Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
1.
Dev Comp Immunol ; 156: 105181, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636698

ABSTRACT

Interferon regulatory factor 7 (IRF7) is considered the master regulator of virus-induced interferon (IFN) production. However, to avoid an autoimmune response, the expression of IRF7 must be tightly controlled. In this study, we report that zebrafish ubiquitin-specific protease 8 (USP8) promotes IRF7 degradation through an autophagy-lysosome-dependent pathway to inhibit IFN production. First, zebrafish usp8 is induced upon spring viremia of carp virus (SVCV) infection and polyinosinic/polycytidylic acid (poly I:C) stimulation. Second, overexpression of USP8 suppresses SVCV or poly I:C-mediated IFN expression. Mechanistically, USP8 interacts with IRF7 and promotes its degradation via an autophagy-lysosome-dependent pathway. Finally, USP8 significantly suppresses cellular antiviral responses and enhances SVCV proliferation. In summary, our discoveries offer a perspective on the role of zebrafish USP8 and provide additional understanding of the regulation of IRF7 in host antiviral immune response.


Subject(s)
Autophagy , Interferon Regulatory Factor-7 , Interferon Regulatory Factors , Lysosomes , Rhabdoviridae , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/immunology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Autophagy/immunology , Lysosomes/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Rhabdoviridae/physiology , Rhabdoviridae/immunology , Interferons/metabolism , Poly I-C/immunology , Rhabdoviridae Infections/immunology , Proteolysis , Fish Diseases/immunology , Fish Diseases/virology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Humans , Immunity, Innate
2.
J Virol ; 97(7): e0053223, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37367226

ABSTRACT

During viral infection, host defensive proteins either enhance the host immune response or antagonize viral components directly. In this study, we report on the following two mechanisms employed by zebrafish mitogen-activated protein kinase kinase 7 (MAP2K7) to protect the host during spring viremia of carp virus (SVCV) infection: stabilization of host IRF7 and degradation of SVCV P protein. In vivo, map2k7+/- (map2k7-/- is a lethal mutation) zebrafish showed a higher lethality, more pronounced tissue damage, and more viral proteins in major immune organs than the controls. At the cellular level, overexpression of map2k7 significantly enhanced host cell antiviral capacity, and viral replication and proliferation were significantly suppressed. Additionally, MAP2K7 interacted with the C terminus of IRF7 and stabilized IRF7 by increasing K63-linked polyubiquitination. On the other hand, during MAP2K7 overexpression, SVCV P proteins were significantly decreased. Further analysis demonstrated that SVCV P protein was degraded by the ubiquitin-proteasome pathway, as the attenuation of K63-linked polyubiquitination was mediated by MAP2K7. Furthermore, the deubiquitinase USP7 was indispensable in P protein degradation. These results confirm the dual functions of MAP2K7 during viral infection. IMPORTANCE Normally, during viral infection, host antiviral factors individually modulate the host immune response or antagonize viral components to defense infection. In the present study, we report that zebrafish MAP2K7 plays a crucial positive role in the host antiviral process. According to the weaker antiviral capacity of map2k7+/- zebrafish than that of the control, we find that MAP2K7 reduces host lethality through two pathways, as follows: enhancing K63-linked polyubiquitination to promote host IRF7 stability and attenuating K63-mediated polyubiquitination to degrade the SVCV P protein. These two mechanisms of MAP2K7 reveal a special antiviral response in lower vertebrates.


Subject(s)
Fish Diseases , Interferon Regulatory Factors , Mitogen-Activated Protein Kinases , Rhabdoviridae Infections , Ubiquitination , Viral Structural Proteins , Animals , Fish Diseases/immunology , Fish Diseases/virology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Rhabdoviridae/genetics , Rhabdoviridae/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Zebrafish/genetics , Zebrafish/immunology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Protein Stability , Proteolysis , Viral Structural Proteins/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Up-Regulation
3.
J Immunol ; 208(5): 1189-1203, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35101889

ABSTRACT

The small HERC family currently comprises four members (HERC3-6) involved in the regulation of various physiological activities. Little is known about the role of HERCs in IFN response. In this study, we identify a novel fish HERC member, named crucian carp HERC7, as a negative regulator of fish IFN response. Genome-wide search of homologs and comprehensive phylogenetic analyses reveal that the small HERC family, apart from HERC3-6 that have been well-characterized in mammals, contains a novel HERC7 subfamily exclusively in nonmammalian vertebrates. Lineage-specific and even species-specific expansion of HERC7 subfamily in fish indicates that crucian carp HERC7 might be species-specific. In virally infected fish cells, HERC7 is induced by IFN and selectively targets three retinoic acid-inducible gene-I-like receptor signaling factors for degradation to attenuate IFN response by two distinct strategies. Mechanistically, HERC7 delivers mediator of IFN regulatory factor 3 activator and mitochondrial antiviral signaling protein for proteasome-dependent degradation at the protein level and facilitates IFN regulatory factor 7 transcript decay at the mRNA level, thus abrogating cellular IFN induction to promote virus replication. Whereas HERC7 is a putative E3 ligase, the E3 ligase activity is not required for its negative regulatory function. These results demonstrate that the ongoing expansion of the small HERC family generates a novel HERC7 to fine-tune fish IFN antiviral response.


Subject(s)
Interferon Regulatory Factor-7/metabolism , Interferons/immunology , Reoviridae/immunology , Rhabdoviridae/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Carps , Cell Line , Fish Proteins/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-7/genetics , Membrane Proteins/metabolism , RNA Stability/genetics , RNA, Messenger/genetics , Signal Transduction/immunology , Trans-Activators/genetics
4.
J Immunol ; 208(5): 1099-1114, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35101892

ABSTRACT

Circular RNA (circRNA) is produced by splicing head to tail and is widely distributed in multicellular organisms, and circRNA reportedly can participate in various cell biological processes. In this study, we discovered a novel exon-intron circRNA derived from probable E3 ubiquitin-protein ligase RNF217 (RNF217) gene, namely, circRNF217, which was related to the antibacterial responses in teleost fish. Results indicated that circRNF217 played essential roles in host antibacterial immunity and inhibited the Vibrio anguillarum invasion into cells. Our study also found a microRNA miR-130-3p, which could inhibit antibacterial immune response and promote V. anguillarum invasion into cells by targeting NOD1. Moreover, we also found that the antibacterial effect inhibited by miR-130-3p could be reversed with circRNF217. In mechanism, our data revealed that circRNF217 was a competing endogenous RNA of NOD1 by sponging miR-130-3p, leading to activation of the NF-κB pathway and then enhancing the innate antibacterial responses. In addition, we also found that circRNF217 can promote the antiviral response caused by Siniperca chuatsi rhabdovirus through targeting NOD1. Our study provides new insights for understanding the impact of circRNA on host-pathogen interactions and formulating fish disease prevention to resist the severely harmful V. anguillarum infection.


Subject(s)
Fish Diseases/immunology , Immunity, Innate/immunology , MicroRNAs/genetics , Perches/immunology , RNA, Circular/genetics , Vibrio/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fish Diseases/microbiology , Fish Diseases/prevention & control , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Perches/virology , Rhabdoviridae/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Front Immunol ; 12: 771277, 2021.
Article in English | MEDLINE | ID: mdl-34868031

ABSTRACT

Histone H2A is a nuclear molecule tightly associated in the form of the nucleosome. Our previous studies have demonstrated the antibacterial property of piscine H2A variants against gram-negative bacteria Edwardsiella piscicida and Gram-positive bacteria Streptococcus agalactiae. In this study, we show the function and mechanism of piscine H2A in the negative regulation of RLR signaling pathway and host innate immune response against spring viremia of carp virus (SVCV) infection. SVCV infection significantly inhibits the expression of histone H2A during an early stage of infection, but induces the expression of histone H2A during the late stage of infection such as at 48 and 72 hpi. Under normal physiological conditions, histone H2A is nuclear-localized. However, SVCV infection promotes the migration of histone H2A from the nucleus to the cytoplasm. The in vivo studies revealed that histone H2A overexpression led to the increased expression of SVCV gene and decreased survival rate. The overexpression of histone H2A also significantly impaired the expression levels of those genes involved in RLR antiviral signaling pathway. Furthermore, histone H2A targeted TBK1 and IRF3 to promote their protein degradation via the lysosomal pathway and impair the formation of TBK1-IRF3 functional complex. Importantly, histone H2A completely abolished TBK1-mediated antiviral activity and enormously impaired the protein expression of IRF3, especially nuclear IRF3. Further analysis demonstrated that the inhibition of histone H2A nuclear/cytoplasmic trafficking could relieve the protein degradation of TBK1 and IRF3, and blocked the negative regulation of histone H2A on the SVCV infection. Collectively, our results suggest that histone H2A nuclear/cytoplasmic trafficking is essential for negative regulation of RLR signaling pathway and antiviral immune response in response to SVCV infection.


Subject(s)
Histones/immunology , Immunity, Innate/immunology , Interferon Regulatory Factor-3/immunology , Lysosomes/immunology , Protein Serine-Threonine Kinases/immunology , Rhabdoviridae/immunology , Zebrafish Proteins/immunology , Zebrafish/immunology , Animals , Cell Line , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cytoplasm/immunology , Cytoplasm/metabolism , Gene Expression Regulation/immunology , Histones/genetics , Histones/metabolism , Host-Pathogen Interactions/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Larva/immunology , Larva/metabolism , Larva/virology , Lysosomes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport/immunology , Proteolysis , Rhabdoviridae/physiology , Zebrafish/metabolism , Zebrafish/virology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Front Immunol ; 12: 769775, 2021.
Article in English | MEDLINE | ID: mdl-34804060

ABSTRACT

The crosstalk between the immune system and microbiota drives an amazingly complex mutualistic symbiosis. In mammals, the upper respiratory tract acts as a gateway for pathogen invasion, and the dynamic interaction between microbiota and mucosal immunity on its surface can effectively prevent disease development. However, the relationship between virus-mediated mucosal immune responses and microbes in lower vertebrates remains uncharacterized. In this study, we successfully constructed an infection model by intraperitoneally injecting common carp (Cyprinus carpio) with spring viremia of carp virus (SVCV). In addition to the detection of the SVCV in the nose and pharynx of common carp, we also identified obvious histopathological changes following viral infection. Moreover, numerous immune-related genes were significantly upregulated in the nose and pharynx at the peak of SVCV infection, after which the expression levels decreased to levels similar to those of the control group. Transcriptome sequencing results revealed that pathways associated with bacterial infection in the Toll-like receptor pathway and the Nod-like receptor pathway were activated in addition to the virus-related Rig-I-like receptor pathway after SVCV infection, suggesting that viral infection may be followed by opportunistic bacterial infection in these mucosal tissues. Using 16S rRNA gene sequencing, we further identified an upward trend in pathogenic bacteria on the mucosal surface of the nose and pharynx 4 days after SVCV infection, after which these tissues eventually reached new homeostasis. Taken together, our results suggest that the dynamic interaction between mucosal immunity and microbiota promotes the host to a new ecological state.


Subject(s)
Bacteria/immunology , Carps/immunology , Fish Diseases/immunology , Immunity, Mucosal/immunology , Pharynx/immunology , Rhabdoviridae/immunology , Animal Structures/immunology , Animal Structures/microbiology , Animal Structures/virology , Animals , Bacteria/classification , Bacteria/genetics , Carps/microbiology , Carps/virology , Fish Diseases/microbiology , Fish Diseases/virology , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/methods , Homeostasis/genetics , Homeostasis/immunology , Immunity, Mucosal/genetics , Pharynx/microbiology , Pharynx/virology , Phylogeny , RNA, Ribosomal, 16S/genetics , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Rhabdoviridae/genetics , Rhabdoviridae/physiology , Signal Transduction/genetics , Signal Transduction/immunology
7.
J Immunol ; 207(10): 2570-2580, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34654690

ABSTRACT

TNFR-associated factor 6 (TRAF6) not only recruits TBK1/IKKε to MAVS upon virus infection but also catalyzes K63-linked polyubiquitination on substrate or itself, which is critical for NEMO-dependent and -independent TBK1/IKKε activation, leading to the production of type I IFNs. The regulation at the TRAF6 level could affect the activation of antiviral innate immunity. In this study, we demonstrate that zebrafish prmt2, a type I arginine methyltransferase, attenuates traf6-mediated antiviral response. Prmt2 binds to the C terminus of traf6 to catalyze arginine asymmetric dimethylation of traf6 at arginine 100, preventing its K63-linked autoubiquitination, which results in the suppression of traf6 activation. In addition, it seems that the N terminus of prmt2 competes with mavs for traf6 binding and prevents the recruitment of tbk1/ikkε to mavs. By zebrafish model, we show that loss of prmt2 promotes the survival ratio of zebrafish larvae after challenge with spring viremia of carp virus. Therefore, we reveal, to our knowledge, a novel function of prmt2 in the negative regulation of antiviral innate immunity by targeting traf6.


Subject(s)
Immunity, Innate/immunology , Protein-Arginine N-Methyltransferases/immunology , Rhabdoviridae Infections/immunology , TNF Receptor-Associated Factor 6/immunology , Animals , Rhabdoviridae/immunology , Zebrafish
8.
J Immunol ; 207(2): 371-375, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34233911

ABSTRACT

The repertoire of Abs is generated by genomic rearrangements during B cell differentiation. Although V(D)J rearrangements lead to repertoires mostly different between individuals, recent studies have shown that they contain a substantial fraction of overrepresented and shared "public" clones. We previously reported a strong public IgHµ clonotypic response against the rhabdovirus viral hemorrhagic septicemia virus in a teleost fish. In this study, we identified an IgL chain associated with this public response that allowed us to characterize its functionality. We show that this public Ab response has a potent neutralizing capacity that is typically associated with host protection during rhabdovirus infections. We also demonstrate that the public response is not restricted to a particular trout isogenic line but expressed in multiple genetic backgrounds and may be used as a marker of successful vaccination. Our work reveals that public B cell responses producing generic Abs constitute a mechanism of protection against infection conserved across vertebrates.


Subject(s)
Antibody Formation/immunology , Fishes/immunology , Mammals/immunology , Animals , B-Lymphocytes/immunology , Clone Cells/immunology , Rhabdoviridae/immunology , Rhabdoviridae Infections/immunology , V(D)J Recombination/immunology , Vaccination/methods
9.
J Fish Dis ; 44(10): 1587-1594, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34165796

ABSTRACT

Spring viraemia of carp (SVC) caused by spring viraemia of carp virus (SVCV) can infect almost all fish of cyprinids, which bring huge economic losses to aquaculture. Glycoprotein (G), as the most important antigenic determinant protein of SVCV, is widely considered as an effective method against SVCV. In our previous study, we found that G3 (131 aa) is the potential dominant antigen epitope that induces strong immune responses similar to G protein (510 aa). Here, in order to further improve the immune effect, we reported a subunit vaccine (PEG-G3) constructed by PEG-modified dominant epitope protein (G3). The results of serum antibody production, enzyme activities and immune-related genes expression showed that PEG-G3 induces significantly stronger immune protective responses against SVCV than G3. PEG modification significantly increased the serum antibody level of the vaccine, which increased significantly after immunization and reached the peak at 21 day post-vaccination. T-AOC and AKP activities in the lowest concentration group (5 µg) of PEG-G3 were significantly higher than those in the highest concentration group (20 µg) of G3. In PEG-G3 group, the expression of almost all genes increased at least 4 times compared with the control group. After 14-day challenge, the RPS (relative percentage survival) of the highest concentration of PEG-G3 group was 53.6%, while that of G3 group is 38.9%. Therefore, this work shows that PEG modification and dominant epitope screening may be effective methods to improve the immune protective effect of vaccines and to resist the infection of aquatic animal viral diseases.


Subject(s)
Carps , Fish Diseases/prevention & control , Immunization/veterinary , Rhabdoviridae Infections/veterinary , Rhabdoviridae/immunology , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Animals , Epitopes/immunology , Fish Diseases/virology , Immunity , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/virology , Vaccines, Subunit/administration & dosage , Viral Vaccines/administration & dosage
10.
Dev Comp Immunol ; 122: 104110, 2021 09.
Article in English | MEDLINE | ID: mdl-33933533

ABSTRACT

Toll/interleukin-1 receptor (TIR) domain-containing adaptors, serve as pivotal signal transduction molecules in Toll-like receptor (TLR) signalling pathway to mediate downstream signalling cascades. In this study, four TIR-domain containing adaptors, MyD88, TRIF, MAL and SARM, were identified in mandarin fish Siniperca chuatsi, and they all contain TIR domains, of which MyD88 and SARM had high sequence homology with their vertebrate homologues. The expression analysis at mRNA level indicated that these genes were ubiquitously distributed in different tissues, being high in immune- and mucosa-related tissues such as head-kidney and intestine. The transcripts of these adaptor genes were up-regulated by poly(I:C) and LPS stimulation in isolated head-kidney lymphocytes (HKLs) of mandarin fish. Fluorescence microscopy revealed that all these molecules were localized in cytoplasm, and further investigations showed that the over-expression of MyD88, TRIF and MAL activated the NF-κB, ISRE or type Ι IFN promoters and inhibited SVCV replication, whereas their antiviral effects were significantly impaired when co-transfected with SARM. It was also confirmed by co-immunoprecipitation (Co-IP) that SARM interacts separately with MyD88, TRIF and MAL, and MAL interacts with MyD88. However, the regulatory mechanisms of these adaptors involved in signalling pathways of different TLRs should be of interest for further research.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Armadillo Domain Proteins/metabolism , Lymphocytes/immunology , Myeloid Differentiation Factor 88/metabolism , Perciformes/immunology , Receptors, Interleukin-1/metabolism , Rhabdoviridae/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Armadillo Domain Proteins/genetics , Cell Line , Fish Diseases/immunology , Fish Diseases/virology , HEK293 Cells , Head Kidney/cytology , Head Kidney/immunology , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Lipopolysaccharides/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Perciformes/virology , Poly I-C/immunology , Protein Domains , Receptors, Interleukin-1/genetics , Signal Transduction/physiology , Transcriptional Activation , Virus Replication/immunology
11.
Dev Comp Immunol ; 122: 104134, 2021 09.
Article in English | MEDLINE | ID: mdl-34000319

ABSTRACT

TUFM is a mitochondrial protein and serves as a regulator of antiviral signaling; nevertheless, the character of TUFM in teleosts remains unidentified. In this study, TUFM homologue of black carp (Mylopharyngodon piceus) has been characterized and its role in innate immunity has been explored. Black carp TUFM (bcTUFM) comprises 447 amino acids and shows the high similarity to human TUFM. bcTUFM was about 50 kDa in the Western blot assay and was determined as a cytosolic protein by immunofluorescent microscopy. Knockdown of bcTUFM by shRNA enhanced the antiviral ability of the host cells. The induction fold of interferon promoter transcription in the cells co-expressing bcTUFM and bcMAVS was much lower than that of the cells expressing bcMAVS alone. Our previous study has identified that bcNLRX1 interacted with bcMAVS and functioned as an inhibitor of bcMAVS. The interaction between bcTUFM and bcNLRX1, but not bcTUFM and bcMAVS, was detected through co-immunoprecipitation. The subsequent reporter assay and plaque assay demonstrated that the inhibition of bcMAVS-mediated interferon production and antiviral activity by bcNLRX1 was enhanced by co-expressed bcTUFM. Thus, our data suggests that bcTUFM cooperates with bcNLRX1 to inhibit bcMAVS-mediated antiviral signaling during host antiviral innate immune response against SVCV.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carps/immunology , Immunity, Innate/immunology , Mitochondrial Proteins/metabolism , Peptide Elongation Factor Tu/metabolism , Rhabdoviridae/immunology , Animals , Carps/metabolism , Carps/virology , Cell Line , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Mitochondrial Proteins/genetics , Peptide Elongation Factor Tu/genetics , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Signal Transduction/immunology
12.
Dev Comp Immunol ; 122: 104105, 2021 09.
Article in English | MEDLINE | ID: mdl-33872658

ABSTRACT

Mammalian Nemo-like kinase (NLK) plays important roles in multiple biological processes including immune response; however, the roles of teleost NLK remain largely unknown. In the present study, the NLK homolog (bcNLK) of black carp (Mylopharyngodon piceus) has been cloned and characterized. The coding region of bcNLK consists of 1427 nucleotides and encodes 476 amino acid, including two low complexity region (LCR) domains at the N-terminus and a serine/threonine protein kinase catalytic (S-TKc) domain in the middle region. The transcription of bcNLK are promoted after spring viremia of carp virus (SVCV) infection and poly (I:C) stimulation in host cells, but not post LPS treatment. bcNLK exhibits weak impact on the transcription of interferon (IFN) promoter in the reporter assay, however, black carp MAVS (bcMAVS)-mediated IFN promoter transcription is remarkably dampened by bcNLK. The interaction between bcNLK and bcMAVS is detected through the co-immunoprecipitation assay. Accordingly, the plaque assay results show that bcMAVS-mediated antiviral ability is impaired by bcNLK. Moreover, knockdown of bcNLK in host cells leads to the enhanced antiviral ability against SVCV. All these data support the conclusion that black carp NLK associates with MAVS and inhibited MAVS-mediated antiviral signaling.


Subject(s)
Carps/immunology , Fish Diseases/immunology , Immunity, Innate/immunology , Interferons/immunology , Mitogen-Activated Protein Kinases/immunology , Rhabdoviridae/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Base Sequence , Carps/virology , Catalytic Domain/genetics , Cell Line , Cloning, Molecular , Fish Diseases/virology , HEK293 Cells , Humans , Immunomodulation/immunology , Interferons/genetics , Mitogen-Activated Protein Kinases/genetics , Poly I-C/immunology , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics , Sequence Analysis, DNA
13.
Front Immunol ; 12: 654758, 2021.
Article in English | MEDLINE | ID: mdl-33897703

ABSTRACT

The mucosa of vertebrates is a particularly complex but dynamic environment in which the host constantly interacts with trillions of commensal microorganisms and pathogens. Although the internal and external mucosal microbiomes with immune defense of mammals have been well investigated, the relationship between mucosal microbes and their host's immune responses has not been systematically understood in the early vertebrates. In this study, we compared the composition and distribution of mucosal microbiota in common carp (Cyprinus carpio), and found that there were significant differences of microbiota between in the internal (gut) and external mucosal (buccal mucosa, gills and skin) tissues. Next, we successfully constructed an infection model with spring viremia of carp virus (SVCV). Specifically, following viral infection, the immune and antiviral related genes showed different up-regulation in all selected mucosal tissues while significant morphological changes were only found in external tissues including buccal mucosa, gills and skin. Using 16S rRNA gene sequence, we revealed that the abundance of Proteobacteria in mucosal tissues including buccal mucosa, gills and gut showed increased trend after viral infection, whereas the abundance of Fusobacteria significantly decreased in gut. In addition, the loss of dominant commensal microorganisms and increased colonization of opportunistic bacteria were discovered in the mucosal surfaces indicating that a secondary bacterial infection might occur in these mucosal tissues after viral infection. Overall, our results firstly point out the distribution of internal and external mucosal microbiota and analyze the changes of mucosal microbiota in common carp after SVCV infection, which may indicated that the potential role of mucosal microbiota in the antiviral process in early vertebrates.


Subject(s)
Fish Diseases/immunology , Fish Diseases/virology , Host-Pathogen Interactions/immunology , Immunity, Mucosal , Microbiota , Rhabdoviridae/immunology , Animals , Biomarkers , Computational Biology/methods , Dysbiosis , Fish Diseases/pathology , Gene Expression , Immunohistochemistry , Metagenome , Metagenomics/methods , Mucous Membrane/immunology , Mucous Membrane/microbiology
14.
Front Immunol ; 12: 581786, 2021.
Article in English | MEDLINE | ID: mdl-33717065

ABSTRACT

Cholesterol is essential for building and maintaining cell membranes and is critical for several steps in the replication cycle of viruses, especially for enveloped viruses. In mammalian cells virus infections lead to the accumulation of the oxysterol 25-hydroxycholesterol (25HC), an antiviral factor, which is produced from cholesterol by the cholesterol 25 hydroxylase (CH25H). Antiviral responses based on CH25H are not well studied in fish. Therefore, in the present study putative genes encoding for CH25H were identified and amplified in common carp and rainbow trout cells and an HPLC-MS method was applied for determination of oxysterol concentrations in these cells under virus infection. Our results give some evidence that the activation of CH25H could be a part of the antiviral response against a broad spectrum of viruses infecting fish, in both common carp and rainbow trout cells in vitro. Quantification of oxysterols showed that fibroblastic cells are capable of producing 25HC and its metabolite 7α,25diHC. The oxysterol 25HC showed an antiviral activity by blocking the entry of cyprinid herpesvirus 3 (CyHV-3) into KFC cells, but not spring viremia of carp virus (SVCV) or common carp paramyxovirus (Para) in the same cells, or viral haemorrhagic septicaemia virus (VHSV) and infectious pancreatic necrosis virus (IPNV) into RTG-2 cells. Despite the fact that the CH25H based antiviral response coincides with type I IFN responses, the stimulation of salmonid cells with recombinant type I IFN proteins from rainbow trout could not induce ch25h_b gene expression. This provided further evidence, that the CH25H-response is not type I IFN dependent. Interestingly, the susceptibility of CyHV-3 to 25HC is counteracted by a downregulation of the expression of the ch25h_b gene in carp fibroblasts during CyHV-3 infection. This shows a unique interplay between oxysterol based immune responses and immunomodulatory abilities of certain viruses.


Subject(s)
Antiviral Agents/immunology , Herpesviridae/immunology , Hydroxycholesterols/immunology , Rhabdoviridae/immunology , Animals , Antiviral Agents/metabolism , Carps/genetics , Carps/metabolism , Carps/virology , Cell Line , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Gene Expression Regulation/immunology , Herpesviridae/physiology , Host-Pathogen Interactions/immunology , Hydroxycholesterols/metabolism , Interferon Type I/genetics , Interferon Type I/immunology , Interferon Type I/metabolism , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/metabolism , Rhabdoviridae/physiology , Virus Internalization , Virus Replication/immunology
15.
Front Immunol ; 12: 647202, 2021.
Article in English | MEDLINE | ID: mdl-33659012

ABSTRACT

The innate immune organs and cells detect the invasion of pathogenic microorganisms, which trigger the innate immune response. A proper immune response can protect the organisms from pathogen invasion. However, excessive immunity can destroy immune homeostasis, leading to uncontrolled inflammation or pathogen transmission. Evidence shows that the miRNA-mediated immune regulatory network in mammals has had a significant impact, but the antibacterial and antiviral responses involved in miRNAs need to be further studied in lower vertebrates. Here, we report that miR-2187 as a negative regulator playing a critical role in the antiviral and antibacterial response of miiuy croaker. We find that pathogens such as Vibrio anguillarum and Siniperca chuatsi rhabdovirus (SCRV) can up-regulate the expression of miR-2187. Elevated miR-2187 is capable of reducing the production of inflammatory factors and antiviral genes by targeting TRAF6, thereby avoiding excessive inflammatory response. Furthermore, we proved that miR-2187 modulates innate immunity through TRAF6-mediated NF-κB and IRF3 signaling pathways. The above results indicate that miR-2187 acts as an immune inhibitor involved in host antibacterial and antiviral responses, thus enriching the immune regulatory network of the interaction between host and pathogen in lower vertebrates.


Subject(s)
Fish Proteins/genetics , Interferon Regulatory Factor-3/genetics , MicroRNAs/genetics , NF-kappa B/genetics , Perciformes/genetics , TNF Receptor-Associated Factor 6/genetics , Animals , Base Sequence , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Fish Diseases/genetics , Fish Diseases/microbiology , Fish Diseases/virology , Fish Proteins/metabolism , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon Regulatory Factor-3/metabolism , NF-kappa B/metabolism , Perciformes/microbiology , Perciformes/virology , Rhabdoviridae/immunology , Rhabdoviridae/physiology , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Signal Transduction/immunology , TNF Receptor-Associated Factor 6/metabolism , Vibrio/immunology , Vibrio/physiology
16.
PLoS Pathog ; 17(3): e1009438, 2021 03.
Article in English | MEDLINE | ID: mdl-33735323

ABSTRACT

Circular RNAs (circRNAs) represent a class of widespread and diverse covalently closed circular endogenous RNAs that exert crucial functions in regulating gene expression in mammals. However, the function and regulation mechanism of circRNAs in lower vertebrates are still unknown. Here, we discovered a novel circRNA derived from Deltex E3 ubiquitin ligase 1 (Dtx1) gene, namely, circDtx1, which was related to the antiviral responses in teleost fish. Results indicated that circDtx1 played essential roles in host antiviral immunity and inhibition of SCRV replication. Our study also found a microRNA miR-15a-5p, which could inhibit antiviral immune response and promote viral replication by targeting TRIF. Moreover, we also found that the antiviral effect inhibited by miR-15a-5p could be reversed with the circDtx1. In mechanism, our data revealed that circDtx1 was a competing endogenous RNA (ceRNA) of TRIF by sponging miR-15a-5p, leading to activation of the NF-κB/IRF3 pathway, and then enhancing the innate antiviral responses. Our results indicated that circRNAs played a regulatory role in immune responses in teleost fish.


Subject(s)
Adaptor Proteins, Vesicular Transport/biosynthesis , Fish Diseases/immunology , Gene Expression Regulation/genetics , Interferon Regulatory Factor-3/immunology , MicroRNAs/immunology , RNA, Circular/immunology , Animals , Down-Regulation , Immunity, Innate/immunology , Perciformes , Rhabdoviridae/immunology , Rhabdoviridae Infections/immunology
17.
Dev Comp Immunol ; 118: 103996, 2021 05.
Article in English | MEDLINE | ID: mdl-33444646

ABSTRACT

Interferon regulatory factors (IRFs) are crucial transcription factors involved in transcriptional regulation of type I interferons (IFNs) and IFN-stimulated genes (ISGs) against viral infection. In teleost fish, eleven IRFs have been found, however, understanding of their roles in the antiviral response remains limited. In the previous study, IRF1 (LcIRF1) and IRF2 (LcIRF2) genes were cloned from large yellow croaker (Larimichthys crocea). Here, we further characterized their function in the antiviral response. LcIRF1 and LcIRF2 were constitutively expressed in primary head kidney monocytes/macrophages (PKMs), lymphocytes (PKLs), granulocytes (PKGs) and large yellow croaker head kidney (LYCK) cell line, and significantly upregulated in PKMs and LYCK cells after stimulation with poly (I:C). LcIRF1 could induce promoter activities of three large yellow croaker type I IFNs, IFNc, IFNd and IFNh, while LcIRF2 could only induce those of IFNd and IFNh, and inhibit IFNc promoter activity. Correspondingly, overexpression of LcIRF1 in LYCK cells increased expression of all three IFNs (IFNc, IFNd and IFNh), while that of LcIRF2 only upregulated the expression levels of IFNd and IFNh, and inhibited expression of IFNc, although both LcIRF1and LcIRF2 induced expression of IFN-stimulated genes (ISGs), MxA, PKR and Viperin. Additionally, both LcIRF1 and LcIRF2 inhibited the Spring Viremia of Carp Virus (SVCV) replication in epithelioma papulosum cyprinid (EPC) cells, thus showing antiviral activity. Taken together, these results indicated that both LcIRF1 and LcIRF2 play positive roles in regulating the antiviral response of large yellow croaker by induction of distinct subgroups of type I IFNs.


Subject(s)
Fish Diseases/immunology , Fish Proteins/metabolism , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/metabolism , Interferon Type I/metabolism , Animals , Cell Line , Fish Diseases/virology , Immunity, Innate , Perciformes , Poly I-C/immunology , Rhabdoviridae/immunology , Up-Regulation/immunology , Virus Replication/immunology
18.
Cell Rep ; 34(1): 108600, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406425

ABSTRACT

Microgravity is a major environmental factor of space flight that triggers dysregulation of the immune system and increases clinical risks for deep-space-exploration crews. However, systematic studies and molecular mechanisms of the adverse effects of microgravity on the immune system in animal models are limited. Here, we establish a ground-based zebrafish disease model of microgravity for the research of space immunology. RNA sequencing analysis demonstrates that the retinoic-acid-inducible gene (RIG)-I-like receptor (RLR) and the Toll-like receptor (TLR) signaling pathways are significantly compromised by simulated microgravity (Sµg). TRIM25, an essential E3 for RLR signaling, is inhibited under Sµg, hampering the K63-linked ubiquitination of RIG-I and the following function-induction positive feedback loop of antiviral immune response. These mechanisms provide insights into better understanding of the effects and principles of microgravity on host antiviral immunity and present broad potential implications for developing strategies that can prevent and control viral diseases during space flight.


Subject(s)
DEAD Box Protein 58/immunology , Immunity , Rhabdoviridae/immunology , Toll-Like Receptors/immunology , Ubiquitin-Protein Ligases/immunology , Weightlessness/adverse effects , Zebrafish/immunology , Aerospace Medicine , Animals , DEAD Box Protein 58/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , HEK293 Cells , Humans , Male , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology , Signal Transduction , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/metabolism
19.
Sci China Life Sci ; 64(7): 1131-1148, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32997329

ABSTRACT

Viral infection induces the initiation of antiviral effectors and cytokines which are critical mediators of innate antiviral responses. The critical molecular determinants are responsible for triggering an appropriate immune response. Long noncoding RNAs (lncRNAs) have emerged as new gene modulators involved in various biological processes, while how lncRNAs operate in lower vertebrates are still unknown. Here, we discover a long noncoding RNA, termed antiviral-associated long noncoding RNA (AANCR), as a novel regulator for innate antiviral responses in teleost fish. The results indicate that fish MITA plays an essential role in host antiviral responses and inhibition of Siniperca chuatsi rhabdovirus (SCRV) production. miR-210 reduces MITA expression and suppress MITA-mediated antiviral responses, which may help viruses evade host antiviral responses. Further, AANCR functions as a competing endogenous RNA (ceRNA) for miR-210 to control protein abundance of MITA, thereby inhibiting SCRV replication and promoting antiviral responses. Our data not only shed new light on understanding the function role of lncRNA in biological processes in teleost fish, but confirmed the hypothesis that ceRNA networks exist widely in vertebrates.


Subject(s)
Antiviral Restriction Factors/genetics , Fish Diseases/immunology , Perciformes/genetics , Perciformes/immunology , RNA, Long Noncoding/genetics , Rhabdoviridae/immunology , Animals , Down-Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , MicroRNAs/genetics , Signal Transduction
20.
FASEB J ; 34(8): 10212-10227, 2020 08.
Article in English | MEDLINE | ID: mdl-32643209

ABSTRACT

Arginine methylation catalyzed by protein arginine methyltransferases (PRMT) is a common post-translational modification in histone and nonhistone proteins, which regulates many cellular functions. Protein arginine methyltransferase 3 (prmt3), a type I arginine methyltransferase, has been shown to carry out the formation of stable monomethylarginine as an intermediate before the establishment of asymmetric dimethylarginine. To date, however, the role of PRMT3 in antiviral innate immunity has not been elucidated. This study showed that zebrafish prmt3 was upregulated by virus infection and that the overexpression of prmt3 suppressed cellular antiviral response. The PRMT3 inhibitor, SGC707, enhanced antiviral capability. Consistently, prmt3-null zebrafish were more resistant to Spring Viremia of Carp Virus (SVCV) and Grass Carp Reovirus (GCRV) infection. Further assays showed that the overexpression of prmt3 diminished the phosphorylation of irf3 and prmt3 interacted with rig-i. In addition, both zinc-finger domain and catalytic domain of prmt3 were required for the suppressive function of prmt3 on IFN activation. Our findings suggested that zebrafish prmt3 negatively regulated the antiviral responses, implicating the vital role of prmt3-or even arginine methylation-in antiviral innate immunity.


Subject(s)
Antiviral Agents/immunology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/immunology , Zebrafish/genetics , Zebrafish/immunology , Animals , Cells, Cultured , Histones/genetics , Histones/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Isoquinolines/immunology , Methylation , Phosphorylation/genetics , Phosphorylation/immunology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology , Rhabdoviridae/immunology , Rhabdoviridae Infections/genetics , Rhabdoviridae Infections/immunology , Up-Regulation/genetics , Up-Regulation/immunology , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/virology , Zebrafish/virology , Zinc Fingers/genetics , Zinc Fingers/immunology
SELECTION OF CITATIONS
SEARCH DETAIL