, DHX15, and DDX23 are responsible for cytoplasmic dsRNA detection in amphioxus. Since the antiviral roles of DDX23 have not been characterized in mammals, we performed further poly(I:C) pull-down assays and found that human DDX23 binds to LMW poly(I:C) through its N-terminal region, suggesting that DDX23 is an evolutionarily conserved dsRNA sensor. Knockdown of human DDX23 enhanced the replication of VSV and reduced the activation of the NF-κB and IRF3. Moreover, when stimulated with poly(I:C) or VSV, human DDX23 translocated from the nucleus to the cytoplasm and formed complexes with TRIF or MAVS to initiate downstream signaling. Collectively, this comparative immunological study not only defined DDX23 as an emerging nuclear pattern recognition receptor (PRR) for the innate sensing of an RNA virus, but also extended the essential role of the DExD/H helicase family in viral RNA sensing from mammals to basal chordates.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/immunology , DEAD-box RNA Helicases/immunology , Immunity, Innate/immunology , A549 Cells , Animals , Biological Evolution , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Lancelets , Poly I-C/immunology , RNA Viruses/immunology , RNA, Double-Stranded
18.
Int J Mol Sci ; 20(18)2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31500298

ABSTRACT

TIR domain-containing proteins are essential for bacterial pathogens to subvert host defenses. This study describes a fish pathogen, Yersinia ruckeri SC09 strain, with a novel TIR domain-containing protein (STIR-2) that affects Toll-like receptor (TLR) function. STIR-2 was identified in Y. ruckeri by bioinformatics analysis. The toxic effects of this gene on fish were determined by in vivo challenge experiments in knockout mutants and complement mutants of the stir-2 gene. In vitro, STIR-2 downregulated the expression and secretion of IL-6, IL-1ß, and TNF-α. Furthermore, the results of NF-κB-dependent luciferase reporter system, co-immunoprecipitation, GST pull-down assays, and yeast two-hybrid assay indicated that STIR-2 inhibited the TLR signaling pathway by interacting with myeloid differentiation factor 88 (MyD88). In addition, STIR-2 promoted the intracellular survival of pathogenic Yersinia ruckeri SC09 strain by binding to the TIR adaptor protein MyD88 and inhibiting the pre-inflammatory signal of immune cells. These results showed that STIR-2 increased virulence in Y. ruckeri and suppressed the innate immune response by inhibiting TLR and MyD88-mediated signaling, serving as a novel strategy for innate immune evasion.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Fish Diseases/microbiology , Myeloid Differentiation Factor 88/metabolism , Yersinia Infections/veterinary , Yersinia ruckeri/pathogenicity , Adaptor Proteins, Vesicular Transport/immunology , Animals , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Fish Diseases/immunology , Fish Proteins/metabolism , Gene Expression Regulation , Immune Evasion , Mice, Knockout , Oncorhynchus mykiss , Protein Domains , Signal Transduction , Toll-Like Receptors/metabolism , Virulence Factors/genetics , Virulence Factors/immunology , Yersinia Infections/immunology , Yersinia ruckeri/genetics , Yersinia ruckeri/immunology
19.
J Cell Mol Med ; 23(9): 5868-5875, 2019 09.
Article in English | MEDLINE | ID: mdl-31350813

ABSTRACT

Gram-negative bacteria (GNB) emerge as important pathogens causing pulmonary infection, which can develop into sepsis due to bacterial resistance to antibiotics. GNB pneumonia poses a huge social and economic burden all over the world. During GNB infection in the lung, Toll-like receptor 4 (TLR4) can form a complex with MD2 and CD14 after recognizing lipopolysaccharide of GNB, initiate the MyD88- and TRIF-dependent signalling pathways and stimulate host non-specific immune response. In this review, we summarize recent progress in our understanding of the role of TLR4 in GNB pneumonia. The latest experimental results, especially in TLR4 knockout animals, suggest a promising potential of targeting TLR4 signalling pathway for the treatment of GNB pneumonia. Furthermore, we highlight the benefits of Traditional Chinese Medicine as novel candidates for the therapy of GNB pneumonia due to the modulation of TLR4 signalling pathway. Finally, we discuss the promise and challenge in the development of TLR4-based drugs for GNB pneumonia.


Subject(s)
Gram-Negative Bacterial Infections/drug therapy , Medicine, Chinese Traditional/methods , Pneumonia, Bacterial/drug therapy , Toll-Like Receptor 4/drug effects , Adaptor Proteins, Vesicular Transport/immunology , Gram-Negative Bacteria/metabolism , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Humans , Lipopolysaccharides/metabolism , Myeloid Differentiation Factor 88/immunology , Pneumonia, Bacterial/microbiology , Sepsis/immunology , Sepsis/pathology , Signal Transduction/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
20.
Fish Shellfish Immunol ; 91: 108-121, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31091461

ABSTRACT

As an adaptor in Toll-like receptor (TLR) signaling pathway, Toll/interleukin-1 receptor (TIR) domain containing adaptor inducing interferon-ß (TRIF) mediates downstream signaling cascades and plays important roles in host innate immune responses. In the present study, a TRIF ortholog named Lc-TRIF was identified in large yellow croaker (Larimichthys crocea). Sequence comparison analysis revealed that Lc-TRIF has a conserved TIR domain but without TRAF6 binding motif. The genome structure of Lc-TRIF is conserved, with two exons and one intron. Syntenic comparison showed that the loci of fish TRIF was different from that in mammals or birds, and TRAM was absent in the genomes of fish, amphibians, and birds, but present in mammals and reptiles. Expression analysis revealed that Lc-TRIF was broadly expressed in examined organs/tissues, with the highest expression level in gill and weakest in brain, and could be up-regulated under poly I:C, LPS, PGN, and Pseudomonas plecoglossicida stimulation. Fluorescence microscopy results showed that Lc-TRIF exhibited a global localization throughout the entire cell including the nucleus in HEK 293T cells. Additionally, luciferase assays demonstrated that Lc-TRIF expression could significantly induce NF-κB, type I IFN, IRF3 as well as IRF7 promoter activation. These results collectively indicated that Lc-TRIF was function in host antiviral and antibacterial responses via NF-κB and IRF3/7 related signaling pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , Perciformes/immunology , Adaptor Proteins, Vesicular Transport/chemistry , Amino Acid Sequence , Animals , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...