Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Fish Dis ; 44(7): 923-937, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33591590

ABSTRACT

Pancreas disease (PD) is a serious challenge in European salmonid aquaculture caused by salmonid alphavirus (SAV). In this study, we report the effect of immunization of Atlantic salmon with three attenuated infectious SAV3 strains with targeted mutations in a glycosylation site of the envelope E2 protein and/or in a nuclear localization signal in the capsid protein. In a pilot experiment, it was shown that the mutated viral strains replicated in fish, transmitted to naïve cohabitants and that the transmission had not altered the sequences. In the main experiment, the fish were immunized with the strains and challenged with SAV3 eight weeks after immunization. Immunization resulted in infection both in injected fish and 2 weeks later in the cohabitant fish, followed by a persistent but declining load of the mutated virus variants in the hearts. The immunized fish developed clinical signs and pathology consistent with PD prior to challenge. However, fish injected with the virus mutated in both E2 and capsid showed little clinical signs and had higher average weight gain than the groups immunized with the single mutated variants. The SAV strain used for challenge was not detected in the immunized fish indicating that these fish were protected against superinfection with SAV during the 12 weeks of the experiment.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus/classification , Fish Diseases/prevention & control , Pancreatic Diseases/veterinary , Viral Vaccines/immunology , Alphavirus/genetics , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Fish Diseases/virology , Immunization/veterinary , Pancreatic Diseases/prevention & control , Salmo salar , Vaccines, Attenuated
2.
Vet Res ; 47(1): 78, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27496170

ABSTRACT

Salmon pancreas disease virus, often referred to as salmonid alphavirus (SAV), causes pancreas disease (PD) in European salmonids. SAV transmits horizontally from fish shedding virus into the water and ocean currents are believed to be a main contributor of viral spread between marine farms. Vaccination against PD is previously shown to reduce mortality and severity of clinical PD. In this study, we demonstrate that vaccination against PD significantly reduces viral shedding from infected individuals. The results suggest that PD vaccination can be an important tool to reduce the infection pressure, a known key risk for PD outbreaks at neighbouring farms.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus/immunology , Fish Diseases/prevention & control , Pancreatic Diseases/veterinary , Salmo salar/virology , Viral Vaccines/therapeutic use , Alphavirus Infections/immunology , Alphavirus Infections/prevention & control , Animals , Fish Diseases/immunology , Fish Diseases/virology , Pancreatic Diseases/immunology , Pancreatic Diseases/prevention & control , Pancreatic Diseases/virology , Salmo salar/immunology , Virus Shedding/immunology
3.
Vaccine ; 32(47): 6206-12, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25269093

ABSTRACT

Salmonid alphavirus (SAV; also known as Salmon pancreas disease virus; family Togaviridae) causes pancreas disease and sleeping disease in Atlantic salmon and rainbow trout, respectively, and poses a major burden to the aquaculture industry. SAV infection in vivo is temperature-restricted and progeny virus is only produced at low temperatures (10-15 °C). Using engineered SAV replicons we show that viral RNA replication is not temperature-restricted suggesting that the viral structural proteins determine low-temperature dependency. The processing/trafficking of SAV glycoproteins E1 and E2 as a function of temperature was investigated via baculovirus vectors in Sf9 insect cells and by transfection of CHSE-214 fish cells with DNA constructs expressing E1 and E2. We identified SAV E2 as the temperature determinant by demonstrating that membrane trafficking and surface expression of E2 occurs only at low temperature and only in the presence of E1. Finally, a vaccination-challenge model in Atlantic salmon demonstrates the biological significance of our findings and shows that SAV replicon DNA vaccines encoding E2 elicit protective immunity only when E1 is co-expressed. This is the first study that identifies E2 as the critical determinant of SAV low-temperature dependent virion formation and defines the prerequisites for induction of a potent immune response in Atlantic salmon by DNA vaccination.


Subject(s)
Alphavirus Infections/veterinary , Cold Temperature , Fish Diseases/prevention & control , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Alphavirus/genetics , Alphavirus Infections/prevention & control , Animals , Cells, Cultured , Fish Diseases/virology , Glycoproteins/immunology , RNA, Viral/genetics , Salmo salar , Sf9 Cells , Virion/immunology
4.
Vaccines (Basel) ; 2(2): 228-51, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-26344619

ABSTRACT

Most commercial vaccines offered to the aquaculture industry include inactivated antigens (Ag) formulated in oil adjuvants. Safety concerns are related to the use of oil adjuvants in multivalent vaccines for fish, since adverse side effects (e.g., adhesions) can appear. Therefore, there is a request for vaccine formulations for which protection will be maintained or improved, while the risk of side effects is reduced. Here, by using an inactivated salmonid alphavirus (SAV) as the test Ag, the combined use of two Toll-like receptor (TLR) ligand adjuvants, CpG oligonucleotides (ODNs) and poly I:C, as well as a genetic adjuvant consisting of a DNA plasmid vector expressing the viral haemorrhagic septicaemia virus (VHSV) glycoprotein (G) was explored. VHSV-G DNA vaccine was intramuscularly injected in combination with intraperitoneal injection of either SAV Ag alone or combined with the oil adjuvant, Montanide ISA763, or the CpG/polyI:C combo. Adjuvant formulations were evaluated for their ability to boost immune responses and induce protection against SAV in Atlantic salmon, following cohabitation challenge. It was observed that CpG/polyI:C-based formulations generated the highest neutralizing antibody titres (nAbs) before challenge, which endured post challenge. nAb responses for VHSV G-DNA- and oil-adjuvanted formulations were marginal compared to the CpG/poly I:C treatment. Interestingly, heat-inactivated sera showed reduced nAb titres compared to their non-heated counterparts, which suggests a role of complement-mediated neutralization against SAV. Consistently elevated levels of innate antiviral immune genes in the CpG/polyI:C injected groups suggested a role of IFN-mediated responses. Co-delivery of the VHSV-G DNA construct with either CpG/polyI:C or oil-adjuvanted SAV vaccine generated higher CD4 responses in head kidney at 48 h compared to injection of this vector or SAV Ag alone. The results demonstrate that a combination of pattern recognizing receptor (PRR) ligands, such as CpG/polyI:C, increases both adaptive and innate responses and represents a promising adjuvant strategy for enhancing the protection of future viral vaccines.

5.
Vaccine ; 30(32): 4828-34, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22634299

ABSTRACT

CpG oligonucleotides and polyinosinic:polycytidylic acid (poly I:C) are toll-like receptor (TLR) agonists that mimic the immunostimulatory properties of bacterial DNA and double-stranded viral RNA respectively, and which have exhibited potential to serve as vaccine adjuvants in previous experiments. Here, a combination of CpGs and poly I:C together with water- or oil-formulated Salmonid Alphavirus (SAV) antigen preparations has been used for a vaccine in Atlantic salmon and tested for protection in SAV challenge trial. The results demonstrate that vaccination with a high dose of the SAV antigen induced protection against challenge with SAV which correlated with production of neutralizing antibodies (NAbs). As the high antigen dose alone induced full protection, no beneficial effect from the addition of CpG and poly I:C could be observed. Nevertheless, these TLR ligands significantly enhanced the levels of NAbs in serum of vaccinated fish. Interestingly, gene expression analysis demonstrated that while addition of oil suppressed the CpG/poly I:C-induced expression of IFN-γ, the upregulation of IFNa1 was substantially enhanced. A low dose of the SAV antigen combined with oil did not induce any detectable levels of NAbs either with or without TLR ligands present, however the addition of CpG and poly I:C to the low SAV antigen dose formulation significantly enhanced the protection against SAV suggesting that CpG/poly I:C may have enhanced a cytotoxic response - a process which is dependent on the up-regulation of type I IFN. These results highlight the immunostimulatory properties of the tested TLR ligands and will serve as a ground for further, more detailed studies aimed to investigate their capacity to serve as adjuvants in vaccine formulations for Atlantic salmon.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alphavirus Infections/veterinary , Fish Diseases/prevention & control , Salmon/immunology , Viral Vaccines/immunology , Alphavirus/immunology , Alphavirus Infections/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Fish Diseases/virology , Interferon Type I/immunology , Oligodeoxyribonucleotides/pharmacology , Poly I-C/pharmacology , Salmon/virology , Toll-Like Receptors/agonists , Virus Inactivation
6.
PLoS One ; 6(10): e25816, 2011.
Article in English | MEDLINE | ID: mdl-21991361

ABSTRACT

Pancreas disease (PD) and sleeping disease (SD) are important viral scourges in aquaculture of Atlantic salmon and rainbow trout. The etiological agent of PD and SD is salmonid alphavirus (SAV), an unusual member of the Togaviridae (genus Alphavirus). SAV replicates at lower temperatures in fish. Outbreaks of SAV are associated with large economic losses of ~17 to 50 million $/year. Current control strategies rely on vaccination with inactivated virus formulations that are cumbersome to obtain and have intrinsic safety risks. In this research we were able to obtain non-infectious virus-like particles (VLPs) of SAV via expression of recombinant baculoviruses encoding SAV capsid protein and two major immunodominant viral glycoproteins, E1 and E2 in Spodoptera frugiperda Sf9 insect cells. However, this was only achieved when a temperature shift from 27°C to lower temperatures was applied. At 27°C, precursor E2 (PE2) was misfolded and not processed by host furin into mature E2. Hence, E2 was detected neither on the surface of infected cells nor as VLPs in the culture fluid. However, when temperatures during protein expression were lowered, PE2 was processed into mature E2 in a temperature-dependent manner and VLPs were abundantly produced. So, temperature shift-down during synthesis is a prerequisite for correct SAV glycoprotein processing and recombinant VLP production.


Subject(s)
Alphavirus/metabolism , Cold Temperature , Glycoproteins/metabolism , Protein Processing, Post-Translational , Salmonidae/virology , Viral Proteins/metabolism , Virion/metabolism , Alphavirus/ultrastructure , Animals , Baculoviridae/genetics , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Protein Transport , Recombination, Genetic/genetics , Spodoptera/cytology , Virion/ultrastructure
7.
Dev Comp Immunol ; 35(11): 1116-27, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21527278

ABSTRACT

Both CpG oligodeoxynucleotides and double-stranded RNA (poly I:C) have documented effects as treatments against several viral diseases in fish. However, as stand-alone treatments their effects have been modest. We have tested here whether CpG and poly I:C, alone or in combination induce protection against Salmonid Alphavirus (SAV), the causative agent of pancreas disease in Atlantic salmon. Our results revealed a significant reduction of viraemia 2 weeks after ip injection of the combined treatment and 1 week after challenge with SAV subtype 3, followed by reduced SAV induced heart pathology 3 weeks later. The SAV titers in blood samples from the combination group were lower as compared to single treatments with either CpG or poly I:C. Surprisingly, reduced SAV levels could also be found in fish as long as 7 weeks after receiving the combination treatment. The expression of IFNγ and to a lesser extent IFNa and Mx was up-regulated in head kidney and spleen 5 days after the fish had been treated with CpG and poly I:C. Furthermore, the complement factor C4 was depleted in serum 8 weeks post treatment, suggesting complement activation leading to C4 consumption. We hypothesize that the CpG/poly I:C-induced protection against SAV3 is mediated by mechanisms involving type I and type II IFN induced antiviral activity and complement mediated protective responses.


Subject(s)
Alphavirus Infections/veterinary , Antiviral Agents/administration & dosage , Fish Diseases/immunology , Oligodeoxyribonucleotides/administration & dosage , Pancreatic Diseases/veterinary , Poly I-C/administration & dosage , Salmo salar/immunology , Alphavirus/immunology , Alphavirus/pathogenicity , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Antiviral Agents/immunology , Complement C4/analysis , Dinucleoside Phosphates , Fish Diseases/virology , Head Kidney/drug effects , Head Kidney/immunology , Immunity, Innate , Interferon Type I/biosynthesis , Interferon-gamma/biosynthesis , Oligodeoxyribonucleotides/immunology , Pancreatic Diseases/immunology , Pancreatic Diseases/virology , Poly I-C/immunology , Salmo salar/virology , Spleen/drug effects , Spleen/immunology
8.
BMC Res Notes ; 3: 241, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20858233

ABSTRACT

BACKGROUND: Salmonid alphavirus (SAV) is a widespread pathogen in European aquaculture of salmonid fish. Distinct viral subtypes have been suggested based on sequence comparisons and some of these have different geographical distributions. In Norway, only SAV subtype 3 have so far been identified. Little is known about viral mechanisms important for pathogenesis and transmission. Tools for detailed exploration of SAV genomes are therefore needed. RESULTS: Infectious cDNA clones in which a genome of subtype 3 SAV is under the control of a CMV promoter were constructed. The clones were designed to express proteins that are putatively identical to those previously reported for the SAVH20/03 strain. A polyclonal antiserum was raised against a part of the E2 glycoprotein in order to detect expression of the subgenomic open reading frame (ORF) encoding structural viral proteins. Transfection of the cDNA clone revealed the expression of the E2 protein by IFAT, and in serial passages of the supernatant the presence of infectious recombinant virus was confirmed through RT-PCR, IFAT and the development of a cytopathic effect similar to that seen during infection with wild type SAV. Confirmation that the recovered virus originated from the infectious plasmid was done by sequence identification of an introduced genetic tag. The recombinant virus was infectious also when an additional ORF encoding an EGFP reporter gene under the control of a second subgenomic alphavirus promoter was added. Finally, we used the system to study the effect of selected point mutations on infectivity in Chinook salmon embryo cells. While introduced mutations in nsP2197, nsP3263 and nsP3323 severely reduced infectivity, a serine to proline mutation in E2206 appeared to enhance the virus titer production. CONCLUSION: We have constructed infectious clones for SAV based on a subtype 3 genome. The clones may serve as a platform for further functional studies.

9.
Arch Virol ; 155(8): 1281-93, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20556445

ABSTRACT

Salmonid alphavirus (SAV) is the most divergent member of the family Togaviridae and constitutes a threat to farming of salmonid fish in Europe. Here, we report cloning, expression and preliminary functional analysis of the capsid protein of SAV, confirming it to be expressed as an approximately 31-kDa protein in infected cells. The protein localizes strictly to the cytoplasm in Chinook salmon embryo cells, and either to the nucleus or cytoplasm in bluegill fry cells. An expression study of full-length and different truncated versions of the SAV capsid fused to the enhanced green fluorescent protein demonstrated that the localization is independent of other viral components in both cell lines, and controlled by the N-terminal 82 aa, which include a conserved, predicted helix and a downstream positively charged region. Thus, the results suggest that the SAV capsid possesses a cell-type-dependent potential for nuclear import and export. Moreover, the SAV capsid and its N-terminal 82 aa were shown to be associated with inhibition of cellular proliferation, a hallmark of the cytopathic effect caused by SAV. These results highlight that the SAV capsid is a multifunctional protein with possible importance for pathogenesis.


Subject(s)
Alphavirus/pathogenicity , Amino Acid Sequence , Capsid Proteins/chemistry , Cell Proliferation/drug effects , Salmonidae/virology , Subcellular Fractions/metabolism , Alphavirus/metabolism , Alphavirus/physiology , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Cloning, Molecular , Cytoplasm/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Host-Pathogen Interactions , Phylogeny , Sequence Alignment , Sequence Analysis, DNA
10.
Virol J ; 6: 173, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19860871

ABSTRACT

Salmonid alphavirus (SAV) causes disease in farmed salmonid fish and is divided into different genetic subtypes (SAV1-6). Here we report the cloning and characterization of the 5'- and 3'- untranslated regions (UTR) of a SAV3 isolated from Atlantic salmon in Norway. The sequences of the UTRs are very similar to those of SAV1 and SAV2, but single nucleotide polymorphisms are present, also in the 3' - conserved sequence element (3'-CSE). Prediction of the RNA secondary structure suggested putative stem-loop structures in both the 5'- and 3'-ends, similar to those of alphaviruses from the terrestrial environment, indicating that the general genome replication initiation strategy for alphaviruses is also utilized by SAV. A DNA replicon vector, pmSAV3, based upon a pVAX1 backbone and the SAV3 genome was constructed, and the SAV3 non-structural proteins were used to express a reporter gene controlled by the SAV3 subgenomic promoter. Transfection of pmSAV3 into CHSE and BF2 cell lines resulted in expression of the reporter protein, confirming that the cloned SAV3 replication apparatus and UTRs are functional in fish cells.


Subject(s)
Alphavirus/genetics , Genetic Vectors , Genome, Viral , Replicon , Salmo salar/virology , Untranslated Regions , Animals , Base Sequence , Models, Molecular , Molecular Sequence Data , Norway , Nucleic Acid Conformation , Polymorphism, Genetic , RNA, Viral/genetics , Sequence Analysis, DNA
11.
J Virol ; 79(19): 12544-53, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160182

ABSTRACT

Infectious salmon anemia virus (ISAV) is an orthomyxovirus causing serious disease in Atlantic salmon (Salmo salar L.). This study presents the characterization of the ISAV 50-kDa glycoprotein encoded by segment 5, here termed the viral membrane fusion protein (F). This is the first description of a separate orthomyxovirus F protein, and to our knowledge, the first pH-dependent separate viral F protein described. The ISAV F protein is synthesized as a precursor protein, F0, that is proteolytically cleaved to F1 and F2, which are held together by disulfide bridges. The cleaved protein is in a metastable, fusion-activated state that can be triggered by low pH, high temperature, or a high concentration of urea. Cell-cell fusion can be initiated by treatment with trypsin and low pH of ISAV-infected cells and of transfected cells expressing F, although the coexpression of ISAV HE significantly improves fusion. Fusion is initiated at pH 5.4 to 5.6, and the fusion process is coincident with the trimerization of the F protein, or most likely a stabilization of the trimer, suggesting that it represents the formation of the fusogenic structure. Exposure to trypsin and a low pH prior to infection inactivated the virus, demonstrating the nonreversibility of this conformational change. Sequence analyses identified a potential coiled coil and a fusion peptide. Size estimates of F1 and F2 and the localization of the putative fusion peptide and theoretical trypsin cleavage sites suggest that the proteolytic cleavage site is after residue K276 in the protein sequence.


Subject(s)
Isavirus , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Fusion , Cell Line , Hemadsorption , Hot Temperature , Hydrogen-Ion Concentration , Molecular Sequence Data , Protein Processing, Post-Translational , Salmo salar , Sequence Analysis, DNA , Sequence Homology , Transfection , Trypsin/metabolism , Urea , Viral Fusion Proteins/genetics
12.
J Virol ; 76(12): 6155-63, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12021349

ABSTRACT

Cell culture isolates of salmon pancreas disease virus (SPDV) of farmed Atlantic salmon and sleeping disease virus (SDV) of rainbow trout were compared. Excluding the poly(A) tracts, the genomic nucleotide sequences of SPDV and SDV RNAs include 11,919 and 11,900 nucleotides, respectively. Phylogenetic analysis places SPDV and SDV between the New World viruses of Venezuelan equine encephalitis virus and Eastern equine encephalitis virus and the Old World viruses of Aura virus and Sindbis virus. When compared to each other, SPDV and SDV show 91.1% nucleotide sequence identity over their complete genomes, with 95 and 93.6% amino acid identities over their nonstructural and structural proteins, respectively. Notable differences between the two viruses include a 24-nucleotide insertion in the C terminus of nsP3 protein of SPDV and amino acid sequence variation at the C termini of the capsid and E1 proteins. Experimental infections of Atlantic salmon and rainbow trout with SPDV and SDV confirmed that the disease lesions induced by SPDV and SDV were similar in nature. Although infections with SPDV and SDV produced similar levels of histopathology in rainbow trout, SDV induced significantly less severe lesions in salmon than did SPDV. Virus neutralization tests performed with sera from experimentally infected salmon indicated that SPDV and SDV belonged to the same serotype; however, antigenic variation was detected among SDV and geographically different SPDV isolates by using monoclonal antibodies. Although SPDV and SDV exhibit minor biological differences, we conclude on the basis of the close genetic similarity that SPDV and SDV are closely related isolates of the same virus species for which the name Salmonid alphavirus is proposed.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus , Fish Diseases/virology , Oncorhynchus mykiss/virology , Salmo salar/virology , Alphavirus/genetics , Alphavirus/immunology , Alphavirus/pathogenicity , Alphavirus Infections/physiopathology , Alphavirus Infections/virology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Cross Reactions , Genome, Viral , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pancreatic Diseases/veterinary , Pancreatic Diseases/virology , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...