Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 304
Filter
1.
J Gen Virol ; 105(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39172037

ABSTRACT

Rhabdoviral vectors can induce lysis of cancer cells. While studied almost exclusively at 37 °C, viruses are subject to a range of temperatures in vivo, including temperatures ≤31 °C. Despite potential implications, the effect of temperatures <37 °C on the performance of rhabdoviral vectors is unknown. We investigated the effect of low anatomical temperatures on two rhabdoviruses, vesicular stomatitis virus (VSV) and Maraba virus (MG1). Using a metabolic resazurin assay, VSV- and MG1-mediated oncolysis was characterized in a panel of cell lines at 28, 31, 34 and 37 °C. The oncolytic ability of both viruses was hindered at 31 and 28 °C. Cold adaptation of both viruses was attempted as a mitigation strategy. Viruses were serially passaged at decreasing temperatures in an attempt to induce mutations. Unfortunately, the cold-adaptation strategies failed to potentiate the oncolytic activity of the viruses at temperatures <37 °C. Interestingly, we discovered that viral replication was unaffected at low temperatures despite the abrogation of oncolytic activity. In contrast, the proliferation of cancer cells was reduced at low temperatures. Equivalent oncolytic effects could be achieved if cells at low temperatures were treated with viruses for longer times. This suggests that rhabdovirus-mediated oncolysis could be compromised at low temperatures in vivo where therapeutic windows are limited.


Subject(s)
Cold Temperature , Oncolytic Viruses , Rhabdoviridae , Virus Replication , Humans , Rhabdoviridae/physiology , Rhabdoviridae/genetics , Animals , Oncolytic Viruses/physiology , Oncolytic Viruses/genetics , Vesiculovirus/physiology , Vesiculovirus/genetics , Oncolytic Virotherapy/methods , Cell Line , Genetic Vectors/genetics , Cell Line, Tumor , Temperature
2.
PLoS Pathog ; 20(8): e1012328, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102417

ABSTRACT

Spring viremia of carp virus (SVCV) has a broad fish host spectrum and is responsible for a disease that generally affects juvenile fishes with a mortality rate of up to 90%. In the absence of treatments or vaccines against SVCV, the search for prophylactic or therapeutic solutions is thus relevant, particularly to identify solutions compatible with mass vaccination. In addition to being a threat to aquaculture and ecosystems, SVCV is a unique pathogen to study virus-host interactions in the zebrafish model. Establishing the first reverse genetics system for SVCV and the design of recombinant SVCV (rSVCV) expressing fluorescent or bioluminescent proteins adds a new dimension for the study of these interactions using innovative imaging techniques. The infection by bath immersion of zebrafish larvae with rSVCV expressing mCherry allows us to define the first SVCV replication sites and the host innate immune responses using different transgenic lines of zebrafish. The fins were found as the main initial sites of infection in both zebrafish and carp, its natural host. Hence, new insights into the physiopathology of SVCV infection have been described. We report that neutrophils are recruited at the sites of infection and persist up to the death of the animal leading to an uncontrolled inflammation correlated with the expression of the pro-inflammatory cytokine IL1ß. Tissue damage was observed at the site of initial replication, a likely consequence of virus-induced injury or the pro-inflammatory response. Interestingly, SVCV infection by bath immersion triggers a persistent pro-inflammatory response rather than activation of the antiviral IFN signaling pathway as observed following intravenous injection, highlighting the importance of the route of infection on the progression of pathogenicity. Thus, this model of zebrafish larvae infection by rSVCV offers new perspectives to study in detail virus-host interactions and to discover new prophylactic or therapeutic solutions.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Zebrafish , Animals , Zebrafish/virology , Rhabdoviridae/physiology , Fish Diseases/virology , Rhabdoviridae Infections/virology , Rhabdoviridae Infections/immunology , Carps/virology , Animals, Genetically Modified , Disease Models, Animal , Immunity, Innate , Viremia
3.
Microbiome ; 12(1): 128, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020382

ABSTRACT

BACKGROUND: Spring viremia of carp virus (SVCV) infects a wide range of fish species and causes high mortality rates in aquaculture. This viral infection is characterized by seasonal outbreaks that are temperature-dependent. However, the specific mechanism behind temperature-dependent SVCV infectivity and pathogenicity remains unclear. Given the high sensitivity of the composition of intestinal microbiota to temperature changes, it would be interesting to investigate if the intestinal microbiota of fish could play a role in modulating the infectivity of SVCV at different temperatures. RESULTS: Our study found that significantly higher infectivity and pathogenicity of SVCV infection in zebrafish occurred at relatively lower temperature. Comparative analysis of the intestinal microbiota in zebrafish exposed to high- and low-temperature conditions revealed that temperature influenced the abundance and diversity of the intestinal microbiota in zebrafish. A significantly higher abundance of Parabacteroides distasonis and its metabolite secondary bile acid (deoxycholic acid, DCA) was detected in the intestine of zebrafish exposed to high temperature. Both colonization of Parabacteroides distasonis and feeding of DCA to zebrafish at low temperature significantly reduced the mortality caused by SVCV. An in vitro assay demonstrated that DCA could inhibit the assembly and release of SVCV. Notably, DCA also showed an inhibitory effect on the infectious hematopoietic necrosis virus, another Rhabdoviridae member known to be more infectious at low temperature. CONCLUSIONS: This study provides evidence that temperature can be an important factor to influence the composition of intestinal microbiota in zebrafish, consequently impacting the infectivity and pathogenicity of SVCV. The findings highlight the enrichment of Parabacteroides distasonis and its derivative, DCA, in the intestines of zebrafish raised at high temperature, and they possess an important role in preventing the infection of SVCV and other Rhabdoviridae members in host fish. Video Abstract.


Subject(s)
Bacteroidetes , Fish Diseases , Gastrointestinal Microbiome , Rhabdoviridae Infections , Rhabdoviridae , Temperature , Zebrafish , Animals , Fish Diseases/microbiology , Fish Diseases/virology , Rhabdoviridae Infections/virology , Rhabdoviridae/physiology , Rhabdoviridae/pathogenicity , Bacteroidetes/pathogenicity , Water , Infectious hematopoietic necrosis virus/pathogenicity
4.
Fish Shellfish Immunol ; 152: 109726, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38944254

ABSTRACT

The immune system of bony fish closely resembles that of mammals, comprising both specific (adaptive) and non-specific (innate) components. Notably, the mucosa-associated lymphoid tissue (MALT) serves as the first line of defense within the non-specific immune system, playing a critical role in protecting these aquatic organisms against invading pathogens. MALT encompasses a network of immune cells strategically distributed throughout the gills and intestines, forming an integral part of the mucosal barrier that interfaces directly with the surrounding aquatic environment. Spring Viremia of Carp Virus(SVCV), a highly pathogenic agent causing substantial harm to common carp populations, has been designated as a Class 2 animal disease by the Ministry of Agriculture and Rural Affairs of China. Utilizing a comprehensive array of research techniques, including Hematoxylin and Eosin (HE)、Alcian Blue Periodic Acid-Schiff (AB-PAS)、transcriptome analysis for global gene expression profiling and Reverse Transcription-Polymerase Chain Reaction (RT-qPCR), this study uncovered several key findings: SVCV is capable of compromising the mucosal architecture in the gill and intestinal tissues of carp, and stimulate the proliferation of mucous cells both in gill and intestinal tissues. Critically, the study revealed that SVCV's invasion elicits a robust response from the carp's mucosal immune system, demonstrating the organism's capacity to resist SVCV invasion despite the challenges posed by the pathogen.


Subject(s)
Carps , Fish Diseases , Gene Expression Profiling , Gills , Intestines , Rhabdoviridae Infections , Rhabdoviridae , Animals , Gills/immunology , Gills/virology , Rhabdoviridae/physiology , Fish Diseases/immunology , Fish Diseases/virology , Carps/immunology , Carps/genetics , Gene Expression Profiling/veterinary , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology , Intestines/immunology , Intestines/virology , Immunity, Innate/genetics , Transcriptome/immunology , Immunity, Mucosal
5.
Fish Shellfish Immunol ; 151: 109719, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38914181

ABSTRACT

Sequestosome 1 (SQSTM1/p62) is a selective autophagy adapter protein that participates in antiviral and bacterial immune responses and plays an important regulatory role in clearing the proteins to be degraded and maintaining intracellular protein homeostasis. In this study, two p62 genes were cloned from common carp (Cyprinus carpio), namely Ccp62-1 and Ccp62-2, and conducted bioinformatics analysis on them. The results showed that Ccp62s had the same structural domain (Phox and Bem1 domain, ZZ-type zinc finger domain, and ubiquitin-associated domain) as p62 from other species. Ccp62s were widely expressed in various tissues of fish, and highly expressed in immune organs such as gills, spleen, head kidney, etc. Subcellular localization study showed that they were mainly distributed in punctate aggregates in the cytoplasm. After stimulation with Aeromonas hydrophila and spring viraemia of carp virus (SVCV), the expression level of Ccp62s was generally up-regulated. Overexpression of Ccp62s in EPC cells could inhibit SVCV replication. Upon A. hydrophila challenge, the bacterial load in Ccp62s-overexpressing group was significantly reduced, the expression levels of pro-inflammatory cytokines and interferon factors were increased, and the survival rate of the fish was improved. These results indicated that Ccp62s were involved in the immune response of common carp to bacterial and viral infections.


Subject(s)
Aeromonas hydrophila , Carps , Fish Diseases , Fish Proteins , Gram-Negative Bacterial Infections , Immunity, Innate , Phylogeny , Rhabdoviridae Infections , Rhabdoviridae , Animals , Carps/immunology , Carps/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Aeromonas hydrophila/physiology , Immunity, Innate/genetics , Rhabdoviridae/physiology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Gene Expression Regulation/immunology , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/immunology , Gene Expression Profiling/veterinary , Sequence Alignment/veterinary , Amino Acid Sequence , Autophagy/immunology
6.
J Virol ; 98(7): e0069724, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38916400

ABSTRACT

Micropterus salmoides rhabdovirus (MSRV) is an important pathogen of largemouth bass. Despite extensive research, the functional receptors of MSRV remained unknown. This study identified the host protein, laminin receptor (LamR), as a cellular receptor facilitating MSRV entry into host cells. Our results demonstrated that LamR directly interacts with MSRV G protein, playing a pivotal role in the attachment and internalization processes of MSRV. Knockdown of LamR with siRNA, blocking cells with LamR antibody, or incubating MSRV virions with soluble LamR protein significantly reduced MSRV entry. Notably, we found that LamR mediated MSRV entry via clathrin-mediated endocytosis. Additionally, our findings revealed that MSRV G and LamR were internalized into cells and co-localized in the early and late endosomes. These findings highlight the significance of LamR as a cellular receptor facilitating MSRV binding and entry into target cells through interaction with the MSRV G protein. IMPORTANCE: Despite the serious epidemic caused by Micropterus salmoides rhabdovirus (MSRV) in largemouth bass, the precise mechanism by which it invades host cells remains unclear. Here, we determined that laminin receptor (LamR) is a novel target of MSRV, that interacts with its G protein and is involved in viral attachment and internalization, transporting with MSRV together in early and late endosomes. This is the first report demonstrating that LamR is a cellular receptor in the MSRV life cycle, thus contributing new insights into host-pathogen interactions.


Subject(s)
Fish Diseases , Receptors, Laminin , Rhabdoviridae , Virus Internalization , Animals , Receptors, Laminin/metabolism , Rhabdoviridae/metabolism , Rhabdoviridae/physiology , Fish Diseases/virology , Fish Diseases/metabolism , Bass/virology , Bass/metabolism , Receptors, Virus/metabolism , Rhabdoviridae Infections/virology , Rhabdoviridae Infections/metabolism , Endocytosis
7.
Int J Biol Macromol ; 269(Pt 2): 132104, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719016

ABSTRACT

Stimulator of interferon genes (STING), as an imperative adaptor protein in innate immune, responds to nucleic acid from invading pathogens to build antiviral responses in host cells. Aberrant activation of STING may trigger tissue damage and autoimmune diseases. Given the decisive role in initiating innate immune response, the activity of STING is intricately governed by several posttranslational modifications, including phosphorylation and ubiquitination. Here, we cloned and characterized a novel RNF122 homolog from common carp (named CcRNF122L). Expression analysis disclosed that the expression of CcRNF122L is up-regulated under spring viremia of carp virus (SVCV) stimulation in vivo and in vitro. Overexpression of CcRNF122L hampers SVCV- or poly(I:C)-mediated the expression of IFN-1 and ISGs in a dose-dependent way. Mechanistically, CcRNF122L interacts with STING and promotes the polyubiquitylation of STING. This polyubiquitylation event inhibits the aggregation of STING and the subsequent recruitment of TBK1 and IRF3 to the signaling complex. Additionally, the deletion of the TM domain abolishes the negative regulatory function of CcRNF122L. Collectively, our discoveries unveil a mechanism that governs the STING function and the precise adjustment of the innate immune response in teleost.


Subject(s)
Carps , Fish Proteins , Immunity, Innate , Membrane Proteins , Rhabdoviridae , Animals , Carps/immunology , Carps/genetics , Carps/virology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Rhabdoviridae/physiology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Fish Diseases/immunology , Fish Diseases/virology , Rhabdoviridae Infections/immunology , Signal Transduction
8.
Fish Shellfish Immunol ; 149: 109559, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636737

ABSTRACT

USP14 regulates the immune related pathways by deubiquitinating the signaling molecules in mammals. In teleost, USP14 is also reported to inhibit the antiviral immune response through TBK1, but its regulatory mechanism remains obscure. To elucidate the role of USP14 in the RLR/IFN antiviral pathway in teleost, the homolog USP14 (bcUSP14) of black carp (Mylopharyngodon piceus) has been cloned and characterize in this paper. bcUSP14 contains 490 amino acids (aa), and the sequence is well conserved among in vertebrates. Over-expression of bcUSP14 in EPC cells attenuated SVCV-induced transcription activity of IFN promoters and enhanced SVCV replication. Knockdown of bcUSP14 in MPK cells led to the increased transcription of IFNs and decreased SVCV replication, suggesting the improved antiviral activity of the host cells. The interaction between bcUSP14 and bcTBK1 was identified by both co-immunoprecipitation and immunofluorescent staining. Co-expressed bcUSP14 obviously inhibited bcTBK1-induced IFN production and antiviral activity in EPC cells. K63-linked polyubiquitination of bcTBK1 was dampened by co-expressed bcUSP14, and bcTBK1-mediated phosphorylation and nuclear translocation of IRF3 were also inhibited by this deubiquitinase. Thus, all the data demonstrated that USP14 interacts with and inhibits TBK1 through deubiquitinating TBK1 in black carp.


Subject(s)
Carps , Fish Diseases , Fish Proteins , Immunity, Innate , Interferons , Protein Serine-Threonine Kinases , Rhabdoviridae Infections , Rhabdoviridae , Signal Transduction , Ubiquitination , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Carps/immunology , Carps/genetics , Fish Diseases/immunology , Rhabdoviridae/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/immunology , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Immunity, Innate/genetics , Ubiquitin Thiolesterase/genetics , Gene Expression Regulation/immunology , Amino Acid Sequence , Sequence Alignment/veterinary , Phylogeny , Gene Expression Profiling/veterinary
9.
Fish Shellfish Immunol ; 149: 109563, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642725

ABSTRACT

HnRNP A/B belongs to the heterogeneous nuclear ribonucleoprotein (hnRNP) family and plays an important role in regulating viral protein translation and genome replication. Here, we found that overexpression of hnRNP A/B promoted spring viremia of carp virus (SVCV) and cyprinid herpesvirus 3 (CyHV3) replication. Further, hnRNP A/B was shown to act as a negative regulator of type I interferon (IFN) response. Mechanistically, hnRNP A/B interacted with MITA, TBK1 and IRF3 to initiate their degradation. In addition, hnRNP A/B bound to the kinase domain of TBK1, the C terminal domain of MITA and IAD domain of IRF3, and the RRM1 domain of hnRNP A/B bound to TBK1, RRM2 domain bound to IRF3 and MITA. Our study provides novel insights into the functions of hnRNP A/B in regulating host antiviral response.


Subject(s)
Fish Diseases , Fish Proteins , Protein Serine-Threonine Kinases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Fish Diseases/immunology , Fish Diseases/virology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Rhabdoviridae/physiology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/immunology , Immunity, Innate/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/immunology , Carps/immunology , Carps/genetics , Herpesviridae/physiology , Herpesviridae Infections/veterinary , Herpesviridae Infections/immunology , Interferon Type I/immunology , Interferon Type I/genetics , Interferon Type I/metabolism , Zebrafish Proteins
10.
Fish Shellfish Immunol ; 149: 109577, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643957

ABSTRACT

A new virus known as snakehead rhabdovirus (SHRV-In) was discovered in South India in striped snakehead (Channa striata) that had hemorrhagic patches and cutaneous ulcerations. The virus is the most potentially harmful pathogen of snakehead because it could cause 100% mortality within 5 days. The goal of the current investigation was to evaluate the infectivity of rhabdovirus in freshwater fishes and to analyze the immune response in snakehead fish after challenge with SHRV-In. The infectivity study of SHRV-In against three freshwater fish such as tilapia, grass carp and loach showed that the virus could not induce mortality in any of them. Snakehead fish challenged with SHRV-In showed significant (p < 0.05) changes in haematological parameters such as red blood cell (RBC), haemoglobin (HGB), haematocrit (HCT), mean corpuscular haemoglobin concentration (MCHC), mean corpuscular volume (MCV), mean corpuscular haemoglobin (MCH), white blood cell (WBC), total platelet (PLT) counts, mean platelet volume (MPV) and immunological markers such as respiratory burst, superoxide dismutase, catalase activity and myeloperoxidase activity at 6, 12, 24 and 48 hpi. Real time PCR was executed to examine the expression profile of innate immune genes such as IRF-7, IL-8 and IL-12 in Snakehead fish at 6, 12, 24 and 48 h post SHRV-In infection. Immune gene expression of IRF-7, IL-8 and IL-12 were up-regulated in the spleen when compared to kidney at 6 and 12 hpi. However, the expression level of all the genes was down-regulated at 24 and 48 hpi. The down regulation of innate immune genes after 24 hpi in these tissues may be the result of increased multiplication of SHRV-In by interfering with the immune signaling pathway.


Subject(s)
Fish Diseases , Immunity, Innate , Rhabdoviridae Infections , Animals , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Fish Diseases/immunology , Fish Diseases/virology , Rhabdoviridae/physiology , India , Perciformes/immunology , Perciformes/virology
11.
Fish Shellfish Immunol ; 149: 109553, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615704

ABSTRACT

Viral diseases have caused great economic losses to the aquaculture industry. However, there are currently no specific drugs to treat these diseases. Herein, we utilized Siniperca chuatsi as an experimental model, and successfully extracted two tissue factor pathway inhibitors (TFPIs) that were highly distributed in different tissues. We then designed four novel peptides based on the TFPIs, named TS20, TS25, TS16, and TS30. Among them, TS25 and TS30 showed good biosafety and high antiviral activity. Further studies showed that TS25 and TS30 exerted their antiviral functions by preventing viruses from invading Chinese perch brain (CPB) cells and disrupting Siniperca chuatsi rhabdovirus (SCRV)/Siniperca chuatsi ranairidovirus (SCRIV) viral structures. Additionally, compared with the control group, TS25 and TS30 could significantly reduce the mortality of Siniperca chuatsi, the relative protection rates of TS25 against SCRV and SCRIV were 71.25 % and 53.85 % respectively, and the relative protection rate of TS30 against SCRIV was 69.23 %, indicating that they also had significant antiviral activity in vivo. This study provided an approach for designing peptides with biosafety and antiviral activity based on host proteins, which had potential applications in the prevention and treatment of viral diseases.


Subject(s)
Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Fish Diseases/virology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae/physiology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Perches , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Peptides/pharmacology , Peptides/chemistry , RNA Virus Infections/veterinary , RNA Virus Infections/immunology , RNA Virus Infections/prevention & control
12.
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
13.
Article in English | MEDLINE | ID: mdl-38621626

ABSTRACT

Hybrid snakehead (male Channa argus × female Channa maculata) is an emerging fish breed with increasing production levels. However, infection with hybrid snakehead rhabdovirus (HSHRV) critically affects hybrid snakehead farming. In this study, a fish cell line called CAMK, derived from the kidneys of hybrid snakehead, was established and characterized. CAMK cells exhibited the maximum growth rate at 28 °C in Leibovitz's-15 medium supplemented with 10% fetal bovine serum(FBS). Karyotyping revealed diploid chromosomes in 54% of the cells at the 50th passage (2n = 66), and 16S rRNA sequencing validated that CAMK cells originated fromhybrid snakehead, and the detection of kidney-specific antibodies suggested that it originated from kidney. .The culture was free from mycoplasma contamination, and the green fluorescent protein gene was effectively transfected into CAMK cells, indicating their potential use for in vitro gene expression investigations. Furthermore, qRT-PCR and immunofluorescence analysis revealed that HSHRV could replicate in CAMK cells, indicating that the cells were susceptible to the virus. Transmission electron microscopy revealed that the viral particles had bullet-like morphology. The replication efficiency of HSHRV was 107.33 TCID50/mL. Altogether, we successfully established and characterized a kidney cell line susceptible to the virus. These findings provide a valuable reference for further genetic and virological studies.


Subject(s)
Fishes , Kidney , Rhabdoviridae , Animals , Kidney/virology , Kidney/cytology , Cell Line , Female , Male , Fishes/virology , Rhabdoviridae/physiology , Fish Diseases/virology , Rhabdoviridae Infections/veterinary , Rhabdoviridae Infections/virology
14.
Insect Mol Biol ; 33(4): 295-311, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38551144

ABSTRACT

Exportin 1 (XPO1) is the major karyopherin-ß nuclear receptor mediating the nuclear export of hundreds of proteins and some classes of RNA and regulates several critical processes in the cell, including cell-cycle progression, transcription and translation. Viruses have co-opted XPO1 to promote nucleocytoplasmic transport of viral proteins and RNA. Maize mosaic virus (MMV) is a plant-infecting rhabdovirus transmitted in a circulative propagative manner by the corn planthopper, Peregrinus maidis. MMV replicates in the nucleus of plant and insect hosts, and it remains unknown whether MMV co-opts P. maidis XPO1 (PmXPO1) to complete its life cycle. Because XPO1 plays multiple regulatory roles in cell functions and virus infection, we hypothesized that RNAi-mediated silencing of XPO1 would negatively affect MMV accumulation and insect physiology. Although PmXPO1 expression was not modulated during MMV infection, PmXPO1 knockdown negatively affected MMV accumulation in P. maidis at 12 and 15 days after microinjection. Likewise, PmXPO1 knockdown negatively affected P. maidis survival and reproduction. PmXPO1 exhibited tissue-specific expression patterns with higher expression in the ovaries compared with the guts of adult females. Survival rate was significantly lower for PmXPO1 knockdown females, compared with controls, but no effect was observed for males. PmXPO1 knockdown experiments revealed a role for PmXPO1 in ovary function and egg production. Oviposition and egg hatch on plants were dramatically reduced in females treated with dsRNA PmXPO1. These results suggest that PmXPO1 is a positive regulator of P. maidis reproduction and that it plays a proviral role in the insect vector supporting MMV infection.


Subject(s)
Exportin 1 Protein , Hemiptera , Insect Vectors , Karyopherins , Ovary , RNA Interference , Receptors, Cytoplasmic and Nuclear , Animals , Female , Hemiptera/virology , Hemiptera/genetics , Hemiptera/growth & development , Karyopherins/metabolism , Karyopherins/genetics , Ovary/virology , Ovary/metabolism , Ovary/growth & development , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Insect Vectors/virology , Insect Vectors/genetics , Rhabdoviridae/physiology , Insect Proteins/metabolism , Insect Proteins/genetics , Zea mays/virology , Zea mays/genetics , Gene Knockdown Techniques
15.
Fish Shellfish Immunol ; 148: 109483, 2024 May.
Article in English | MEDLINE | ID: mdl-38458501

ABSTRACT

The precise control of interferon (IFN) production is indispensable for the host to eliminate invading viruses and maintain a homeostatic state. In mammals, stimulator of interferon genes (STING) is a prominent adaptor involved in antiviral immune signaling pathways. However, the regulatory mechanism of piscine STING has not been thoroughly investigated. Here, we report that autophagy related 16 like 1 (bcATG16L1) of black carp (Mylopharyngodon piceus) is a negative regulator in black carp STING (bcSTING)-mediated signaling pathway. Initially, we substantiated that knockdown of bcATG16L1 increased the transcription of IFN and ISGs and enhanced the antiviral activity of the host cells. Subsequently, we identified that bcATG16L1 inhibited the bcSTING-mediated IFN promoter activation and proved that bcATG16L1 suppressed bcSTING-mediated antiviral ability. Furthermore, we revealed that bcATG16L1 interacted with bcSTING and the two proteins shared a similar subcellular distribution. Mechanically, we found that bcATG16L1 attenuated the oligomerization of bcSTING, which was a key step for bcSTING activation. Taken together, our results indicate that bcATG16L1 interacts with bcSTING, dampens the oligomerization of bcSTING, and negatively regulates bcSTING-mediated antiviral activity.


Subject(s)
Carps , Fish Diseases , Reoviridae Infections , Reoviridae , Rhabdoviridae Infections , Rhabdoviridae , Animals , Rhabdoviridae/physiology , Reoviridae/physiology , Rhabdoviridae Infections/veterinary , Carps/genetics , Carps/metabolism , Fish Proteins , Immunity, Innate/genetics , Interferons , Mammals/metabolism
16.
Dev Comp Immunol ; 154: 105145, 2024 May.
Article in English | MEDLINE | ID: mdl-38316233

ABSTRACT

Spring viremia of carp virus (SVCV) is a globally distributed virus that causes severe clinical symptoms and high mortality in fish belonging to the families Cyprinidae and Siluridae. To protect the host against viral infection, understanding the relatedness between viral susceptibility and antiviral mechanisms must be crucial. Thus, we evaluated the viral suppression efficacy of ribavirin by measuring the transcription levels of viral and immune genes in vitro. The results showed that following ribavirin treatment after SVCV infection (MOI 0.1), ribavirin inhibited SVCV replication in epithelioma papulosum cyprini (EPC) cells and completely inhibited viral gene (G and N) expression at concentrations above 10 µg/mL at 48 h post-infection. Ribavirin does not directly damage SVCV particles but inhibits early viral replication. In the absence of SVCV infection, the immunological dynamics triggered by ribavirin resulted in upregulated pattern recognition receptors and proinflammatory cytokine-related genes (i.e., PI3K, MYD88, IRAK1, RIG-І, MAVS, Mx1, TNF-α, and NF-κB). Furthermore, EPC cells treated with ribavirin following SVCV infection showed upregulation of PI3K, MYD88, IRAK1, RIG-І, TNF-α, and NF-κB genes within 24 h post-SVCV infection, suggesting that ribavirin positively inhibits the SVCV infection in vitro.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Humans , Animals , Ribavirin/therapeutic use , Ribavirin/pharmacology , Viremia/drug therapy , NF-kappa B , Tumor Necrosis Factor-alpha , Myeloid Differentiation Factor 88/genetics , Rhabdoviridae/physiology , Adaptor Proteins, Signal Transducing , Phosphatidylinositol 3-Kinases
17.
Fish Shellfish Immunol ; 146: 109426, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38316349

ABSTRACT

Glutathione S-transferase P1 (GSTP1), the most ubiquitous member of the GST superfamily, plays vital roles in the detoxification, antioxidant defense, and modulation of inflammatory responses. However, limited studies have been conducted on the function of GSTP1 in antiviral innate immunity. In this study, we have cloned the homolog of GSTP1 in triploid hybrid crucian carp (3nGSTP1) and investigated its regulatory role in the interferon signaling pathway. The open reading frame of 3nGSTP1 is composed of 627 nucleotides, encoding 209 amino acids. In response to spring viremia of carp virus (SVCV) infection, the mRNA level of 3nGSTP1 was up-regulated in the liver, kidney, and caudal fin cell lines (3 nF C) of triploid fish. The knockdown of 3nGSTP1 in 3 nF C improved host cell's antiviral capacity and attenuated SVCV replication. Additionally, overexpression of 3nGSTP1 inhibited the activation of IFN promoters induced by SVCV infection, poly (I:C) stimulation, or the RLR signaling factors. The co-immunoprecipitation assays further revealed that 3nGSTP1 interacts with 3nMAVS. In addition, 3nGSTP1 dose-dependently inhibited 3nMAVS-mediated antiviral activity and reduced 3nMAVS protein level. Mechanistically, 3nGSTP1 promoted ubiquitin-proteasome degradation of MAVS by promoting its K48-linked polyubiquitination. To conclude, our results indicate that GSTP1 acts as a novel inhibitor of MAVS, which negatively regulates the IFN signaling.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Triploidy , Signal Transduction , Rhabdoviridae/physiology , Rhabdoviridae Infections/veterinary , Immunity, Innate/genetics , Poly I-C/pharmacology , Antiviral Agents
18.
Fish Shellfish Immunol ; 142: 109164, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37839543

ABSTRACT

Stimulator of interferon genes (STING) serve as an endoplasmic reticulum (ER) protein and modulates innate immune responses to viral contagion. Most investigations involving teleost STING antiviral immunity have examined DNA viruses. Therefore, fish STING signaling events against RNA viruses require additional exploration. Here, common carp STING (named CcSTING) was cloned and characterized. The bioinformatics analyses of CcSTING showed evolutionary conservations and were most closely related to other cyprinid STINGs. Immunofluorescence staining discovered that the CcSTING was chiefly placed in the cytoplasm, specifically within the ER. CcSTING was ubiquitously generated in all analyzed organs, with especially strong expression in the gills and head kidney. Spring viremia of carp virus (SVCV) stimulation and poly(I:C) infection induced the generation of CcSTING in immune-associated organs, as well as in peripheral blood leukocytes. Additional investigations revealed that CcSTING overexpression strongly suppressed SVCV replication in EPC cells. Mechanistically, CcSTING enhanced IFN-1 and ISGs expression following SVCV infection. CcSTING also substantially increased both IFN and NF-κB promoter luciferase activity via a dosage-dependent fashion. Lastly, CcSTING significantly up-regulated both TBK1 and p65 phosphorylation. Collectively, these findings demonstrated the critical role and underlying mechanism of fish STING in response to RNA virus.


Subject(s)
Carps , Fish Diseases , RNA Viruses , Rhabdoviridae Infections , Rhabdoviridae , Animals , Viremia , Carps/genetics , Carps/metabolism , Rhabdoviridae/physiology , Fish Proteins
19.
J Virol ; 97(11): e0122623, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37861337

ABSTRACT

IMPORTANCE: Although a virus can regulate many cellular responses to facilitate its replication by interacting with host proteins, the host can also restrict virus infection through these interactions. In the present study, we showed that the host eukaryotic translation elongation factor 1 alpha (eEF1A), an essential protein in the translation machinery, interacted with two proteins of a fish rhabdovirus, Siniperca chuatsi rhabdovirus (SCRV), and inhibited virus infection via two different mechanisms: (i) inhibiting the formation of crucial viral protein complexes required for virus transcription and replication and (ii) promoting the ubiquitin-proteasome degradation of viral protein. We also revealed the functional regions of eEF1A that are involved in the two processes. Such a host protein inhibiting a rhabdovirus infection in two ways is rarely reported. These findings provided new information for the interactions between host and fish rhabdovirus.


Subject(s)
Fish Diseases , Fish Proteins , Peptide Elongation Factor 1 , Rhabdoviridae Infections , Rhabdoviridae , Animals , Fishes , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Rhabdoviridae/physiology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/veterinary , Viral Proteins/genetics , Viral Proteins/metabolism , Fish Proteins/metabolism , Fish Diseases/metabolism
20.
Fish Shellfish Immunol ; 142: 109128, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37777100

ABSTRACT

In recent years, the exploration of natural compounds possessing both immunostimulatory and antiviral activities has attracted growing attention in aquaculture research. Consequently, the pursuit of identifying natural products exhibiting anti-SVCV potential as immunostimulants holds significant promise, offering a pathway to mitigate the economic ramifications inflicted by SVCV outbreaks in aquaculture settings. Among them, rhein emerges as a particularly compelling contender. Boasting a widespread distribution, well-established extraction methods, and multiple biological activities, it has exhibited the capacity to enhance the antiviral activity of host cells in vitro by blocking the viral internalization process, with a peak inhibition rate of 44.0%. Based on this intervention, rhein inhibited apoptosis and mitochondrial damage triggered by SVCV infection, ultimately producing a significant antiviral effect. Moving beyond the laboratory setting, rhein's efficacy translates effectively into in vivo scenarios. It has demonstrated substantial antiviral potency by increasing the expression of antiviral-related genes, most notably, retinoic acid-inducible gene I (RIG-I), interferon-φ (IFN-φ) and IFN-stimulated gene product 15 (ISG15). In concert with this genetic modulation, rhein efficiently reduces the viral load, precipitating a consequential enhancement in the survival rate of SVCV-infected fish, elevating it to an encouraging 16%. In conclusion, the outcomes of our investigation offer a compelling testament to rhein's potential as a valuable immunomodulator in the battle against SVCV infections in aquaculture, and the remarkable attributes exhibited by rhein underscore its viability for future commercial deployment.


Subject(s)
Carps , Fish Diseases , Rhabdoviridae Infections , Rhabdoviridae , Animals , Rhabdoviridae/physiology , Viremia/drug therapy , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL