Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Fish Shellfish Immunol ; 121: 332-341, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35032679

ABSTRACT

The ubiquitin-specific proteases (USPs) have attracted particular attention due to their multiple functions in different biological processes. USP12, a member of the USP family, has been demonstrated to exert critical roles in diverse cellular processes, including cell death, cancer and antiviral immunity. Here, we cloned a USP12 homolog from orange spotted grouper (Epinephelus coioides, E. coioides), and its roles in fish RNA virus replication were investigated. EcUSP12 contained a 1119-bp open reading frame (ORF) encoding a 372-amino acid polypeptide, which shared 100.00% and 91.32% identity with USP12 homolog of Etheostoma cragini and Homo sapiens, respectively. Sequence analysis indicated that EcUSP12 contained a conserved peptidase-C19G domain (aa 40-369). qPCR analysis showed that EcUSP12 transcript was most abundant in head kidney and spleen of grouper E. coioides. The expression of EcUSP12 was significantly upregulated in grouper spleen (GS) cells in response to red-spotted grouper nervous necrosis virus (RGNNV) infection. Subcellular localization analysis showed that EcUSP12 was evenly distributed throughout the cytoplasm, and mainly co-localized with endoplasmic reticulum (ER). Interestingly, during RGNNV infection, the endogenous distribution of EcUSP12 was obviously altered, and mostly overlapped with viral coat protein (CP). Co-Immunoprecipitation (Co-IP) assay indicated that EcUSP12 interacted with viral CP. In addition, overexpression of EcUSP12 significantly inhibited the replication of RGNNV in vitro, as evidenced by the decrease in viral gene transcription and protein synthesis during infection. Consistently, knockdown of EcUSP12 by small interfering RNA (siRNA) promoted the replication of RGNNV. Furthermore, EcUSP12 overexpression also increased the transcription level of inflammatory factors and interferon-related genes, including tumor necrosis factor α (TNF-α), interleukin (IL)-1ß, IL-6, IL-8, interferon regulatory factor 3 (IRF3), and IRF7. Taken together, our results demonstrated that EcUSP12, as a positive regulator of IFN signaling, interacted with viral CP to inhibit virus infection.


Subject(s)
Bass , DNA Virus Infections , Fish Diseases , Fish Proteins/immunology , Immunity, Innate , Ubiquitin-Specific Proteases/immunology , Amino Acid Sequence , Animals , Bass/immunology , Bass/virology , DNA Virus Infections/immunology , DNA Virus Infections/veterinary , Fish Diseases/virology , Nodaviridae , Phylogeny , Sequence Alignment
2.
J Crohns Colitis ; 16(1): 122-132, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-34232309

ABSTRACT

BACKGROUND AND AIMS: The inflammatory bowel disease [IBD]-associated immune response is marked by excessive production of a variety of inflammatory cytokines, which are supposed to sustain and amplify the pathological process. OTUD5 is a deubiquitinating enzyme, which regulates cytokine production by both innate and adaptive immune cells. Here, we investigated the expression and role of OTUD5 in IBD. METHODS: OTUD5 expression was evaluated in mucosal samples of patients with Crohn's disease [CD], patients with ulcerative colitis [UC], and controls, as well as in mice with trinitrobenzene-sulphonic acid [TNBS]-induced colitis by real-time polymerase chain reaction, western blotting, immunohistochemistry, and immunofluorescence. Moreover, OTUD5 was assessed in lamina propria mononuclear cells [LPMC] stimulated with inflammatory cytokines. TNF-α, IL-6, and IL-10 were evaluated in LPMCs of IBD patients and in colitic mice transfected with a specific OTUD5 antisense oligonucleotide [AS]. RESULTS: OTUD5 protein, but not RNA, expression was increased in inflamed ileal and colonic mucosal samples of patients with CD and patients with UC as compared with controls. In IBD, OTUD5-expressing cells were abundant in both epithelial and lamina propria compartments, and non-CD3+, HLA-DR+ LPMC were one of the major sources of the protein. OTUD5 expression was enhanced by IFN-γ through a p38/MAPK-dependent mechanism, and the AS-induced knockdown of OTUD5 in LPMCs of IBD patients and colitic mice reduced TNF-α. CONCLUSIONS: Our data show that OTUD5 is overexpressed in both CD and UC and suggest the involvement of such a protein in the amplification of the aberrant cytokine response in IBD.


Subject(s)
Cytokines/immunology , Endopeptidases/immunology , Inflammatory Bowel Diseases/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Biopsy , Female , Humans , Male , Mice , Mice, Inbred BALB C
3.
Life Sci ; 281: 119720, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34144056

ABSTRACT

AIMS: Asthma is characterized by chronic inflammation and airway hyperresponsiveness (AHR). It is controllable, but not curable. Ubiquitin-specific peptidase 4 (USP4) has been verified as a regulator of regulatory T (Treg) cells and Th17 cells in vitro. In this study, we aim to investigate whether USP4 could serve as a therapeutic target for asthma. MAIN METHODS: Age-matched USP4 wild-type and knockout mice received an intraperitoneal injection of 100 µg ovalbumin (OVA) mixed in 2 mg aluminum hydroxide in 1 × PBS on days 0, 7 and 14. On days 21 to 27, the mice were challenged with aerosolized 1% OVA in 1 × PBS for 30 min. Tissue histology, ELISA and flow cytometry were applied 24 h after the last OVA challenge. KEY FINDINGS: USP4 deficiency protected mice from OVA-induced AHR and decreased the production of several inflammatory cytokines in T cells in vivo. Compared to the lung cells isolated from WT mice, Usp4-/- lung cells decreased secretion of IL-4, IL-13 and IL-17A upon stimulation in vitro. Meanwhile, the percentage of CD4+Foxp3+ Treg cells was elevated, with more CCR6+Foxp3+ Treg cells accumulating in the lungs of OVA-challenged USP4 deficient mice than in their wild-type counterparts. Treatment with the USP4 inhibitor, Vialinin A, reduced inflammatory cell infiltration in the lungs of OVA-challenged mice in vivo. SIGNIFICANCE: We found USP4 deficiency contributes to attenuated airway inflammation and AHR in allergen-induced murine asthma, and Vialinin A treatment alleviates asthma pathogenesis and may serve as a promising therapeutic target for asthma.


Subject(s)
Asthma/immunology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Bronchoalveolar Lavage Fluid , Cell Differentiation , Disease Models, Animal , Female , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , Ubiquitin-Specific Proteases/genetics
5.
Cell Rep ; 33(3): 108297, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086059

ABSTRACT

The immune system is not only required for preventing threats exerted by pathogens but also essential for developing immune tolerance to avoid tissue damage. This study identifies a distinct mechanism by which MYSM1 suppresses innate immunity and autoimmunity. The expression of MYSM1 is induced upon DNA virus infection and by intracellular DNA stimulation. MYSM1 subsequently interacts with STING and cleaves STING K63-linked ubiquitination to suppress cGAS-STING signaling. Notably, Mysm1-deficient mice exhibit a hyper-inflammatory response, acute tissue damage, and high mortality upon virus infection. Moreover, in the PBMCs of patients with systemic lupus erythematosus (SLE), MYSM1 production decreases, while type I interferons and pro-inflammatory cytokine expressions increase. Importantly, MYSM1 treatment represses the production of IFNs and pro-inflammatory cytokines in the PBMCs of SLE patients. Thus, MYSM1 is a critical repressor of innate immunity and autoimmunity and is thus a potential therapeutic agent for infectious, inflammatory, and autoimmune diseases.


Subject(s)
Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/metabolism , Adult , Animals , Autoimmune Diseases , Autoimmunity/immunology , China , Female , Humans , Immunity, Innate/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon Type I/physiology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nucleotidyltransferases/physiology , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology
6.
Fish Shellfish Immunol ; 103: 239-247, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32437860

ABSTRACT

Deubiquitinases are widely involved in the regulation of the virus-triggered type I interferon (IFN) signaling. Here, we found sea perch (Lateolabrax japonicus) ubiquitin-specific protease 5 (LjUSP5) was a negative regulatory factor of the red-spotted grouper nervous necrosis virus (RGNNV)-triggered IFN response. LjUSP5 encoded a polypeptide of 830 amino acids, containing a zinc finger UBP domain (residues 197-270 aa), two ubiquitin-associated domains (residues 593-607 aa; 628-665 aa), and one UBP domain (residues 782-807 aa), and shared the closest genetic relationship with the USP5 of Larimichthys crocea. Quantitative RT-PCR analysis showed that LjUSP5 was ubiquitously expressed and up-regulated significantly in all inspected tissues post RGNNV infection, and its transcripts significantly increased in brain, liver and kidney tissues post RGNNV infection. LjUSP5 was up-regulated in cultured LJB cells after poly I:C and RGNNV treatments. In addition, overexpression of LjUSP5 significantly inhibited the activation of zebrafish IFN 1 promoter and promoted RGNNV replication in vitro. Furthermore, LjUSP5 inhibited the activation of zebrafish IFN 1 promoter induced by key genes of retinoic acid-inducible gene I-like receptors signaling pathway. Our findings provides useful information for further elucidating the mechanism underlying NNV infection.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Perciformes/genetics , Perciformes/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Nodaviridae/physiology , Phylogeny , RNA Virus Infections/immunology , RNA Virus Infections/veterinary , Ubiquitin-Specific Proteases/chemistry
7.
Cell Res ; 30(10): 914-927, 2020 10.
Article in English | MEDLINE | ID: mdl-32457395

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is an essential sensor of cytosolic DNA and critically mediates innate immune responses and autoimmunity. Modulating the activity and stability of cGAS provides potential strategies for treating viral or autoimmune diseases. Here, we report that ubiquitin-specific protease 29 (USP29) deubiquitinates and stabilizes cGAS and promotes cellular antiviral responses and autoimmunity. Knockdown or knockout of USP29 severely impairs Herpes simplex virus 1 (HSV-1)- or cytosolic DNA-induced expression of type I interferons (IFNs) and proinflammatory cytokines. Consistently, Usp29m/m mice produce decreased type I IFNs and proinflammatory cytokines after HSV-1 infection and are hypersensitive to HSV-1 infection compared to the wild-type littermates. In addition, genetic ablation of USP29 in Trex1-/- mice eliminated the detectable pathological and molecular autoimmune phenotypes. Mechanistically, USP29 constitutively interacts with cGAS, deconjugates K48-linked polyubiquitin chains from cGAS and stabilizes cGAS in uninfected cells or after HSV-1 infection. Reconstitution of cGAS into Usp29-/- cells fully rescues type I IFN induction and cellular antiviral responses after HSV-1 infection. Our findings thus reveal a critical role of USP29 in the innate antiviral responses against DNA viruses and autoimmune diseases and provide insight into the regulation of cGAS.


Subject(s)
Antiviral Agents/immunology , Autoimmune Diseases/immunology , Herpes Simplex/immunology , Nucleotidyltransferases/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Bone Marrow Cells , HEK293 Cells , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
PLoS Pathog ; 16(2): e1008293, 2020 02.
Article in English | MEDLINE | ID: mdl-32027733

ABSTRACT

RIG-I plays important roles in pathogen sensing and activation of antiviral innate immune responses in response to RNA viruses. RIG-I-mediated signaling must be precisely controlled to maintain innate immune signaling homeostasis. Previous studies demonstrated that lysine 63 (K63)-linked polyubiquitination of RIG-I is vital for its activation, but the mechanisms through which RIG-I is deubiquitinated to control innate immune responses are not well understood. Here we identified USP27X as a negative regulator of antiviral signaling in response to RNA viruses through siRNA library screening. Further functional studies indicated that USP27X negatively modulated RIG-I-mediated antiviral signaling in a deubiquitinase-dependent manner. Mechanistically, we found that USP27X removed K63-linked polyubiquitin chains from RIG-I to negatively modulate type I interferon signaling. Collectively, these studies uncover a novel negative regulatory role of USP27X in targeting RIG-I to balance innate immune responses.


Subject(s)
DEAD Box Protein 58 , Immunity, Innate/genetics , Signal Transduction , Ubiquitin-Specific Proteases , Viruses/immunology , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HeLa Cells , Hep G2 Cells , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Mice , RAW 264.7 Cells , Receptors, Immunologic , Signal Transduction/genetics , Signal Transduction/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology , Ubiquitination/genetics , Ubiquitination/immunology
9.
J Cell Mol Med ; 23(5): 3737-3746, 2019 05.
Article in English | MEDLINE | ID: mdl-30895711

ABSTRACT

Adipose-derived stem cells (ASCs) are highly attractive for cell-based therapies in tissue repair and regeneration because they have multilineage differentiation capacity and are immunosuppressive. However, the detailed epigenetic mechanisms of their immunoregulatory capacity are not fully defined. In this study, we found that Mysm1 was induced in ASCs treated with inflammatory cytokines. Adipose-derived stem cells with Mysm1 knockdown exhibited attenuated immunosuppressive capacity, evidenced by less inhibition of T cell proliferation, more pro-inflammatory factor secretion and less nitric oxide (NO) production in vitro. Mysm1-deficient ASCs exacerbated inflammatory bowel diseases but inhibited tumour growth in vivo. Mysm1-deficient ASCs also showed depressed miR-150 expression. When transduced with Mysm1 overexpression lentivirus, ASCs exhibited enhanced miR-150 expression. Furthermore, Mysm1-deficient cells transduced with lentivirus containing miR-150 mimics produced less pro-inflammatory factors and more NO. Our study reveals a new role of Mysm1 in regulating the immunomodulatory activities of ASCs by targeting miR-150. These novel insights into the mechanisms through which ASCs regulate immune reactions may lead to better clinical utility of these cells.


Subject(s)
Adipose Tissue/cytology , Epigenesis, Genetic/immunology , MicroRNAs/immunology , Stem Cells/immunology , Trans-Activators/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression/drug effects , Gene Expression/immunology , Interferon-gamma/pharmacology , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Nitric Oxide/immunology , Nitric Oxide/metabolism , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
10.
J Exp Med ; 215(11): 2850-2867, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30224386

ABSTRACT

Th2 immune response is critical for allergic asthma pathogenesis. Molecular mechanisms for regulating Th2 immunity are still not well understood. Here we report that the ubiquitin-specific protease USP38 is crucial for Th2-mediated allergic asthma. TCR stimulation up-regulated the USP38 level, and USP38 in turn mediated the protein stabilization of JunB, a transcription factor specific for Th2 development. Consequently, USP38 was specifically required for TCR-induced production of Th2 cytokines and Th2 development both in vitro and in vivo, and USP38-deficient mice were resistant to asthma pathogenesis induced by OVA or HDM. Mechanistically, USP38 directly associated with JunB, deubiquitinated Lys-48-linked poly-ubiquitination of JunB, and consequently blocked TCR-induced JunB turnover. USP38 represents the first identified deubiquitinase specifically for Th2 immunity and the associated asthma.


Subject(s)
Asthma/immunology , Th2 Cells/immunology , Transcription Factors/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Asthma/genetics , Asthma/pathology , Cytokines/genetics , Cytokines/immunology , Mice , Mice, Knockout , Polyubiquitin/genetics , Polyubiquitin/immunology , Protein Stability , Th2 Cells/pathology , Transcription Factors/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
11.
Monoclon Antib Immunodiagn Immunother ; 37(4): 180-184, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30130141

ABSTRACT

Ovarian tumor domain-containing protein 1 (OTUD1), an OTU-family deubiquitinating enzyme, has been reported to be involved in cancer progression through the regulation of p53 and SMAD7. However, the precise pathophysiological functions of OTUD1 remain elusive. Here, we report the establishment of OTUD1-specific monoclonal antibodies (mAbs), using the rat medial iliac lymph node method. The generated antibodies recognize the N-terminal portion (aa. 1-290) of human and mouse OTUD1 proteins. In addition, immunofluorescent staining and subcellular fractionation analyses using these antibodies indicated that OTUD1 is predominantly localized in the cytosol. Thus, these mAbs can be further used to elucidate cellular functions of OTUD1 and its involvement in processes such as cancer progression.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/immunology , Ubiquitin-Specific Proteases/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Humans , Lymph Nodes/immunology , Mice , Neoplasms/genetics , Neoplasms/pathology , Rats , Smad7 Protein/immunology , Tumor Suppressor Protein p53/immunology , Ubiquitin-Specific Proteases/antagonists & inhibitors
12.
J Autoimmun ; 94: 156-165, 2018 11.
Article in English | MEDLINE | ID: mdl-30100102

ABSTRACT

Dysregulation of innate immunity accompanied by excessive interferon production contributes to autoimmune disease. However, the mechanism by which the immune response is modulated in autoimmune disorders is largely unknown. Here we identified loss-of-function mutations of OTUD1 associated with multiple autoimmune diseases. Under inflammatory conditions, inducible OTUD1 acts as an immune checkpoint and blocks RIG-I-like receptors signaling. As a deubiquitinase, OTUD1 directly interacts with transcription factor IRF3 and removes the K63-linked poly-ubiquitin chains on IRF3 Lysine 98, which inhibits IRF3 nuclear translocation and transcriptional activity. In contrast, OTUD1 mutants impair its suppressive effects on IRF3 via attenuating the OTUD1 deubiquinase activity or its association with IRF3. Moreover, we found FOXO3 signaling is required for OTUD1 induction upon antigenic stimulation. Our data demonstrate that OTUD1 is involved in maintaining immune homeostasis and loss-of-function mutations of OTUD1 enhance the immune response and are associated with autoimmunity.


Subject(s)
Arthritis, Rheumatoid/genetics , Colitis, Ulcerative/genetics , Hashimoto Disease/genetics , Lupus Erythematosus, Systemic/genetics , Lymphocytes/immunology , Ubiquitin-Specific Proteases/genetics , Adult , Amino Acid Sequence , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Case-Control Studies , Cell Line, Tumor , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/immunology , Gene Expression Regulation , HEK293 Cells , Hashimoto Disease/immunology , Hashimoto Disease/pathology , Homeostasis/immunology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lymphocytes/pathology , Male , Mutation , Protein Transport , Receptors, Immunologic , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Ubiquitin-Specific Proteases/immunology
14.
PLoS Pathog ; 14(5): e1007067, 2018 05.
Article in English | MEDLINE | ID: mdl-29734366

ABSTRACT

During RNA virus infection, the adaptor protein MAVS recruits TRAF3 and TRAF6 to form a signalosome, which is critical to induce the production of type I interferons (IFNs) and proinflammatory cytokines. While activation of the MAVS/TRAF3/TRAF6 signalosome is well studied, the negative regulation of the signalosome remains largely unknown. Here we report that RNA viruses specifically promote the deubiquitinase OTUD1 expression by NF-κB-dependent mechanisms at the early stage of viral infection. Furthermore, OTUD1 upregulates protein levels of intracellular Smurf1 by removing Smurf1 ubiquitination. Importantly, RNA virus infection promotes the binding of Smurf1 to MAVS, TRAF3 and TRAF6, which leads to ubiquitination-dependent degradation of every component of the MAVS/TRAF3/TRAF6 signalosome and subsequent potent inhibition of IFNs production. Consistently, OTUD1-deficient mice produce more antiviral cytokines and are more resistant to RNA virus infection. Our findings reveal a novel immune evasion mechanism exploited by RNA viruses, and elucidate a negative feedback loop of MAVS/TRAF3/TRAF6 signaling mediated by the OTUD1-Smurf1 axis during RNA virus infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate/immunology , RNA Viruses/physiology , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , RNA Virus Infections/immunology , RNA Virus Infections/prevention & control , RNA, Messenger/metabolism , Signal Transduction , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology
15.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263274

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a key pattern recognition receptor that senses viral RNA and interacts with the mitochondrial adaptor MAVS, triggering a signaling cascade that results in the production of type I interferons (IFNs). This signaling axis is initiated by K63-linked ubiquitination of RIG-I mediated by the E3 ubiquitin ligase TRIM25, which promotes the interaction of RIG-I with MAVS. USP15 was recently identified as an upstream regulator of TRIM25, stabilizing the enzyme through removal of degradative K48-linked polyubiquitin, ultimately promoting RIG-I-dependent cytokine responses. Here, we show that the E6 oncoprotein of human papillomavirus type 16 (HPV16) as well as of other HPV types form a complex with TRIM25 and USP15 in human cells. In the presence of E6, the K48-linked ubiquitination of TRIM25 was markedly increased, and in line with this, TRIM25 degradation was enhanced. Our results further showed that E6 inhibited the TRIM25-mediated K63-linked ubiquitination of RIG-I and its CARD-dependent interaction with MAVS. HPV16 E6, but not E7, suppressed the RIG-I-mediated induction of IFN-ß, chemokines, and IFN-stimulated genes (ISGs). Finally, CRISPR-Cas9 gene targeting in human keratinocytes showed that the TRIM25-RIG-I-MAVS triad is important for eliciting an antiviral immune response to HPV16 infection. Our study thus identifies a novel immune escape mechanism that is conserved among different HPV strains and further indicates that the RIG-I signaling pathway plays an important role in the innate immune response to HPV infection.IMPORTANCE Persistent infection and tumorigenesis by HPVs are known to require viral manipulation of a variety of cellular processes, including those involved in innate immune responses. Here, we show that the HPV E6 oncoprotein antagonizes the activation of the cytoplasmic innate immune sensor RIG-I by targeting its upstream regulatory enzymes TRIM25 and USP15. We further show that the RIG-I signaling cascade is important for an antiviral innate immune response to HPV16 infection, providing evidence that RIG-I, whose role in sensing RNA virus infections has been well characterized, also plays a crucial role in the antiviral host response to small DNA viruses of the Papillomaviridae family.


Subject(s)
DEAD Box Protein 58/immunology , Human papillomavirus 6/immunology , Immunity, Innate , Keratinocytes/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Specific Proteases/immunology , DEAD Box Protein 58/genetics , HEK293 Cells , Human papillomavirus 6/genetics , Humans , Keratinocytes/pathology , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Receptors, Immunologic , Signal Transduction/genetics , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Specific Proteases/genetics
16.
Poult Sci ; 97(3): 1022-1031, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29267974

ABSTRACT

Ubiquitin-specific protease 18 (USP18) is known as an inhibition factor and has been associated with the innate immune response to pathogens. USP18 is the only deconjugating protease with specificity for interferon-stimulated gene 15 (ISG15), which is supposed to be missing in birds. To analyze the efficacy of goose USP18 (goUSP18) against Tembusu virus (TMUV) infection, we first cloned USP18 homologous cDNA from TMUV infected geese. The coding sequence was 1131 bp, and the deduced amino acid sequence shared conserved motifs with its homologues. Tissue-specific expression has shown that goUSP18 transcripts are strongly expressed in the spleen and liver of adult geese, as well as in the pancreas of goslings. Moreover, the goUSP18 transcripts were induced by goose interferons (goIFN) in goose embryo fibroblasts (GEF) and by TLR ligands in peripheral blood mononuclear cells (PBMC). Notably, goUSP18 transcripts were highly up-regulated by TMUV infection compared to the basal level in uninfected birds. Taken together, these results suggested that goUSP18 was involved in host innate immunity against TMUV infection.


Subject(s)
Geese/genetics , Geese/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Poultry Diseases/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Avian Proteins/immunology , Flavivirus/physiology , Flavivirus Infections/immunology , Flavivirus Infections/veterinary , Gene Expression Profiling/veterinary , Interferons/genetics , Leukocytes, Mononuclear/immunology , Sequence Alignment/veterinary , Ubiquitin-Specific Proteases/chemistry
17.
Int J Mol Med ; 40(4): 979-986, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28791349

ABSTRACT

We speculated that ubiquitin specific peptidase 4 (USP4) may deubiquitinate interferon regulatory factor 4 (IRF4) and affect T helper type 2 (Th2) cell function. This study aimed to validate this hypothesis. Here, the interaction between USP4 and IRF4 were analyzed by co-immunoprecipitation assay. The deubiquitin effect of USP4 on IRF4 was analyzed by the Ni-NTA pull down assay. Luciferase reporter gene constructs were used to analyze the effects of USP4, IRF4 and nuclear factor of activated T cell-2 (NFATc2) on the activation of the interleukin-4 (IL-4) promoter. Then, the Th2 cells were infected with sh-USP4 to analyze the effects of USP4 on the expression levels of IRF4 and Th2-related cytokines. Western blotting and RT-qPCR were used to detect the protein and mRNA expression levels, respectively. To determine the levels of IL-4 and IRF4 in rheumatic heart disease (RHD) patients, peripheral blood mononuclear cells (PBMCs) were separated by density gradient centrifugation from RHD patients and healthy controls, and flow cytometric analysis was performed. Our results validated the interaction between USP4 and IRF4, and effects of USP4 on stabilization and deubiquitination of IRF4 were also found. Importantly, USP4 and IRF4 synergized with NFATc2 to specifically enhance NFAT-mediated activation of the IL-4 promoter. USP4 knockdown not only decreased the expression level of IRF4, but also affected the expression level of Th2-related cytokines. Finally, the increased level of IL-4 and IRF4 in PBMCs of RHD patients were observed. On the whole, our data indicate that USP4 interacts with and deubiquitinates IRF4, and also stabilizes IRF4 protein and promotes IRF4 function to facilitate IL-4 expression in Th2 cells, which may be related to the pathological process of RHD.


Subject(s)
Interferon Regulatory Factors/genetics , Interleukin-4/genetics , RNA, Small Interfering/genetics , Rheumatic Heart Disease/genetics , Th2 Cells/immunology , Ubiquitin-Specific Proteases/genetics , Case-Control Studies , Cell Separation , Female , Gene Expression Regulation , Genes, Reporter , Humans , Interferon Regulatory Factors/immunology , Interleukin-4/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Luciferases/genetics , Luciferases/metabolism , Male , Middle Aged , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/immunology , Rheumatic Heart Disease/immunology , Rheumatic Heart Disease/pathology , Signal Transduction , Th2 Cells/pathology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/immunology
18.
Mol Cell ; 64(2): 267-281, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27692986

ABSTRACT

TBK1 is a component of the type I interferon (IFN) signaling pathway, yet the mechanisms controlling its activity and degradation remain poorly understood. Here we report that USP38 negatively regulates type I IFN signaling by targeting the active form of TBK1 for degradation in vitro and in vivo. USP38 specifically cleaves K33-linked poly-ubiquitin chains from TBK1 at Lys670, and it allows for subsequent K48-linked ubiquitination at the same position mediated by DTX4 and TRIP. Knockdown or knockout of USP38 increases K33-linked ubiquitination, but it abrogates K48-linked ubiquitination and degradation of TBK1, thus enhancing type I IFN signaling. Our findings identify an essential role for USP38 in negatively regulating type I IFN signaling, and they provide insights into the mechanisms by which USP38 regulates TBK1 ubiquitination through the NLRP4 signalosome.


Subject(s)
Immunity, Innate , Interferon Type I/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Signal Transduction/immunology , Ubiquitin-Specific Proteases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/immunology , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions , Interferon Type I/genetics , Interferon Type I/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/virology , Mice , Mice, Knockout , Phosphorylation , Polyubiquitin/genetics , Polyubiquitin/immunology , Polyubiquitin/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proteins/genetics , Proteins/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/immunology , Ubiquitination , Vesiculovirus/growth & development , Vesiculovirus/immunology
19.
Nat Immunol ; 17(3): 259-68, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808229

ABSTRACT

The proinflammatory cytokines interleukin 12 (IL-12) and IL-23 connect innate responses and adaptive immune responses and are also involved in autoimmune and inflammatory diseases. Here we describe an epigenetic mechanism for regulation of the genes encoding IL-12 (Il12a and Il12b; collectively called 'Il12' here) and IL-23 (Il23a and Il12b; collectively called 'Il23' here) involving the deubiquitinase Trabid. Deletion of Zranb1 (which encodes Trabid) in dendritic cells inhibited induction of the expression of Il12 and Il23 by Toll-like receptors (TLRs), which impaired the differentiation of inflammatory T cells and protected mice from autoimmune inflammation. Trabid facilitated TLR-induced histone modifications at the promoters of Il12 and Il23, which involved deubiqutination and stabilization of the histone demethylase Jmjd2d. Our findings highlight an epigenetic mechanism for the regulation of Il12 and Il23 and establish Trabid as an innate immunological regulator of inflammatory T cell responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Epigenesis, Genetic , Interleukin-12/genetics , Interleukin-23/genetics , Ubiquitin-Specific Proteases/genetics , Animals , Cell Differentiation , Chromatin Immunoprecipitation , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Gene Expression Regulation , Gene Knockdown Techniques , Immunoblotting , Immunoprecipitation , Interleukin-12/immunology , Interleukin-23/immunology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptors/metabolism , Ubiquitin-Specific Proteases/immunology , Zinc Fingers/genetics , Zinc Fingers/immunology
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 44(5): 578-83, 2015 09.
Article in Chinese | MEDLINE | ID: mdl-26713535

ABSTRACT

Ubiquitin-specific protease(USP), which belongs to cysteine protease, is an important member of the deubiquitinating enzyme family(DUB). USP plays an important role in the immune response against viral infections, in which it can regulate the production of type I interferon through various ways to initiate or weaken the antiviral immune response. USP2b, USP3, USP18, USP25, UL36USP and HAUSP play a role of antivirus; while USP4, USP13, USP15 and USP17 negatively regulate antiviral immune response. In this article we review the recent progress on roles of USP family in antiviral immune response.


Subject(s)
Ubiquitin-Specific Proteases/immunology , Virus Diseases/immunology , Humans , Interferon Type I/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...