Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Methods Mol Biol ; 2786: 289-300, 2024.
Article in English | MEDLINE | ID: mdl-38814400

ABSTRACT

In this protocol, we outline how to produce a chimeric viral vaccine in a biosafety level 1 (BSL1) environment. An animal viral vector RNA encapsidated with tobacco mosaic virus (TMV) coat protein can be fully assembled in planta. Agrobacterium cultures containing each component are inoculated together into tobacco leaves and the self-assembled hybrid chimeric viral vaccine is harvested 4 days later and purified with a simple PEG precipitation. The viral RNA delivery vector is derived from the BSL1 insect virus, Flock House virus (FHV), and replicates in human and animal cells but does not spread systemically. A polyethylene glycol purification protocol is also provided to collect and purify these vaccines for immunological tests. In this update, we also provide a protocol for in trans co-inoculation of a modified FHV protein A, which significantly increased the yield of in planta chimeric viral vaccine.


Subject(s)
Nicotiana , Replicon , Tobacco Mosaic Virus , Viral Vaccines , Nicotiana/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics , Animals , Tobacco Mosaic Virus/genetics , Tobacco Mosaic Virus/immunology , Replicon/genetics , RNA, Viral/genetics , Genetic Vectors/genetics , Nodaviridae/genetics , Nodaviridae/immunology , Plants, Genetically Modified/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Agrobacterium/genetics , Humans
2.
J Fish Dis ; 47(7): e13945, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38523313

ABSTRACT

The extensive growth of intensive fish farming has led to a massive spread of infectious diseases. Nervous necrosis virus (NNV) is the causative agent of the viral encephalo- and retinopathy disease which has become a major threat for fish farming all over the globe. The devastating mortality rates recorded in disease outbreaks, especially when infected specimens are at early stages of development, have a high economic impact on the sector. Currently, vaccines are the most cost-effective preventing tool in the fight against viruses. Inactivated vaccines have the advantage of simplicity in their development at the same time as present the antigen in a similar manner than the natural infection in the host. Nevertheless, they usually trigger weaker immune responses needing adjuvants to boost their effectiveness. In this work, we have intraperitoneally vaccinated Senegalese sole juveniles (Solea senegalensis) with a previously designed inactivated vaccine against NNV based on binary ethylenimine (BEI), mixed or not with an oil-adjuvant. Our results demonstrated the potential activation of different immune pathways when the vaccine was administered alone compared to the oil-adjuvanted vaccine, both resulting in an equivalent partial improvement in survival following a NNV challenge. However, whilst the vaccine alone led to a significant increase in specific antibodies, in the adjuvanted version those antibodies were kept basal although with a slight improvement in their neutralization capacity. At transcriptional level, neither vaccine (adjuvanted or not) triggered the immune system activation during the vaccination period. However, after NNV infection, the BEI-inactivated vaccines alone and oil-adjuvanted both elicited the stimulation of antiviral responsive genes (rtp3, herc4), antigen presentation molecules (mhcii) and T-cell markers (cd8a) in the head-kidney. Additionally, the oil-adjuvanted vaccine appears to stimulate mediator cytokines (il6) and B-cell markers (ight and ighm). Surprisingly, when the adjuvant was administered alone, fish showed the highest survival rates concomitantly with a lack of NNV-IgM production, pointing to the possible induction of different immune pathways than the B-cell responses via antibodies by the adjuvant. Since this combined vaccine did not succeed in the full extension of protection against the pathogen, further studies should be performed focusing on unravelling the molecular mechanisms through which adjuvants trigger the immune response, both independently and when added to a vaccine antigen.


Subject(s)
Fish Diseases , Flatfishes , Nodaviridae , RNA Virus Infections , Vaccines, Inactivated , Viral Vaccines , Animals , Fish Diseases/prevention & control , Fish Diseases/virology , Fish Diseases/immunology , Flatfishes/immunology , Flatfishes/virology , Nodaviridae/immunology , RNA Virus Infections/veterinary , RNA Virus Infections/prevention & control , RNA Virus Infections/immunology , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Vaccination/veterinary , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine/administration & dosage
3.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163406

ABSTRACT

Nanoplastics (NPs) might cause different negative effects on aquatic organisms at different biological levels, ranging from single cells to whole organisms, including cytotoxicity, reproduction, behavior or oxidative stress. However, the impact of NPs on disease resistance is almost unknown. The objective of this study was to assess whether exposure to 50 nm functionalized polystyrene NPs impacts fish susceptibility to viral diseases both in vitro and in vivo. In particular, we focused on the nervous necrosis virus (NNV), which affects many fish species, producing viral encephalopathy and retinopathy (VER), and causes great economic losses in marine aquaculture. In vitro and in vivo approaches were used. A brain cell line (SaB-1) was exposed to 1 µg mL-1 of functionalized polystyrene NPs (PS-NH2, PS-COOH) and then infected with NNV. Viral titers were increased in NP-exposed cells whilst the transcription of inflammatory and antiviral markers was lowered when compared to those cells only infected with NNV. In addition, European sea bass (Dicentrarchus labrax) juveniles were intraperitoneally injected with the same NPs and then challenged with NNV. Our results indicated that NPs increased the viral replication and clinical signs under which the fish died although the cumulate mortality was unaltered. Again, exposure to NPs produced a lowered inflammatory and antiviral response. Our results highlight that the presence of NPs might impact the infection process of NNV and fish resistance to the disease, posing an additional risk to marine organisms.


Subject(s)
Bass , Fish Diseases , Microplastics/toxicity , Nodaviridae/immunology , RNA Virus Infections , Animals , Bass/immunology , Bass/virology , Cell Line , Fish Diseases/immunology , Fish Diseases/virology , RNA Virus Infections/immunology , RNA Virus Infections/virology
4.
Microbiol Spectr ; 9(3): e0148721, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34817277

ABSTRACT

Viral diseases of the central nervous system (CNS) represent a major global health concern. Difficulties in treating these diseases are caused mainly by the biological tissues and barriers, which hinder the transport of drugs into the CNS. To counter this, a nanobody-mediated virus-targeting drug delivery platform (SWCNTs-P-A-Nb) is constructed for CNS viral disease therapy. Viral encephalopathy and retinopathy (VER), caused by nervous necrosis virus (NNV), is employed as a disease model. SWCNTs-P-A-Nb is successfully constructed by employing single-walled carbon nanotubes, amantadine, and NNV-specific nanobody (NNV-Nb) as the nanocarrier, anti-NNV drug, and targeting ligand, respectively. Results showed that SWCNTs-P-A-Nb has a good NNV-targeting ability in vitro and in vivo, improving the specific distribution of amantadine in NNV-infected sites under the guidance of NNV-Nb. SWCNTs-P-F-A-Nb can pass through the muscle and gill and be excreted by the kidney. SWCNTs-P-A-Nb can transport amantadine in a fast manner and prolong the action time, improving the anti-NNV activity of amantadine. Results so far have indicated that the nanobody-mediated NNV-targeting drug delivery platform is an effective method for VER therapy, providing new ideas and technologies for control of the CNS viral diseases. IMPORTANCE CNS viral diseases have resulted in many deadly epidemics throughout history and continue to pose one of the greatest threats to public health. Drug therapy remains challenging due to the complex structure and relative impermeability of the biological tissues and barriers. Therefore, development in the intelligent drug delivery platform is highly desired for CNS viral disease therapy. In the study, a nanobody-mediated virus-targeting drug delivery platform is constructed to explore the potential application of targeted therapy in CNS viral diseases. Our findings hold great promise for the application of targeted drug delivery in CNS viral disease therapy.


Subject(s)
Amantadine/pharmacology , Central Nervous System Viral Diseases/therapy , Central Nervous System Viral Diseases/veterinary , Drug Delivery Systems/methods , Nodaviridae/drug effects , Single-Domain Antibodies/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Central Nervous System/virology , Encephalitis, Viral/therapy , Encephalitis, Viral/virology , Fishes , Nanotubes, Carbon , Nodaviridae/immunology , Perciformes/virology , Single-Domain Antibodies/immunology
5.
Fish Shellfish Immunol ; 116: 91-97, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34224855

ABSTRACT

The objective of this study was to analyze the efficiency of the killed vaccine against nervous necrosis virus on Acipenser stellutus. Heat inactivated VNN vaccine was administrated in 7 g juveniles of Acipenser stellutus as a laboratory model and it was included in three different adjuvants that were used as injection and immersion forms with different doses. Ten groups consisting of 30 A. stellutus fish in each group (group 1-4 with 3 replications, others with no replicate) were divided totally into 18 aquariums. Two steps of vaccination were done with a one-month interval and after that, all treatments and control groups were challenged by the virulent VNN virus. The mortality rate of immersion and injection groups were 12.9% and 19.8% respectively, compared to 100% mortality in the control group. Histopathology and immunohistochemistry findings were evaluated. According to the mortality rate one month after challenging, a low range mortality of 12.5% was seen in group 2 with no pathological lesion and negative IHC test in the brain and eye tissues, whereas 100% of the control group (unvaccinated group) died with severe vacuolation in the brain and eye tissues and also positive IHC test. The correlation assay between these results concluded that the immersion form with 75% of aquatic-specific Montanide IMS 1312 Seppic adjuvant made better immunization with no pathological sign or forming the complex of antigen-antibody in IHC assay. These findings are important because of the impossibility of injection in the larval stage and also due to the occurrence of the disease in the first stage of sturgeon life which could cause high mortality in susceptible fish in the larval stage.


Subject(s)
Fish Diseases/prevention & control , Nodaviridae/immunology , RNA Virus Infections/prevention & control , Vaccines, Inactivated/administration & dosage , Viral Vaccines/administration & dosage , Animals , Antigen-Antibody Complex , Brain/immunology , Brain/pathology , Eye/immunology , Eye/pathology , Fish Diseases/immunology , Fish Diseases/pathology , Fishes/immunology , Immunohistochemistry , RNA Virus Infections/immunology , RNA Virus Infections/pathology , RNA Virus Infections/veterinary
6.
J Virol ; 95(17): e0236420, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34133901

ABSTRACT

Nervous necrosis virus (NNV) belongs to the Betanodavirus genus of the Nodaviridae family and is the main cause of viral nervous necrosis disease in marine fish larvae and juveniles worldwide. The NNV virion contains two positive-sense, single-stranded RNA genomes, which encode RNA-dependent RNA polymerase, coat protein, and B2 protein. Interestingly, NNV infection can shut off host translation in orange-spotted grouper (Epinephelus coioides) brain cells; however, the detailed mechanisms of this action remain unknown. In this study, we discovered that the host translation factor, polyadenylate binding protein (PABP), is a key target during NNV takeover of host translation machinery. Additionally, ectopic expression of NNV coat protein is sufficient to trigger nuclear translocalization and degradation of PABP, followed by translation shutoff. A direct interaction between NNV coat protein and PABP was demonstrated, and this binding requires the NNV coat protein N-terminal shell domain and PABP proline-rich linker region. Notably, we also showed that degradation of PABP during later stages of infection is mediated by the ubiquitin-proteasome pathway. Thus, our study reveals that the NNV coat protein hijacks host PABP, causing its relocalization to the nucleus and promoting its degradation to stimulate host translation shutoff. IMPORTANCE Globally, more than 200 species of aquacultured and wild marine fish are susceptible to NNV infection. Devastating outbreaks of this virus have been responsible for massive economic damage in the aquaculture industry, but the molecular mechanisms by which NNV affects its host remain largely unclear. In this study, we show that NNV hijacks translation in host brain cells, with the viral coat protein binding to host PABP to promote its nuclear translocalization and degradation. This previously unknown mechanism of NNV-induced host translation shutoff greatly enhances the understanding of NNV pathogenesis and provides useful insights and novel tools for development of NNV treatments, such as the use of orange-spotted grouper brain cells as an in vitro model system.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/metabolism , Fish Diseases/immunology , Nodaviridae/immunology , Poly(A)-Binding Proteins/metabolism , Protein Biosynthesis , RNA Virus Infections/veterinary , Animals , Bass , Capsid Proteins/genetics , Poly(A)-Binding Proteins/genetics , Protein Transport , RNA Virus Infections/immunology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
7.
Int J Mol Sci ; 22(4)2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33672018

ABSTRACT

Hepatitis B is a major global health challenge. In the absence of an effective treatment for the disease, hepatitis B vaccines provide protection against the viral infection. However, some individuals do not have positive immune responses after being vaccinated with the hepatitis B vaccines available in the market. Thus, it is important to develop a more protective vaccine. Previously, we showed that hepatitis B virus (HBV) 'a' determinant (aD) displayed on the prawn nodavirus capsid (Nc) and expressed in Spodoptera frugiperda (Sf9) cells (namely, Nc-aD-Sf9) self-assembled into virus-like particles (VLPs). Immunisation of BALB/c mice with the Nc-aD-Sf9 VLPs showed significant induction of humoral, cellular and memory B-cell immunity. In the present study, the biophysical properties of the Nc-aD-Sf9 VLPs were studied using dynamic light scattering (DLS) and circular dichroism (CD) spectroscopy. Enzyme-linked immunosorbent assay (ELISA) was used to determine the antigenicity of the Nc-aD-Sf9 VLPs, and multiplex ELISA was employed to quantify the cytokine response induced by the VLPs administered intramuscularly into BALB/c mice (n = 8). CD spectroscopy of Nc-aD-Sf9 VLPs showed that the secondary structure of the VLPs predominantly consisted of beta (ß)-sheets (44.8%), and they were thermally stable up to ~52 °C. ELISA revealed that the aD epitope of the VLPs was significantly antigenic to anti-HBV surface antigen (HBsAg) antibodies. In addition, multiplex ELISA of serum samples from the vaccinated mice showed a significant induction (p < 0.001) of IFN-γ, IL-4, IL-5, IL-6, IL-10, and IL-12p70. This cytokine profile is indicative of natural killer cell, macrophage, dendritic cell and cytotoxic T-lymphocyte activities, which suggests a prophylactic innate and adaptive cellular immune response mediated by Nc-aD-Sf9 VLPs. Interestingly, Nc-aD-Sf9 induced a more robust release of the aforementioned cytokines than that of Nc-aD VLPs produced in Escherichia coli and a commercially used hepatitis B vaccine. Overall, Nc-aD-Sf9 VLPs are thermally stable and significantly antigenic, demonstrating their potential as an HBV vaccine candidate.


Subject(s)
Capsid Proteins/immunology , Cytokines/metabolism , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Immunodominant Epitopes/immunology , Nodaviridae/immunology , Signal Transduction/immunology , Vaccination/methods , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hot Temperature , Mice , Mice, Inbred BALB C , Sf9 Cells , Spodoptera , Vaccines, Virus-Like Particle/administration & dosage
8.
Sci Rep ; 11(1): 3608, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574489

ABSTRACT

Nervous necrosis virus (NNV) is a pathogenic fish-virus belonging to the genus Betanodavirus (Nodaviridae). Surface protrusions on NNV particles play a crucial role in both antigenicity and infectivity. We exposed purified NNV particles to different physicochemical conditions to investigate the effects on antigenicity and infectivity, in order to reveal information regarding the conformational stability and spatial relationships of NNV neutralizing-antibody binding sites and cell receptor binding sites. Treatment with PBS at 37 °C, drastically reduced NNV antigenicity by 66-79% on day one, whereas its infectivity declined gradually from 107.6 to 105.8 TCID50/ml over 10 days. When NNV was treated with carbonate/bicarbonate buffers at different pHs, both antigenicity and infectivity of NNV declined due to higher pH. However, the rate of decline with respect to antigenicity was more moderate than for infectivity. NNV antigenicity declined 75-84% after treatment with 2.0 M urea, however, there was no reduction observed in infectivity. The antibodies used in antigenicity experiments have high NNV-neutralizing titers and recognize conformational epitopes on surface protrusions. The maintenance of NNV infectivity means that receptor binding sites are functionally preserved. Therefore, it seems highly likely that NNV neutralizing-antibody binding sites and receptor binding sites are independently located on surface protrusions.


Subject(s)
Antigens, Viral/immunology , Epitopes/immunology , Fish Diseases/immunology , Nodaviridae/immunology , Animals , Antigens, Viral/drug effects , Bicarbonates/pharmacology , Buffers , Carbonates/pharmacology , Epitopes/genetics , Fish Diseases/virology , Fishes/virology , Molecular Conformation , Nodaviridae/genetics , Nodaviridae/pathogenicity
9.
J Gen Virol ; 102(3)2021 03.
Article in English | MEDLINE | ID: mdl-33507144

ABSTRACT

The zebrafish (Danio rerio) possesses evolutionarily conserved innate and adaptive immunity as a mammal and has recently become a popular vertebrate model to exploit infection and immunity. Antiviral RNA interference (RNAi) has been illuminated in various model organisms, including Arabidopsis thaliana, Drosophila melanogaster, Caenorhabditis elegans and mice. However, to date, there is no report on the antiviral RNAi pathway of zebrafish. Here, we have evaluated the possible use of zebrafish to study antiviral RNAi with Sindbis virus (SINV), vesicular stomatitis virus (VSV) and Nodamura virus (NoV). We find that SINVs and NoVs induce the production of virus-derived small interfering RNAs (vsiRNAs), the hallmark of antiviral RNAi, with a preference for a length of 22 nucleotides, after infection of larval zebrafish. Meanwhile, the suppressor of RNAi (VSR) protein, NoV B2, may affect the accumulation of the NoV in zebrafish. Furthermore, taking advantage of the fact that zebrafish argonaute-2 (Ago2) protein is naturally deficient in cleavage compared with that of mammals, we provide evidence that the slicing activity of human Ago2 can virtually inhibit the accumulation of RNA virus after being ectopically expressed in larval zebrafish. Thus, zebrafish may be a unique model organism to study the antiviral RNAi pathway.


Subject(s)
RNA Interference , RNA Virus Infections/virology , RNA Viruses/physiology , RNA, Small Interfering/metabolism , RNA, Viral/metabolism , Zebrafish/virology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Immunity, Innate , Models, Animal , Nodaviridae/immunology , Nodaviridae/physiology , RNA Virus Infections/immunology , RNA Viruses/immunology , Sindbis Virus/immunology , Sindbis Virus/physiology , Vesiculovirus/immunology , Vesiculovirus/physiology , Zebrafish/genetics , Zebrafish/immunology , Zebrafish Proteins/metabolism
10.
Gene ; 774: 145430, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33444680

ABSTRACT

The transcriptomic response of Senegalese sole (Solea senegalensis) triggered by two betanodaviruses with different virulence to that fish species has been assessed using an OpenArray® platform based on TaqMan™ quantitative PCR. The transcription of 112 genes per sample has been evaluated at two sampling times in two organs (head kidney and eye/brain-pooled samples). Those genes were involved in several roles or pathways, such as viral recognition, regulation of type I (IFN-1)-dependent immune responses, JAK-STAT cascade, interferon stimulated genes, protein ubiquitination, virus responsive genes, complement system, inflammatory response, other immune system effectors, regulation of T-cell proliferation, and proteolysis and apoptosis. The highly virulent isolate, wSs160.3, a wild type reassortant containing a RGNNV-type RNA1 and a SJNNV-type RNA2 segments, induced the expression of a higher number of genes in both tested organs than the moderately virulent strain, a recombinant harbouring mutations in the protruding domain of the capsid protein. The number of differentially expressed genes was higher 2 days after the infection with the wild type isolate than at 3 days post-inoculation. The wild type isolate also elicited an exacerbated interferon 1 response, which, instead of protecting sole against the infection, increases the disease severity by the induction of apoptosis and inflammation-derived immunopathology, although inflammation seems to be modulated by the complement system. Furthermore, results derived from this study suggest a potential important role for some genes with high expression after infection with the highly virulent virus, such as rtp3, sacs and isg15. On the other hand, the infection with the mutant does not induce immune response, probably due to an altered recognition by the host, which is supported by a different viral recognition pathway, involving myd88 and tbkbp1.


Subject(s)
Fish Diseases/genetics , Fish Diseases/virology , Flatfishes/genetics , Flatfishes/virology , Immunogenetic Phenomena/genetics , Nodaviridae , Animals , Brain/metabolism , Eye/metabolism , Fish Diseases/immunology , Flatfishes/immunology , Gene Expression Profiling , Head Kidney/metabolism , Interferon Type I/metabolism , Nodaviridae/immunology , Nodaviridae/pathogenicity , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/veterinary , RNA-Seq , Virulence , Virus Replication
11.
Dev Comp Immunol ; 117: 103966, 2021 04.
Article in English | MEDLINE | ID: mdl-33338519

ABSTRACT

Some members of genus Macrobrachium are important economically prawns and valuable objects for studying the innate immune defense mechanism of crustaceans. Studies have focused on immune responses against bacterial and fungal infections and have expanded to include antiviral immunity over the past two decades. Similar to all living organisms, prawns are exposed to viruses, including white spot syndrome virus, Macrobrachium rosenbergii nodavirus, and Decapod iridescent virus 1 and develop effective defense mechanisms. Here, we review current understanding of the antiviral host defense in two species of Macrobrachium. The main antiviral defense of Macrobrachium is the activation of intracellular signaling cascades, leading to the activation of cellular responses (apoptosis) and humoral responses (immune-related signaling pathways, antimicrobial and antiviral peptides, lectins, and prophenoloxidase-activating system).


Subject(s)
Arthropod Proteins/immunology , Immunity, Innate/immunology , Iridoviridae/immunology , Nodaviridae/immunology , Palaemonidae/immunology , White spot syndrome virus 1/immunology , Animals , Apoptosis/immunology , Arthropod Proteins/metabolism , Host-Pathogen Interactions/immunology , Iridoviridae/physiology , Nodaviridae/physiology , Palaemonidae/metabolism , Palaemonidae/virology , Signal Transduction/immunology , White spot syndrome virus 1/physiology
12.
Virus Res ; 292: 198227, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33186642

ABSTRACT

The objective of this study was to investigate safety and efficacy using a low-temperature immunization protocol with NNV in sevenband grouper, Epinephelus septemfasciatus. Further, NNV specific antibody post immunization and intramuscularly challenge was also evaluated. Immunization at low temperature resulted in a low titer virus infection in brain tissues without any clinical symptoms of infection such as sluggish behavior and/or spinning, rotating swimming being observed, and no mortality was observed. Post challenge, NNV titer NNV giving an RPS of 100 %, increased in brain tissues of naïve (non-immunized) sevenband grouper NNV giving an RPS of 100 %, with a cumulative mortality of 100 % at 25 days post-infection. No mortality or disease symptoms NNV giving an RPS of 100 %, as NNV giving and of 100 %, observed in the groups immunized at low temperature with live NNV giving an RPS of 100 %. NNV giving an RPS of 100 %. NNV specific antibody was not detected in live NNV vaccinated sevenband grouper. This is the first study that confirms that field-scale NNV immersion vaccine can protect sevenband grouper against lethal infection with NNV at natural seawater temperature under the gradually increased from 14.3-24.8 °C.


Subject(s)
Fish Diseases/prevention & control , Nodaviridae/immunology , RNA Virus Infections/veterinary , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/immunology , Bass/immunology , Bass/virology , Cold Temperature , Fish Diseases/immunology , Fish Diseases/virology , Immunization/veterinary , Nodaviridae/genetics , RNA Virus Infections/immunology , RNA Virus Infections/prevention & control , RNA Virus Infections/virology , Viral Vaccines/immunology
13.
Sci Rep ; 10(1): 20051, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208823

ABSTRACT

Amphiprion ocellaris (ocellaris clownfish) is one of the most commercially important marine ornamental fish. A cell line designated as OCF was developed for the first time from the caudal fin of this fish species. The cell line was maintained in Leibovitz's-15 medium supplemented with 15% FBS (Fetal Bovine Serum) and was successfully subcultured up to 34 passages. The cell line was authenticated by sequencing mitochondrial cytochrome C oxidase subunit I (COI) and 16S rRNA genes. The growth rate of the OCF cell line was maximum in medium containing 20% FBS and 1% of 0.2 M NaCl at 28 °C. Chromosome analysis revealed 48 diploid chromosomes. The OCF cell line was transfected with the pMaxGFP plasmid vector with 7% efficiency and GFP expression was observed. The OCF cell line was used for testing nervous necrosis virus (NNV) susceptibility. Cytopathic effect (CPE) was observed in terms of plaque formation after virus inoculation. Nested PCR confirmed the susceptibility of the OCF cell line to NNV. The cell line was successfully cryopreserved by a slow freezing procedure at - 80 °C with a revival efficiency of 70-75%. The study revealed that the OCF cell line would be useful for virological studies. In addition, the cell line would play an important role as an in vitro tool for carrying out toxicological and biotechnological studies.


Subject(s)
Chromosomes/genetics , Fish Diseases/immunology , Nodaviridae/immunology , Perciformes/immunology , RNA Virus Infections/veterinary , Animals , Cell Culture Techniques , Cell Line , Fish Diseases/virology , Nodaviridae/genetics , Perciformes/genetics , RNA Virus Infections/immunology , RNA Virus Infections/virology , Virus Replication
14.
mBio ; 11(4)2020 08 04.
Article in English | MEDLINE | ID: mdl-32753500

ABSTRACT

Distinct mammalian RNA viruses trigger Dicer-mediated production of virus-derived small-interfering RNAs (vsiRNA) and encode unrelated proteins to suppress vsiRNA biogenesis. However, the mechanism and function of the mammalian RNA interference (RNAi) response are poorly understood. Here, we characterized antiviral RNAi in a mouse model of infection with Nodamura virus (NoV), a mosquito-transmissible positive-strand RNA virus encoding a known double-stranded RNA (dsRNA)-binding viral suppressor of RNAi (VSR), the B2 protein. We show that inhibition of NoV RNA replication by antiviral RNAi in mouse embryonic fibroblasts (MEFs) requires Dicer-dependent vsiRNA biogenesis and Argonaute-2 slicer activity. We found that VSR-B2 of NoV enhances viral RNA replication in wild-type but not RNAi-defective MEFs such as Argonaute-2 catalytic-dead MEFs and Dicer or Argonaute-2 knockout MEFs, indicating that VSR-B2 acts mainly by suppressing antiviral RNAi in the differentiated murine cells. Consistently, VSR-B2 expression in MEFs has no detectable effect on the induction of interferon-stimulated genes or the activation of global RNA cleavages by RNase L. Moreover, we demonstrate that NoV infection of adult mice induces production of abundant vsiRNA active to guide RNA slicing by Argonaute-2. Notably, VSR-B2 suppresses the biogenesis of both vsiRNA and the slicing-competent vsiRNA-Argonaute-2 complex without detectable inhibition of Argonaute-2 slicing guided by endogenous microRNA, which dramatically enhances viral load and promotes lethal NoV infection in adult mice either intact or defective in the signaling by type I, II, and III interferons. Together, our findings suggest that the mouse RNAi response confers essential protective antiviral immunity in both the presence and absence of the interferon response.IMPORTANCE Innate immune sensing of viral nucleic acids in mammals triggers potent antiviral responses regulated by interferons known to antagonize the induction of RNA interference (RNAi) by synthetic long double-stranded RNA (dsRNA). Here, we show that Nodamura virus (NoV) infection in adult mice activates processing of the viral dsRNA replicative intermediates into small interfering RNAs (siRNAs) active to guide RNA slicing by Argonaute-2. Genetic studies demonstrate that NoV RNA replication in mouse embryonic fibroblasts is inhibited by the RNAi pathway and enhanced by the B2 viral RNAi suppressor only in RNAi-competent cells. When B2 is rendered nonexpressing or nonfunctional, the resulting mutant viruses become nonpathogenic and are cleared in adult mice either intact or defective in the signaling by type I, II, and III interferons. Our findings suggest that mouse antiviral RNAi is active and necessary for the in vivo defense against viral infection in both the presence and absence of the interferon response.


Subject(s)
Nodaviridae/genetics , RNA Interference , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , Virus Replication , Animals , Argonaute Proteins/genetics , Cell Line , Cells, Cultured , DEAD-box RNA Helicases/genetics , Female , Fibroblasts/immunology , Fibroblasts/virology , Male , Mice , Mice, Inbred C57BL , Nodaviridae/immunology , RNA Virus Infections/virology , Ribonuclease III/genetics
15.
Front Immunol ; 11: 1718, 2020.
Article in English | MEDLINE | ID: mdl-32849607

ABSTRACT

Receptor interacting protein 1 (RIP1) is an essential sensor of cellular stress, which may respond to apoptosis or cell survival and participate in antiviral pathways. To investigate the roles of fish RIP1 in Singapore grouper iridovirus (SGIV) and red-spotted grouper nervous necrosis virus (RGNNV) infection, a RIP1 homolog from orange-spotted grouper (Epinephelus coioides) (EcRIP1) was cloned and characterized. EcRIP1 encoded a 679 amino acid protein that shares 83.28% identity with that of Perca flavescens and contained a homologous N-terminal kinase (S-TKc) domain, a RIP isotype interaction motif (RHIM), and a C-terminal domain (DD). EcRIP1 was predominantly detected in immune tissues, and its expression was induced by RGNNV or SGIV infection in vitro. Subcellular localization showed that EcRIP1 was distributed in the cytoplasm with point-like uniform and dot-like aggregation forms. Overexpression of EcRIP1 inhibited SGIV and RGNNV replication and positively regulated the expression levels of interferon (IFN) and IFN-stimulated genes and pro-inflammatory factors. EcRIP1 may interact with grouper tumor necrosis factor receptor type 1-associated DEATH domain protein (EcTRADD) to promote SGIV-induced apoptosis, and interact with grouper Toll/interleukin-1 receptor (TIR) domain containing adapter inducing interferon-ß (EcTRIF) and participate in Myeloid Differentiation Factor 88 (MyD88)-independent toll-like receptor (TLR) signaling. EcRIP1 may also interact with grouper tumor necrosis factor receptor-associated factors (TRAFs) as intracellular linker proteins and mediate the signaling of various downstream signaling pathways, including NF-κB and IFN. These results suggest that EcRIP1 may inhibit SGIV and RGNNV infection by regulating apoptosis and various signaling molecules. Our study offers new insights into the regulatory mechanism of RIP1-related signaling, and provides a novel perspective on fish diseases mediated by RIP1.


Subject(s)
Bass/virology , DNA Virus Infections/veterinary , Fish Diseases/virology , Fish Proteins/metabolism , Immunity, Innate , Iridovirus/pathogenicity , Nodaviridae/pathogenicity , RNA Virus Infections/veterinary , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bass/genetics , Bass/immunology , Bass/metabolism , Cells, Cultured , Cytokines/metabolism , DNA Virus Infections/immunology , DNA Virus Infections/metabolism , DNA Virus Infections/virology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Host-Pathogen Interactions , Iridovirus/immunology , Nodaviridae/immunology , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , RNA Virus Infections/virology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction
16.
Fish Shellfish Immunol ; 104: 431-438, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32580003

ABSTRACT

The giant freshwater prawn/giant river prawn, Macrobrachium rosenbergii is one of the high market value crustaceans cultured worldwide. The intensified aquaculture of the species has led to the outbreak of infectious diseases, prominently, the white tail disease (WTD). It is caused by the infection of Macrobrachium rosenbergii nodavirus (MrNV), which was classified in the family of Nodaviridae. To-date, there are no effective prophylactic and therapeutic agents available against MrNV infection. Vaccination is known to be the most effective prophylactic agent in disease prevention. However, vaccine development against virus infection in crustaceans is equivocal. The feasibility of vaccination in conferring immune protection in crustaceans against infectious diseases is disputable. The argument lies in the fact that crustaceans do not possess adaptive immunity, which is the main immune component that functions to establish immunological memory upon vaccination. Nevertheless, an increasing number of literatures has been documented, which concerns the development of vaccines against infectious diseases in crustaceans. The current review deliberates different approaches in vaccine development against MrNV, which were documented in the past years. It is noteworthy that the live-attenuated MrNV vaccine has not been experimented by far. Thus, the potential of live-attenuated MrNV vaccine in conferring long-term immune protection through the establishment of innate immune memory is currently being discussed.


Subject(s)
Nodaviridae/immunology , Palaemonidae/virology , Vaccination , Viral Vaccines/pharmacology , Animals , Aquaculture
17.
Fish Shellfish Immunol ; 104: 18-24, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32473363

ABSTRACT

Understanding the functions of genes related to disease resistance and identifying polymorphisms in these genes are essential in molecular breeding for disease resistance. Viral nervous necrosis (VNN) is one of the major diseases in the Asian seabass, Lates calcarifer. Our previous works on QTL mapping, GWAS and cell-line transcriptome analysis of the Asian seabass after NNV challenge revealed that the gene GAB3 might be a candidate gene for VNN resistance. In this study, we cloned and characterized GAB3, and identified SNPs in the gene of the Asian seabass. The cDNA of the gene was 2165 bp, containing an ORF of 1674 bp encoding 557 amino acids. The gene consisted of 10 exons and nine introns. It was ubiquitously expressed in normal fish. An analysis of the association between two SNPs in the second intron and NNV resistance in 1035 fish descended from 43 families revealed that the two SNPs were significantly associated with VNN resistance. After NNV infection, the expression of GAB3 was significantly increased in the brain, spleen, muscle and gut, and was suppressed in the liver. The GAB3 protein was localized in the nucleus. Overexpression of GAB3 with specific GAB3-pcDNA was positively correlated to increased viral RNA and titer in NNV-infected Asian seabass cells. Our study provides new evidence to support that GAB3 may be an important gene related to NNV resistance. In addition, the SNPs provide DNA markers for the selection of candidate genes resistance to NNV at the juvenile stage of Asian seabass.


Subject(s)
Bass/genetics , Bass/immunology , Fish Diseases/immunology , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Animals , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , GRB2 Adaptor Protein/chemistry , Gene Expression Profiling/veterinary , Nodaviridae/immunology , Phylogeny , RNA Virus Infections/immunology , RNA Virus Infections/veterinary
18.
Front Immunol ; 10: 2579, 2019.
Article in English | MEDLINE | ID: mdl-31736981

ABSTRACT

Granzymes (Gzm) are serine proteases, contained into the secretory granules of cytotoxic cells, responsible for the cell-mediated cytotoxicity (CMC) against tumor cells and intracellular pathogens such as virus and bacteria. In fish, they have received little attention to their existence, classification or functional characterization. Therefore, we aimed to identify and evaluate their functional and transcriptomic relevance in the innate CMC activity of two relevant teleost fish species, gilthead seabream and European sea bass. Afterwards, we wanted to focus on their regulation upon nodavirus (NNV) infection, a virus that causes great mortalities to sea bass specimens while seabream is resistant. In this study, we have identified genes encoding GzmA and GzmB in both seabream and sea bass, as well as GzmM in seabream, which showed good phylogenetic relation to their mammalian orthologs. In addition, we found enzymatic activity related to tryptase (GzmA and/or GzmK), aspartase (GzmB), metase (GzmM), or chymase (GzmH) in resting head-kidney leucocytes (HKLs), with the following order of activity: GzmA/K ~ GzmM >> GzmH >>> GzmB. In addition, during innate CMC assays consisting on HKLs exposed to either mock- or NNV-infected target cells, though all the granzyme transcripts were increased only the tryptase activity did. Thus, our data suggest a high functional activity of GzmA/K in the innate CMC and a marginal one for GzmB. Moreover, GzmB activity was detected into target cells during the CMC assays. However, the percentage of target cells with GzmB activity after the CMC assays was about 10-fold lower than the death target cells, demonstrating that GzmB is not the main inductor of cell death. Moreover, in in vivo infection with NNV, gzm transcription is differently regulated depending on the fish species, genes and tissues. However, the immunohistochemistry study revealed an increased number of GzmB stained cells and areas in the brain of seabream after NNV infection, which was mainly associated with the lesions detected. Further studies are needed to ascertain the molecular nature, biological function and implication of fish granzymes in the CMC activity, and in the antiviral defense in particular.


Subject(s)
Bass/immunology , Fish Proteins/immunology , Granzymes/immunology , Immunity, Innate , Sea Bream/immunology , Animals , Bass/genetics , Bass/virology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Granzymes/genetics , Nodaviridae/immunology , RNA Virus Infections/genetics , RNA Virus Infections/immunology , RNA Virus Infections/veterinary , Sea Bream/genetics , Sea Bream/virology
19.
Sci Rep ; 9(1): 8647, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31201359

ABSTRACT

Nervous necrosis virus (NNV) is a pathogenic fish virus belonging to family Nodaviridae. The objective of this study was to analyze stabilities of NNV surface protrusion and free coat protein (CP) conformational structures by analyzing changes of NNV infectivity and antigenicity after incubation at moderate-low temperatures. When cultured NNV suspension was incubated at 45 °C, its infectivity declined gradually but its antigenicity maintained. In contrast, both infectivity and antigenicity of purified NNV declined after incubation at 45 °C. After heat-treatment, surface protrusions of NNV particles disappeared completely, although viral particle structures maintained. Therefore, the reduction in NNV infectivity appeared to specifically occur as a result of heat-denaturation of virus surface protrusions. The loss of NNV infectivity in the presence of fetal bovine serum (FBS) was delayed compared to virus heated in the absence of FBS, demonstrating that FBS could function as a stabilizer for conformational structures of NNV surface protrusions. Moreover, the stabilizing function of FBS changed depending on salt concentration. Continued maintenance of antigenicity for heated cultured NNV suspension containing free-CPs may suggest that conformational structures corresponding to protrusion-domain of free-CP are more heat-stable than those of surface protrusions on NNV particles.


Subject(s)
Capsid Proteins/chemistry , Cold Temperature , Molecular Conformation , Nodaviridae/chemistry , Antigens, Viral/immunology , Nodaviridae/immunology , Nodaviridae/pathogenicity , Nodaviridae/ultrastructure , Suspensions , Virion/ultrastructure
20.
Front Immunol ; 10: 517, 2019.
Article in English | MEDLINE | ID: mdl-30941145

ABSTRACT

Autophagy is an important biological activity that maintains homeostasis in eukaryotic cells. However, little is known about the functions of fish autophagy-related genes (Atgs). In this study, we cloned and characterized Atg5, a key gene in the autophagy gene superfamily, from orange-spotted grouper (Epinephelus coioides) (EcAtg5). EcAtg5 encoded a 275-amino acid protein that shared 94 and 81% identity to seabass (Lates calcarifer) and humans (Homo sapiens), respectively. The transcription level of EcAtg5 was significantly increased in cells infected with red-spotted grouper nervous necrosis virus (RGNNV). In cells infected with Singapore grouper iridovirus (SGIV), EcAtg5 expression declined during the early stage of infection and increased in the late stage. Fluorescence microscopy revealed that EcAtg5 mainly localized with a dot-like pattern in the cytoplasm of grouper cells. Overexpression of EcAtg5 significantly increased the replication of RGNNV and SGIV at different levels of detection, as indicated by increased severity of the cytopathic effect, transcription levels of viral genes, and levels of viral proteins. Knockdown of EcAtg5 decreased the replication of RGNNV and SGIV. Further studies showed that overexpression EcAtg5 activated autophagy, decreased expression levels of interferon related cytokines or effectors and pro-inflammatory factors, and inhibited the activation of nuclear factor κB, IFN-sensitive response element, and IFNs. In addition, ectopic expression of EcAtg5 affected cell cycle progression by hindering the G1/S transition. Taken together, our results demonstrated that fish Atg5 exerted a crucial role in virus replication by promoting autophagy, down-regulating antiviral IFN responses, and affecting the cell cycle.


Subject(s)
Autophagy-Related Protein 5/immunology , Autophagy/immunology , Fish Diseases/immunology , Fish Proteins/immunology , Fishes/immunology , Iridovirus/immunology , Nodaviridae/immunology , Animals , Cell Cycle/immunology , Cell Line , Gene Expression Regulation/immunology , Immunity, Innate/immunology , Inflammation/genetics , RNA Virus Infections/immunology , Ranavirus/immunology , Transcription, Genetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL