Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Fish Shellfish Immunol ; 149: 109581, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670412

ABSTRACT

Deubiquitinating enzyme A (DUBA), a member of the ovarian tumor (OTU) subfamily of deubiquitinases (DUBs), is recognized for its negative regulatory role in type I interferon (IFN) expression downstream of Toll-like receptor 3 (TLR3). However, its involvement in the TLR3 signaling pathway in fish remains largely unexplored. In this study, we investigated the regulatory role of DUBA (OmDUBA) in the TLR3 response in rainbow trout (Oncorhynchus mykiss). OmDUBA features a conserved OTU domain, and its expression increased in RTH-149 cells following stimulation with the TLR3 agonist poly(I:C). Gain- and loss-of-function experiments demonstrated that OmDUBA attenuated the activation of TANK-binding kinase 1 (TBK1), resulting in a subsequent reduction in type I IFN expression and IFN-stimulated response element (ISRE) activation in poly(I:C)-stimulated cells. OmDUBA interacted with TRAF3, a crucial mediator in TLR3-mediated type I IFN production. Under poly(I:C) stimulation, there was an augmentation in the K63-linked polyubiquitination of TRAF3, a process significantly inhibited upon OmDUBA overexpression. These findings suggest that OmDUBA may function similarly to its mammalian counterparts in downregulating the poly(I:C)-induced type I IFN response in rainbow trout by removing the K63-linked ubiquitin chain on TRAF3. Our study provides novel insights into the role of fish DUBA in antiviral immunity.


Subject(s)
Fish Proteins , Interferon Type I , Oncorhynchus mykiss , Poly I-C , Signal Transduction , TNF Receptor-Associated Factor 3 , Animals , Oncorhynchus mykiss/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 3/immunology , Interferon Type I/immunology , Interferon Type I/genetics , Interferon Type I/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Signal Transduction/immunology , Poly I-C/pharmacology , Immunity, Innate , Gene Expression Regulation/immunology , Ubiquitination , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/immunology
2.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38597360

ABSTRACT

Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-ß, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.


Subject(s)
DEAD Box Protein 58 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Mice, Knockout , Oligosaccharides , Orthomyxoviridae Infections , Signal Transduction , TNF Receptor-Associated Factor 3 , Animals , Mice , Oligosaccharides/administration & dosage , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Humans , Signal Transduction/drug effects , Signal Transduction/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 3/immunology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/immunology , Pneumonia/immunology , Pneumonia/prevention & control , Pneumonia/metabolism , Pneumonia/virology , Mice, Inbred C57BL , Lung/immunology , Lung/metabolism , Lung/drug effects , Lung/virology , Cytokines/metabolism , Cytokines/immunology , Cytokines/genetics , Female , NF-kappa B/immunology , NF-kappa B/genetics , NF-kappa B/metabolism , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology
3.
J Immunol ; 207(1): 322-332, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34145060

ABSTRACT

The adaptor protein TNFR-associated factor 3 (TRAF3) is required for in vivo T cell effector functions and for normal TCR/CD28 signaling. TRAF3-mediated enhancement of TCR function requires engagement of both CD3 and CD28, but the molecular mechanisms underlying how TRAF3 interacts with and impacts TCR/CD28-mediated complexes to enhance their signaling remains an important knowledge gap. We investigated how TRAF3 is recruited to, and regulates, CD28 as a TCR costimulator. Direct association with known signaling motifs in CD28 was dispensable for TRAF3 recruitment; rather, TRAF3 associated with the CD28-interacting protein linker of activated T cells (LAT) in human and mouse T cells. TRAF3-LAT association required the TRAF3 TRAF-C domain and a newly identified TRAF2/3 binding motif in LAT. TRAF3 inhibited function of the LAT-associated negative regulatory protein Dok1, which is phosphorylated at an inhibitory tyrosine residue by the tyrosine kinase breast tumor kinase (Brk/PTK6). TRAF3 regulated Brk activation in T cells, limiting the association of protein tyrosine phosphatase 1B (PTP1B) with the LAT complex. In TRAF3-deficient cells, LAT complex-associated PTP1B was associated with dephosphorylation of Brk at an activating tyrosine residue, potentially reducing its ability to inhibit Dok1. Consistent with these findings, inhibiting PTP1B activity in TRAF3-deficient T cells rescued basal and TCR/CD28-mediated activation of Src family kinases. These results reveal a new mechanism for promotion of TCR/CD28-mediated signaling through restraint of negative regulation of LAT by TRAF3, enhancing the understanding of regulation of the TCR complex.


Subject(s)
CD28 Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
4.
J Immunol ; 206(11): 2668-2681, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34011520

ABSTRACT

The antiviral innate immune responses are crucial steps during host defense and must be strictly regulated, but the molecular mechanisms of control remain unclear. In this study, we report increased expression of human ATPase Na+/K+ transporting subunit ß 1(ATP1B1) after DNA and RNA virus infections. We found that the expression of ATP1B1 can inhibit viral replication and increase the levels of IFNs, IFN-stimulated genes, and inflammatory cytokines. Knockdown of ATP1B1 by specific short hairpin RNA had the opposite effects. Upon viral infection, ATP1B1 was induced, interacted with TRAF3 and TRAF6, and potentiated the ubiquitination of these proteins, leading to increased phosphorylation of downstream molecules, including TGF-ß-activated kinase 1 (TAK1) and TANK-binding kinase 1 (TBK1). These results reveal a previously unrecognized role of ATP1B1 in antiviral innate immunity and suggest a novel mechanism for the induction of IFNs and proinflammatory cytokines during viral infection.


Subject(s)
Immunity, Innate/immunology , Intracellular Signaling Peptides and Proteins/immunology , Sodium-Potassium-Exchanging ATPase/immunology , TNF Receptor-Associated Factor 3/immunology , Up-Regulation/immunology , Animals , Cells, Cultured , Chlorocebus aethiops , DNA Virus Infections/immunology , DNA Viruses/immunology , Humans , RNA Virus Infections/immunology , RNA Viruses/immunology , Sodium-Potassium-Exchanging ATPase/genetics , Ubiquitination/immunology , Virus Replication
5.
Front Immunol ; 12: 573078, 2021.
Article in English | MEDLINE | ID: mdl-33692778

ABSTRACT

Swine acute diarrhea syndrome coronavirus (SADS-CoV), first discovered in 2017, is a porcine enteric coronavirus that can cause acute diarrhea syndrome (SADS) in piglets. Here, we studied the role of SADS-CoV nucleocapsid (N) protein in innate immunity. Our results showed that SADS-CoV N protein could inhibit type I interferon (IFN) production mediated by Sendai virus (Sev) and could block the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Simultaneously, the IFN-ß promoter activity mediated by TANK binding kinase 1 (TBK1) or its upstream molecules in the RLRs signal pathway was inhibited by SADS-CoV N protein. Further investigations revealed that SADS-CoV N protein could counteract interaction between TNF receptor-associated factor 3 (TRAF3) and TBK1, which led to reduced TBK1 activation and IFN-ß production. Our study is the first report of the interaction between SADS-CoV N protein and the host antiviral innate immune responses, and the mechanism utilized by SADS-CoV N protein provides a new insight of coronaviruses evading host antiviral innate immunity.


Subject(s)
Alphacoronavirus/metabolism , Coronavirus Nucleocapsid Proteins/immunology , Interferon-beta/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , TNF Receptor-Associated Factor 3/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Alphacoronavirus/immunology , Animals , Cell Line , Coronavirus/immunology , Coronavirus/metabolism , Coronavirus Nucleocapsid Proteins/metabolism , HEK293 Cells , Host-Pathogen Interactions , Humans , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interferon-beta/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Swine , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649212

ABSTRACT

The recently elucidated proresolving conjugates in tissue regeneration (CTR) maresin-CTR (MCTR), protectin-CTR (PCTR), and resolvin-CTR (RCTR), termed cysteinyl-specialized proresolving mediators (cys-SPMs) each promotes regeneration, controls infection, and accelerates resolution of inflammation. Here, we sought evidence for cys-SPM activation of primordial pathways in planaria (Dugesia japonica) regeneration that might link resolution of inflammation and regeneration. On surgical resection, planaria regeneration was enhanced with MCTR3, PCTR3, or RCTR3 (10 nM), each used for RNA sequencing. The three cys-SPMs shared up-regulation of 175 known transcripts with fold-change > 1.25 and combined false discovery rate (FDR) < 0.002, and 199 canonical pathways (FDR < 0.25), including NF-κB pathways and an ortholog of human TRAF3 (TNFR-associated factor 3). Three separate pathway analyses converged on TRAF3 up-regulation by cys-SPMs. With human macrophages, three cys-SPMs each dose-dependently increased TRAF3 expression in a cAMP-PKA-dependent manner. TRAF3 overexpression in macrophages enhanced Interleukin-10 (IL-10) and phagocytosis of Escherichia coli IL-10 also increased phagocytosis in a dose-dependent manner. Silencing of mouse TRAF3 in vivo significantly reduced IL-10 and macrophage phagocytosis. TRAF3 silencing in vivo also relieved cys-SPMs' actions in limiting polymorphonuclear neutrophil in E. coli exudates. These results identify cys-SPM-regulated pathways in planaria regeneration, uncovering a role for TRAF3/IL-10 in regulating mammalian phagocyte functions in resolution. Cys-SPM activation of TRAF3 signaling is a molecular component of both regeneration and resolution of infectious inflammation.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/immunology , Planarians/immunology , Regeneration/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Escherichia coli Infections/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Neutrophils/immunology , Phagocytosis , Planarians/genetics , Regeneration/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 3/genetics
7.
Dev Comp Immunol ; 119: 104015, 2021 06.
Article in English | MEDLINE | ID: mdl-33460679

ABSTRACT

Tumour necrosis factor receptor associated factor 3 (TRAF3) is a crucial transducing protein for linking upstream receptor signals and downstream antiviral signalling pathways. Previous studies mostly clarified the functions of TRAF3 in mammals, birds and fish, but little is known about the characterization and function of TRAF3 in amphibians. In this study, the molecular and functional identification of two TRAF3 genes, AdTRAF3A and AdTRAF3B, were investigated in the Chinese giant salamander Andrias davidianus. The complete open reading frames (ORFs) of AdTRAF3A and AdTRAF3B were 1698 bp and 1743 bp in length, encoding 565 and 580 amino acids, respectively. Both AdTRAF3A and AdTRAF3B deduced proteins contained a RING finger, two TRAF-type zinc fingers, a coiled-coil and a MATH domain. Phylogenetic analysis showed that the AdTRAF3 protein clustered together with other known TRAF3 proteins. Gene expression analysis showed that AdTRAF3s were broadly distributed in all examined tissues with similar distribution patterns. AdTRAF3s in the blood or spleen positively responded to Giant salamander iridovirus (GSIV) and poly (I:C) induction but exhibited distinct response patterns. Silencing AdTRAF3A/B remarkably suppressed the expression of IFN signalling pathway-related genes when leukocytes were treated with DNA virus and the viral RNA analogue. Moreover, overexpression of AdTRAF3A may induce the activation of the IFN-ß promoter, and the zinc finger, coiled coil and MATH domains of AdTRAF3A were essential for IFN-ß promoter activation. However, the overexpression of AdTRAF3B significantly suppressed IFN-ß promoter activity, and its inhibitory effect was enhanced when the RING finger or MATH domain was deleted. Furthermore, AdTRAF3A rather than AdTRAF3B significantly induced NF-κB activation, implying that AdTRAF3A may function as an enhancer in both the IFN and NF-κB signalling pathways. Taken together, our results suggest that the two TRAF3 genes play different crucial regulatory roles in innate antiviral immunity in Chinese giant salamanders.


Subject(s)
Immunity, Innate/immunology , Iridovirus/immunology , TNF Receptor-Associated Factor 3/immunology , Urodela/immunology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Computational Biology/methods , Gene Expression Profiling/methods , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Iridovirus/physiology , NF-kappa B/immunology , NF-kappa B/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction/genetics , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/classification , TNF Receptor-Associated Factor 3/genetics , Urodela/genetics , Urodela/virology
8.
J Immunol ; 205(3): 830-841, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591397

ABSTRACT

The BCR recognizes foreign Ags to initiate humoral immunity that needs isotype-switched Abs generated via class switch recombination (CSR); however, stimulating the BCR in the absence of costimulation (e.g., CD40) does not induce CSR; thus, it remains elusive whether and how the BCR induces CSR mechanistically. Autoreactive B cells can maintain anergy via unresponsiveness of their BCRs to self-antigens. However, it remains unknown what molecule(s) restrict BCR signaling strength for licensing BCR-induced CSR and whether deficiency of such molecule(s) disrupts autoreactive B cell anergy and causes B cell-mediated diseases by modulating BCR signaling. In this study, we employ mouse models to show that the BCR's capacity to induce CSR is restrained by B cell-intrinsic checkpoints TRAF3 and TRAF2, whose deletion in B cells enables the BCR to induce CSR in the absence of costimulation. TRAF3 deficiency permits BCR-induced CSR by elevating BCR-proximal signaling intensity. Furthermore, NF-κB2 is required for BCR-induced CSR in TRAF3-deficient B cells but not for CD40-induced or LPS-induced CSR, suggesting that TRAF3 restricts NF-κB2 activation to specifically limit the BCR's ability to induce CSR. TRAF3 deficiency also disrupts autoreactive B cell anergy by elevating calcium influx in response to BCR stimulation, leading to lymphoid organ disorders and autoimmune manifestations. We showed that TRAF3 deficiency-associated autoimmune phenotypes can be rectified by limiting BCR repertoires or attenuating BCR signaling strength. Thus, our studies highlight the importance of TRAF3-mediated restraint on BCR signaling strength for controlling CSR, B cell homeostasis, and B cell-mediated disorders.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy , Immunoglobulin Class Switching/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , B-Lymphocytes/cytology , Mice , Mice, Transgenic , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/immunology , Signal Transduction/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 3/genetics
9.
Fish Shellfish Immunol ; 104: 592-604, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32589928

ABSTRACT

Tumor necrosis factor receptor-associated factor 3 (TRAF3) is a multifunctional adaptor protein primarily involved in both bacterial defense and antiviral immunity in living organisms. However, the knowledge on TRAF3 in blunt snout bream (Megalobrama amblycephala), a freshwater fish with economic values, remained unclear. In the present study, we identified and characterized successfully Traf3 gene from M. amblycephala (maTraf3). The maTraf3 cDNA contained a 1722 bp open reading frame that encoded a protein of 573 amino acid residues. The deduced amino acid sequence comprised of a RING finger domain, two zinc finger motifs, a coiled-coil region and a MATH domain. Analysis of the transcriptional patterns of maTraf3 revealed that it was ubiquitously distributed in various tissues tested from M. amblycephala, with the abundance of expression in spleen and muscle. Following a challenge with Aeromonas hydrophila and lipopolysaccharide stimulation, the expression of maTraf3 was strongly enhanced at different time points in vitro and in vivo. MaTRAF3 was identified as a cytosolic protein and suggested to form aggregates or be associated with vesicles scattering in the cytoplasm. NF-κB transcription was activated by maTraf3 in reporter assay. The overexpression of maTraf3 produced high levels of pro-inflammatory cytokines such as IL-1ß, IL-6, IL-8 and TNF-α, implying its immune-regulatory role in M. amblycephala. Taken together, our results obtained in this study demonstrated the crucial role of maTraf3 in mediating host innate immune response to pathogen invasion via NF-κB signaling pathway, which might indicate a novel therapeutic approach to combat bacterial infection in fish.


Subject(s)
Cyprinidae/genetics , Cyprinidae/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Lipopolysaccharides/pharmacology , Phylogeny , Sequence Alignment/veterinary , TNF Receptor-Associated Factor 3/chemistry
10.
Protein Cell ; 11(12): 894-914, 2020 12.
Article in English | MEDLINE | ID: mdl-32562145

ABSTRACT

Tripartite motif (TRIM) family proteins are important effectors of innate immunity against viral infections. Here we identified TRIM35 as a regulator of TRAF3 activation. Deficiency in or inhibition of TRIM35 suppressed the production of type I interferon (IFN) in response to viral infection. Trim35-deficient mice were more susceptible to influenza A virus (IAV) infection than were wild-type mice. TRIM35 promoted the RIG-I-mediated signaling by catalyzing Lys63-linked polyubiquitination of TRAF3 and the subsequent formation of a signaling complex with VISA and TBK1. IAV PB2 polymerase countered the innate antiviral immune response by impeding the Lys63-linked polyubiquitination and activation of TRAF3. TRIM35 mediated Lys48-linked polyubiquitination and proteasomal degradation of IAV PB2, thereby antagonizing its suppression of TRAF3 activation. Our in vitro and in vivo findings thus reveal novel roles of TRIM35, through catalyzing Lys63- or Lys48-linked polyubiquitination, in RIG-I antiviral immunity and mechanism of defense against IAV infection.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Proteolysis , TNF Receptor-Associated Factor 3/immunology , Ubiquitination/immunology , Viral Proteins/immunology , A549 Cells , Animals , Apoptosis Regulatory Proteins/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Dogs , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype/genetics , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , RAW 264.7 Cells , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells , TNF Receptor-Associated Factor 3/genetics , Viral Proteins/genetics
11.
J Immunol ; 204(5): 1085-1090, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31969387

ABSTRACT

Lymphotoxin ß receptor (LTßR) signaling is crucial for lymphoid tissue organogenesis and immune homeostasis. To identify novel regulatory mechanisms for signaling, we implemented a two-step screen that uses coexpression analysis of human fibroblasts undergoing LTßR stimulation and affinity-purification mass spectrometry for the LTßR signaling protein TNFR-associated factor 3 (TRAF3). We identify Ewing sarcoma (EWS) protein as a novel LTßR signaling component that associates with TRAF3 but not with TNFR-associated factor 2 (TRAF2). The EWS:TRAF3 complex forms under unligated conditions that are disrupted following activation of the LTßR. We conclude that EWS limits expression of proinflammatory molecules, GM-CSF, and ERK-2, promoting immune homeostasis.


Subject(s)
Lymphotoxin beta Receptor/immunology , MAP Kinase Signaling System/immunology , Multiprotein Complexes/immunology , RNA-Binding Protein EWS/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HEK293 Cells , Humans , Lymphotoxin beta Receptor/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Multiprotein Complexes/genetics , RNA-Binding Protein EWS/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology
12.
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
13.
J Fish Biol ; 96(1): 185-193, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31721203

ABSTRACT

In this study, we cloned the complementary (c)DNA sequences of tumour necrosis factor receptor (TNFR)-associated factor 3 (traf3) in Nile tilapia, Oreochromis niloticus. The expression patterns of the traf3 gene were investigated and preliminary functional analyses were performed. In healthy fish, traf3 transcript was broadly expressed in all examined tissues, with the highest expression level in the blood and the lowest in the liver. The traf3 gene reached its highest expression at 8 days post-fertilisation (dpf) during embryonic development. Moreover, we found that expression of traf3 was clearly altered following stimulation with Streptococcus agalactiae in vivo and that traf3 could be induced by lipopolysaccharides (LPS), Poly I: C and S. agalactiae WC1535 in Nile tilapia macrophages. Overexpression in 293T cells showed that Traf3 protein was mainly distributed in the cytoplasm and could significantly increase nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. Taken together, these results implied that traf3 could play important roles in the immune response to pathogen invasion.


Subject(s)
Cichlids/immunology , NF-kappa B/metabolism , TNF Receptor-Associated Factor 3 , Animals , Cell Culture Techniques , Cichlids/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Gene Expression , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , NF-kappa B/immunology , Streptococcus agalactiae/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism
14.
Protein & Cell ; (12): 894-914, 2020.
Article in English | WPRIM (Western Pacific) | ID: wpr-880885

ABSTRACT

Tripartite motif (TRIM) family proteins are important effectors of innate immunity against viral infections. Here we identified TRIM35 as a regulator of TRAF3 activation. Deficiency in or inhibition of TRIM35 suppressed the production of type I interferon (IFN) in response to viral infection. Trim35-deficient mice were more susceptible to influenza A virus (IAV) infection than were wild-type mice. TRIM35 promoted the RIG-I-mediated signaling by catalyzing Lys63-linked polyubiquitination of TRAF3 and the subsequent formation of a signaling complex with VISA and TBK1. IAV PB2 polymerase countered the innate antiviral immune response by impeding the Lys63-linked polyubiquitination and activation of TRAF3. TRIM35 mediated Lys48-linked polyubiquitination and proteasomal degradation of IAV PB2, thereby antagonizing its suppression of TRAF3 activation. Our in vitro and in vivo findings thus reveal novel roles of TRIM35, through catalyzing Lys63- or Lys48-linked polyubiquitination, in RIG-I antiviral immunity and mechanism of defense against IAV infection.


Subject(s)
Animals , Dogs , Humans , Mice , A549 Cells , Apoptosis Regulatory Proteins/immunology , DEAD Box Protein 58/immunology , HEK293 Cells , Influenza A Virus, H1N1 Subtype/immunology , Madin Darby Canine Kidney Cells , Mice, Knockout , Orthomyxoviridae Infections/pathology , Proteolysis , Signal Transduction/immunology , THP-1 Cells , TNF Receptor-Associated Factor 3/immunology , Ubiquitination/immunology , Viral Proteins/immunology
15.
J Med Virol ; 91(3): 482-492, 2019 03.
Article in English | MEDLINE | ID: mdl-30267576

ABSTRACT

The mitochondrial antiviral signal protein mitochondrial antiviral signaling protein, also known as virus-induced signaling adaptor (VISA), plays a key role in regulating host innate immune signaling pathways. This study identifies FK506 binding protein 8 (FKBP8) as a candidate interacting protein of VISA through the yeast two-hybrid technique. The interaction of FKBP8 with VISA, retinoic acid inducible protein 1 (RIG-I), and IFN regulatory factor 3 (IRF3) was confirmed during viral infection in mammalian cells by coimmunoprecipitation. Overexpression of FKBP8 using a eukaryotic expression plasmid significantly attenuated Sendai virus-induced activation of the promoter interferons ß (IFN-ß), and transcription factors nuclear factor κ-light chain enhancer of activated B cells (NF-κB) and IFN-stimulated response element (ISRE). Overexpression of FKBP8 also decreased dimer-IRF3 activity, but enhanced virus replication. Conversely, knockdown of FKBP8 expression by RNA interference showed opposite effects. Further studies indicated that FKBP8 acts as a negative interacting partner to regulate RLR-VISA signaling by acting on VISA and TANK binding kinase 1 (TBK1). Additionally, FKBP8 played a negative role on virus-induced signaling by inhibiting the formation of TBK1-IRF3 and VISA-TRAF3 complexes. Notably, FKBP8 also promoted the degradation of TBK1, RIG-I, and TRAF3 resulting from FKBP8 reinforced Sendai virus-induced endogenous polyubiquitination of RIG-I, TBK1, and TNF receptor-associated factor 3 (TRAF3). Therefore, a novel function of FKBP8 in innate immunity antiviral signaling regulation was revealed in this study.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Sendai virus , Signal Transduction , Tacrolimus Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Protein Binding , Protein Serine-Threonine Kinases/immunology , Receptors, Immunologic , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Two-Hybrid System Techniques , Ubiquitination
16.
FEBS J ; 286(3): 523-535, 2019 02.
Article in English | MEDLINE | ID: mdl-30536547

ABSTRACT

Fas (CD95) signalling is best known for its role in apoptosis, however, recent reports have shown it to be involved in other cellular responses as well, including inflammation. Fas and its adaptor protein FADD are known to negatively regulate LPS-induced proinflammatory responses, but their role in LPS-induced type I interferon production is unknown. Here, we demonstrate that Fas engagement on macrophages, using an agonistic Fas antibody CH11, augments LPS-induced NF-κB responses, causing increased production of TNFα, IL-8, IL-6 and IL-12. Conversely, costimulation with both LPS and CH11 causes a significant reduction in the level of interferon-beta (IFNß) production. This differential effect involves the Fas adaptor FADD because while LPS-induced IL-6 production increased in FADD-/- murine embryonic fibroblasts, LPS-induced IFNß production was significantly reduced in these cells. Overexpression of a dominant negative form of FADD (FADD-DD) inhibits LPS-induced IFNß luciferase but not LPS-induced NF-κB luciferase. In contrast, overexpression of full-length FADD inhibited LPS-induced NF-κB luciferase activation but was seen to augment LPS-induced IFNß luciferase. Moreover, FADD-DD inhibits TRIF-, TRAM-, IKKε-, TBK-1- and TRAF3-induced IFNß luciferase production, with coimmunoprecipitation experiments demonstrating an interaction between FADD and TRIF. These data identify FADD as a novel component of the noncanonical Toll-like receptor 4/IFNß signalling pathway and demonstrate that both Fas and its adaptor FADD can differentially regulate the production of LPS-induced proinflammatory cytokines and type I interferons.


Subject(s)
Fas-Associated Death Domain Protein/genetics , Interferon-beta/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Toll-Like Receptor 4/genetics , fas Receptor/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Animals , Antibodies/pharmacology , Fas-Associated Death Domain Protein/immunology , Gene Expression Regulation , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Interferon-beta/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Jurkat Cells , Macrophages/cytology , Macrophages/immunology , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RAW 264.7 Cells , Signal Transduction , THP-1 Cells , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , fas Receptor/antagonists & inhibitors , fas Receptor/immunology
17.
Mol Immunol ; 106: 53-62, 2019 02.
Article in English | MEDLINE | ID: mdl-30579117

ABSTRACT

The inhibition of tumor necrosis factor receptor-associated factor 3 (TRAF3) degradation induces endotoxin tolerance (ET) in macrophages. However, the mechanisms leading to TRAF3 inhibition by ET are largely unknown. Here, we found that ubiquitin-specific peptidase 25 (USP25), a deubiquitinating enzyme (DUB), interacted with TRAF3 and stabilized ET in Kupffer cells (KCs). Lentiviral knockdown of USP25 activated K48-linked ubiquitination of TRAF3 and the cytoplasmic translocation of the Myd88-associated multiprotein complex in tolerized KCs. This outcome led to a subsequent activation of Myd88-dependent c-Jun N-terminal kinase (JNK) and p38-mediated downregulation of inflammatory cytokines. The overexpression of TRAF3 attenuated the proinflammatory effects of USP25 knockdown in tolerized KCs. Thus, our findings reveal a novel mechanism of endotoxin-mediated TRAF3 degradation in KCs.


Subject(s)
Endotoxins/immunology , Immune Tolerance , Kupffer Cells/immunology , Proteolysis , TNF Receptor-Associated Factor 3/immunology , Ubiquitin Thiolesterase/immunology , Ubiquitination/immunology , Animals , Gene Knockdown Techniques , Lentivirus , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Male , Mice , TNF Receptor-Associated Factor 3/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitination/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
18.
Nat Commun ; 9(1): 4964, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470758

ABSTRACT

The inflammasome plays a critical role in inflammation and immune responses against pathogens. However, whether or how inflammasome activation regulates type I interferon (IFN-I) signaling in the context of malaria infection remain unknown. Here we show mice deficient in inflammasome sensors AIM2, NLRP3 or adaptor Caspase-1 produce high levels of IFN-I cytokines and are resistant to lethal Plasmodium yoelii YM infection. Inactivation of inflammasome signaling reduces interleukin (IL)-1ß production, but increases IFN-I production. Mechanistically, we show inflammsome activation enhances IL-1ß-mediated MyD88-TRAF3-IRF3 signaling and SOCS1 upregulation. However, SOCS1 inhibits MyD88-IRF7-mediated-IFN-I signaling and cytokine production in plasmacytoid dendritic cells. By contrast, ablation of inflammsome components reduces SOCS1 induction, and relieves its inhibition on MyD88-IRF7-dependent-IFN-I signaling, leading to high levels of IFN-α/ß production and host survival. Our study identifies a previously unrecognized role of inflammasome activation in the negative regulation of IFN-I signaling pathways and provides potential targets for developing effective malaria vaccines.


Subject(s)
Inflammasomes/immunology , Interferon Regulatory Factor-7/immunology , Malaria/immunology , Myeloid Differentiation Factor 88/immunology , Plasmodium yoelii/physiology , Animals , Down-Regulation , Female , Humans , Inflammasomes/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Malaria/genetics , Malaria/parasitology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology
19.
Front Immunol ; 9: 2161, 2018.
Article in English | MEDLINE | ID: mdl-30319624

ABSTRACT

The adaptor protein TNF receptor-associated factor 3 (TRAF3) serves as a powerful negative regulator in multiple aspects of B cell biology. Early in vitro studies in transformed cell lines suggested the potential of TRAF3 to inhibit signaling by its first identified binding receptor, CD40. However, because the canonical TRAF3 binding site on many receptors also mediates binding of other TRAFs, and whole-mouse TRAF3 deficiency is neonatally lethal, an accurate understanding of TRAF3's specific functions was delayed until conditional TRAF3-deficient mice were produced. Studies of B cell-specific TRAF3-deficient mice, complemented by investigations in normal and malignant mouse and human B cells, reveal that TRAF3 has powerful regulatory roles that are unique to this TRAF, as well as functions context-specific to the B cell. This review summarizes the current state of knowledge of these roles and functions. These include inhibition of signaling by plasma membrane receptors, negative regulation of intracellular receptors, and restraint of cytoplasmic NF- κB pathways. TRAF3 is also now known to function as a resident nuclear protein, and to impact B cell metabolism. Through these and additional mechanisms TRAF3 exerts powerful restraint upon B cell survival and activation. It is thus perhaps not surprising that TRAF3 has been revealed as an important tumor suppressor in B cells. The many and varied functions of TRAF3 in B cells, and new directions to pursue in future studies, are summarized and discussed here.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/immunology , Tumor Suppressor Proteins/immunology , Animals , Cell Survival/immunology , Humans , Mice , Mice, Knockout , NF-kappa B/immunology
20.
Fish Shellfish Immunol ; 82: 27-31, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30075247

ABSTRACT

The large yellow croaker (Larimichthys crocea) has a well-developed innate immune system. To gain a better understanding of the defense mechanisms involved in this system, we studied tumor necrosis factor receptor-associated factors (TRAFs), which play important roles in the Toll-like receptor (TLR) pathway. We characterized the full-length open reading frames and protein structures of TRAF3 and TRAF6 to determine their identities, and conducted phylogenetic analysis to determine their evolutionary relationships. To assess the roles of TRAFs in innate immune responses in the large yellow croaker, we performed quantitative reverse-transcription PCR (qRT-PCR) to characterize expression profiles in a range of tissues at different stages after challenge with polyinosinic polycytidylic acid (poly I:C) and Vibrio anguillarum. Following poly I:C challenge, the expression levels of TRAF3 and TRAF6 were highest in the kidneys and lowest in the spleen, whereas after infection with V. anguillarum, TRAF6 expression was the highest in the kidneys and lowest in the liver.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 6/genetics , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Perciformes/immunology , Phylogeny , Sequence Alignment/veterinary , TNF Receptor-Associated Factor 3/chemistry , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...