NO344698B1 - Novel fish virus - Google Patents

Novel fish virus Download PDF

Info

Publication number
NO344698B1
NO344698B1 NO20180836A NO20180836A NO344698B1 NO 344698 B1 NO344698 B1 NO 344698B1 NO 20180836 A NO20180836 A NO 20180836A NO 20180836 A NO20180836 A NO 20180836A NO 344698 B1 NO344698 B1 NO 344698B1
Authority
NO
Norway
Prior art keywords
seq
sequences
sequence
virus
nucleic acid
Prior art date
Application number
NO20180836A
Other languages
Norwegian (no)
Other versions
NO20180836A1 (en
Inventor
Håvard Aanes
Linda Ramsevik Teigene
Magnus Andreas Devold
Vidar Teis Aspehaug
Kristoffer Alnes Røyset
Teresa Garzon
David Cockerill
Meritxell Diez Padrisa
Original Assignee
Patogen As
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Patogen As filed Critical Patogen As
Priority to NO20180836A priority Critical patent/NO344698B1/en
Priority to PCT/NO2019/050124 priority patent/WO2019240596A1/en
Priority to EP19737262.6A priority patent/EP3807403A1/en
Priority to CA3103433A priority patent/CA3103433A1/en
Publication of NO20180836A1 publication Critical patent/NO20180836A1/en
Publication of NO344698B1 publication Critical patent/NO344698B1/en
Priority to CL2020003238A priority patent/CL2020003238A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23KFODDER
    • A23K50/00Feeding-stuffs specially adapted for particular animals
    • A23K50/80Feeding-stuffs specially adapted for particular animals for aquatic animals, e.g. fish, crustaceans or molluscs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Analytical Chemistry (AREA)
  • Polymers & Plastics (AREA)
  • Animal Husbandry (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Birds (AREA)
  • Insects & Arthropods (AREA)
  • Food Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Description

Novel Fish Virus
The present invention relates to use of a novel virus for vaccines and prophylactic treatment for a disease caused by the virus. The invention also relates to nucleic acid sequences, amino acid sequences, primers and primer pairs, probes and methods for diagnostic according to the preamble of the independent patent claims
Fish are an increasingly important source of food and income; global annual consumption is projected to rise from 110 million tons in 2010 to more than 200 million tons in 2030. However, the emergence of infectious diseases in aquaculture threatens production and may also impact wild fish populations.
During summer 2017 it was discovered particularly high mortality in several cages in a fish farm in Scotland. A novel fish virus was characterized from samples of the fish.
The novel virus was isolated in a salmon farm with high losses caused by disease. The virus shows unique characters and 28% similarity to unclassified Fisavirus1, that has previously been identified in Carp (Cyprinus carpio) (Reuter 2016). The sequence of Fisavirus is 8712 NT long and relates to Posavirus (Porcine stool associated virus), and it is assumed that the host of Fisavirus could be the Nematoda phylum (Reuter 2016). Nematodes (Nematoda) belong to the most frequent and the most important parasites of fishes in the freshwater, brackish-water and marine environments throughout the world (Moravec 2007). Small numbers of nematodes often occur in healthy fish, but high numbers cause illness or even death (Roy P. Yanong). Thus it is expected that the novel virus may have a broad range of hosts among fish and shellfish, such as shrimp, carp, tilapia, seabass and salmon, among others.
There is therefore a need for a rapid method to identify the virus, and a tool to monitor the production of virus and avoid outbreak. The present invention as described below, identifies the novel virus from fish, and describes methods for identification, detection and vaccine development. Another object is to develop further treatment to treat infected fish.
The virus may also be used in challenge methods using a virus according to the invention for testing susceptibility of animals, with the purpose of testing for instance efficacy of a vaccine or functional feed, or resistance towards disease. Further, the virus may be used in breeding programs to obtain animals that are resistant to the virus as defined above.
The new virus is isolated from Atlantic salmon.
The invention relates to an isolated nucleic acid sequence originating from the virus. The sequence is selected from the group consisting of SEQ ID No 2 and 3 and sequences complementary to any of SEQ ID No 2 and 3, and variants thereof being at least 70% identical.
The isolated nucleic acid sequence may further be at least 80%, preferably 90%, more preferably 95 % identical with any of the sequences SEQ ID No 2 or 3, or any sequences being complementary to SEQ ID No 2 or 3.
In another aspect of the invention, it relates to a nucleic acid sequence of at least 10 nucleotides, wherein said sequence hybridizes specifically to a nucleic acid sequence having a sequence selected from the group consisting of. SEQ ID No 4-12, sequences being complementary to SEQ ID No 4-12, and variants being at least 70 % identical with any of the sequences SEQ ID NO 4-12 and sequences being complementary to the variants of SEQ ID NO 4-12. Such a nucleic acid may be used as a primer for instance for PCR or a probe for instance for identification of a nucleic acid sequence or gene. Further, the said nucleotide sequence may be at least 15 or at least 20 nucleotides long.
In another aspect the invention provides a primer or probe comprising a sequence selected from the group consisting of SEQ ID NO 4-12, sequences being complementary to SEQ ID NO 4-12, and variants being at least 90 % identical with any of the sequences SEQ ID NO 4-12 and sequences being complementary to the variants of SEQ ID NO 4-12. In some aspects the primer may be 15 nucleotides or 20 nucleotides long.
The above said nucleic acid sequence being at least 10 nucleotides long, or the primer or probe, may further be at least 95 %, even more preferred 99% identical with the sequence SEQ ID No 2-12, or any sequences being complementary to SEQ ID No 2-12.
According to another aspect of the invention, a method for detection of a virus in a biological sample is provided. The method comprises the following steps:
a) preparing a sample comprising nucleic acid sequences isolated from the biological sample for a reverse transcription reaction,
b) subjecting the mixture of a) to a polymerase chain reaction with a primer pair, wherein each primer of said primer pair comprises at least 10 nucleotides and hybridizes to a nucleic acid sequence selected from the group consisting of SEQ ID No 4-12, sequences being complementary to SEQ ID No 4-12, and variants being at least 90 % identical with any of the sequences,
c) determining whether the binding of the nucleotide sequences to nucleotide sequences in the biological sample and amplification of the sequence between them have occurred indicating the presence of virus in the sample tested.
In a preferred embodiment, the primers of the method above are selected from a group consisting for SEQ ID No 4-12, preferably SEQ ID NO 7-12.
In step b) of the method above, the primers may further hybridize to a nucleic acid being at least 95 % or 100 % identical with the sequences SEQ ID No 4-12, or any sequences being complementary to SEQ ID No 4-12.
According to another aspect of the invention, another method for detection of a virus in a biological sample is provided. The method comprises the following steps: a) preparing a sample comprising nucleic acid sequences isolated from the biological sample for a reverse transcription reaction,
b) sequencing the mixture of a), and
c) comparing the resulting sequence with a sequence selected of the group consisting of SEQ ID No 2 and 3 and sequences being complementary to SEQ ID NO 2 and 3, wherein 70 % identity indicates the presence of virus in the biological sample tested.
Further, in step c) of the method above, 80 %, 90%, 95% or 100 % identity may be required to confirm the presence of virus in the biological sample.
In a preferred embodiment, the sequencing of the mixture in the method above is performed by a method selected from the group consisting of Next Generation Sequencing, preferably Illumina (Solexa) sequencing, Roche 454 sequencing, Ion Torrent or SOLiD sequencing (Goodwin S, et al., (2016) Coming of age: Ten years of next-generation sequencing technologies. Nature reviews, Genetics, 17, 333-351).
The invention also relates to use of a nucleic acid sequence comprising at the least 10 contiguous nucleotides of any of the sequences 2-12, or 10 contiguous nucleotides being complementary of any of the sequences 2-12, for confirming the presence of the virus in a biological sample. In yet another embodiment it is used a nucleic acid sequence having a sequence selected from the group of SEQ ID No 2 and 3 and sequences being complementary to the any of the sequences SEQ ID No 2 and 3.
The methods and uses above may be used to confirm that the virus is present in the biological samples, but it may also be used to confirm that the virus is not present in the biological samples. Further, it may be used to monitor a fish population, for instance for disease/sickness control. Information in this regard, desirable with methods for identifying other diseases and/or sicknesses, may be valuable information as to if or when the fish population should be treated. The biological samples to be analysed are preferably from dead fish, but may also be samples removed from live fish without harming the fish.
Another aspect of the invention relates to a vector comprising nucleic acid sequences according to the present invention, and host cells comprising said vectors.
According to yet another aspect of the invention, DNA vaccines are provided comprising a nucleic acid sequence selected from the group consisting of SEQ ID No. 2 and 3, sequences being complementary to sequences of SEQ ID No 2 and 3, and variants thereof being at least 70% identical with any of the sequences SEQ ID No. 2 and 3 or sequences being complementary to variants of SEQ ID No 2 and 3.
The nucleic acid sequence of the DNA vaccine may further be at least 80%, preferably 90%, more preferably 95 % identical with any of the sequences SEQ ID No 2 and 3, or any sequences being complementary to SEQ ID No 2 and 3.
Another aspect of the invention relates to recombinant proteins encoded by a nucleic acid sequence selected from the group consisting of SEQ ID No.2 and 3, and variants thereof being at least 70 %, 80 %, preferably 90%, more preferably 95 % identical with any of the sequences SEQ ID No.2 and 3. In one aspect the amino acid sequence of the recombinant protein is given in SEQ ID No 1.
The present invention also provides a recombinant vaccine comprising at least one of the recombinant proteins according to the present invention.
The immunogenic composition may further comprise at least one excipient, additive or adjuvant, and may be administrated orally, by immersion or by injection.
Furthermore, according to another aspect of the invention, it is provided an antibody that recognises and binds to a recombinant protein according to the present invention.
The invention also relates to a diagnostic kit, which may be used to decide whether a sample comes from an organism being infected by the virus according to the invention.
Finally, the present invention relates to the use of the nucleic acid sequences having a sequences selected from the group consisting of SEQ ID NO 2 and 3, sequences being complementary to SEQ ID NO 2 and 3, and variants thereof being at least 70 %, 80 %, preferably 90%, more preferably 95 % identical with any of these sequences, for the preparation of DNA vaccine, recombinant vaccine or a live recombinant microorganism.
The present invention relates to isolated nucleic acid sequences and variants thereof being at least 70% identical with the isolated nucleic acid sequences. The term "% identity" is to be understood to refer to the percentage of nucleotides that two or more sequences or fragments thereof contains that are the same. The term "at least 70 % identical" thus means that at least 70 % of the nucleotides over the entire sequences which are compared, are identical. A specified percentage of nucleotides can be referred to as e.g. 70% identical, 80% identical, 85% identical, 90% identical, 95% identical, 99% identical or more over a specified region when compared and aligned for maximum correspondence. The skilled person will acknowledge that various means for comparing sequences are available.
The term "variants thereof" used in respect of the nucleic acid sequences and recombinant proteins according to the present invention, is to be understood to encompass nucleic acid sequences and recombinant proteins that only differs from the isolated sequences SEQ ID No.1, 2, and 3 by way of some amino acid or nucleotide additions, deletions or alteration that have little effect, if any, on the functional activity of the claimed sequences. The skilled person will acknowledge that modifications of a protein coding nucleotide sequence may be introduced which does not alter the amino acid sequence, e.g. the substitution of a nucleotide resulting in that the triplett affected by the substitution still codes for the same amino acid.
Such alterations may be introduced to adapt the nucleic acid sequence to the codons preferably used by a host cell and thus to enhance the expression of a desired recombinant protein. Furthermore, the addition of nucleic acid sequences coding polypeptides which facilitates purification may be added without affecting the activity of the resulting recombinant protein.
The skilled person will further acknowledge that also alterations of the nucleic acid sequence resulting in modifications of the amino acid sequence of the recombinant protein it codes may have little, if any, effect on e.g. the proteins' ability to induce protection against the virus if the alteration does not have any impact on the resulting three dimensional structure of the recombinant protein. For example, a codon for the amino acid alanine, a hydrophobic amino acid, may be substituted by a codon encoding another less hydrophobic residue, such as glycine, or a more hydrophobic residue, such as valine, leucine, or isoleucine. Similarly, changes which result in substitution of one negatively charged residue for another, such as aspartic acid for glutamic acid, or one positively charged residue for another, such as lysine for arginine, can also be expected to produce a recombinant protein with substantially the same functional activity as the protein encoded by the nucleotide sequence SEQ ID No.2 or 3 and thus to be expected to constitute a biologically equivalent product. Nucleotide changes which result in alteration of the N-terminal or C-terminal portions of the protein molecule would also not be expected to alter the functional activity of the protein. Each of the proposed modifications is well within the routine skills in the art, as is determination of retention of biological activity of the encoded products. Therefore, where the terms "nucleic acid sequences of the invention" or "recombinant protein of the invention" are used in either the specification or the claims each will be understood to encompass all such modifications and variations which result in the production of a biologically equivalent protein.
Thus, the present invention encompasses recombinant proteins and variants thereof which differ in respect of amino acid substitutions, addition or deletions compared with the protein of SEQ ID No 1, and proteins being encoded by the sequence SEQ ID No.2 or 3.
The primers and probes according to the present invention, will hybridize under stringent conditions with the sequence in question. The term “hybridizing under stringent conditions” refers to conditions of high stringency, i.e. in term of temperature, ionic strength, and the presence of other compounds such as organic solvents, under which nucleic acid hybridizations are conducted. With “high stringency” conditions, nucleic acid base pairing will occur only between nucleic acids having a high frequency of complementary base sequences. Stringent hybridization conditions are known to the skilled person (see e.g. Green M. R., Sambrook, J., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press; 4th edition, 2012).
The precise conditions for stringent hybridization are typically sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium. Typically, stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C. for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60°C. for longer probes, primers and oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
The term "antigen" when used in connection with the present invention is to be understood to refer to a recombinant protein or fragment thereof according to the invention being able to induce protection against the virus in fish or to be able to bind to an antibody which recognise and bind to the virus.
The term "vaccine" as used herein refers to a material that can produce an immune response that blocks the infectivity, either partially or fully, of an infectious agent, which in respect of the present invention is the virus affecting fish such as e.g. salmonids. Thus, when administering the vaccines of the invention to a fish, the fish becomes immunised against the disease caused by the virus. The immunising component of the vaccine may be e.g. DNA as in a DNA vaccine, RNA as in a RNA vaccine, a recombinant protein or fragment thereof according to the present invention, or a live recombinant microorganism. The vaccines may be administered by immersion, orally or by injection.
The present invention also provides short nucleotide sequences having a length of at least 10 nucleotides. These sequences may be primers or probes useful in polymerase chain reaction techniques to be used as diagnostic tools. The terms "primer" and "probes" as used herein refers to an oligonucleotide either naturally occurring or produced synthetically which is significantly complementary to a virus target sequence and thus capable of hybridizing to nucleic acid sequences of the present invention. When a primer pair is used, it is generally one forward and one reverse primer, and the sequence between the primers will be multiplied during a PCR. This is well known to a skilled person, and he/she would know which primers constitute a suitable pair. The amplified sequence may be labelled to facilitate detection, e.g. using fluorescent labels on a probe, or other label means well known to the skilled person.
Figures
The invention will now be described in detail with reference to the enclosed Figures, where
Figure 1 shows the cumulated mortality of infected fish during 2017,
Figure 2 shows the amino acid sequence encoded by SEQ ID No.3, the amino acid sequence is corresponding to SEQ ID No 1,
Figure 3 shows the genomic sequence of the novel virus (PV) corresponding to SEQ ID No.2,
Figure 4 shows the genomic sequence of the CDS of the novel virus (PV), corresponding to SEQ ID No. 3,
Figure 5 shows the genomic sequence of PV1P corresponding to SEQ ID No.4, Figure 6 shows the genomic sequence of PV2P corresponding to SEQ ID No.5, Figure 7 shows the genomic sequence of PV3P corresponding to SEQ ID No.6, Figure 8 shows the genomic sequence of PV1F corresponding to SEQ ID No.7, Figure 9 shows the genomic sequence of PV2F corresponding to SEQ ID No.8, Figure 10 shows the genomic sequence of PV3F corresponding to SEQ ID No.9, Figure 11 shows the genomic sequence of PV1R corresponding to SEQ ID No.10, Figure 12 shows the genomic sequence of PV2R corresponding to SEQ ID No.11, and
Figure 13 shows the genomic sequence of PV3R corresponding to SEQ ID No.12.
Examples
Reference throughout the description to "an embodiment" signifies that a particular feature, structure or property specified in connection with an embodiment is included in the least in one embodiment. The expressions "in one embodiment", "in a preferred embodiment" or "in an alternative embodiment" different places in the description does not necessarily point to the same embodiment. Further, the different features, structures or properties may be combined in any suitable way in one or more of the embodiments.
Disease outbreak
A group of 70000 Atlantic salmon were stocked in sea water October 1<st>2016 at a commercial fish farm. Some mortality was observed during stocking, but the mortality was low in the period from March 2017 through June 2017. From June 2017 the mortality increased steadily. In September 2017, the mortality had reached more than 75% (Figure 1).
The fish showed symptoms of severe anaemia.
Tissue:
RNA sequencing and virus detection
On the day of slaughter, 2 September 2017, samples from gill, heart and kidney were collected from 10 fish, from the outbreak above, and sent to PatoGen for analysis by Real-Time qPCR. The samples were analysed for Paramoeba perurans, Branchiomonas, Piscine Reovirus (PRV), Pancreas Disease Virus (PDV), Paranucleospora theridion, Pravicapsula pseudobranchicola, Salmon gill poxvirus (SGPVD), Tenacibaculum maritimum, Yersinia ruckeri, Pasteurella skyensis, Piscirickettsia salmonis, and Infectious Pancreatic Necrosis Virus (IPNV) (Table 1). Although the gills were particularly affected by many agents, none of the detected agents could explain the observed anaemia and mortality, considering the symptoms and the results of the analysis.
np=not performed; nd=no data
Number of samples analysed (N)=10 except for * where N=1, **N=2, ***N=7
Tissue preparation, RNA isolation and analysis
RNA was extracted from samples of kidney, heart and gills using RNAeasy Universal kit (Qiagen) on an automated system at the PatoGen AS accredited commercial laboratory. The RNA quality and concentration of the samples were assessed with the Agilent 2100 Bioanalyzer (Agilent Technologies) and the Qubit 3.0 Fluorometer (Thermo Fischer scientific) using the RNA 6000 Pico Kit (Agilent) and the Qubit<TM>RNA HS Assay kit (Thermo Fischer Scientific).
Library preparation and RNA sequencing with Ion Torrent S5
3,2 µg of RNA from one gill sample was used as starting input for the RNA sequencing library preparation. The RNA from the sample was treated with RiboMinus TM Eukaryote Kit v2 (Thermo Fischer Scientific) to remove ribosomal RNA. The rRNA-depleted RNA was fragmented and library was constructed using the Ion Total-RNA Seq Kit v2 (Thermo Fischer Scientific). The library was bar-coded and further quantified with qRT-PCR.
Using the Ion Chef and the Ion 520<TM>and Ion 530<TM>kit Chef (Thermo Fischer Scientific), the library was clonally amplified on Ion Sphere Particles, loaded into one 530 Chip and sequenced on the Ion Torrent S5 (Thermo Fischer Scientific).
Description of bioinformatics analysis
Approximately five million reads were generated for the RNA sequencing experiment. BAM-files were converted to FASTQ files using SAMTOOLS fastq command, and we applied our in-house pathogen detection pipeline using the reads. Our approach utilizes publicly available research tools, as described in the following.
First, reads were first trimmed 25bp in both the 5’ and 3’ end, and short reads (<10 bps) were removed using VSEARCH (https://github.com/torognes/vsearch). Reads mapping to the salmon genome were then removed using Kraken (Wood and Salzberg, 2014), before sequence construction with SPADES using the remaining reads were performed (Bankevich et al., 2012). The resulting scaffold sequences were then tested for complementarity with known sequences using blastn and blastx algorithms against the nucleotide (nt) and Swiss-Prot databases, respectively.
The obtained sequence matched partly to members of the Picornaviridae family (Posavirus and Fisavirus).
The final genome is 8,713 bps, and divided into the following sections;
Bases 1-602; 5’UTR
Bases 603-8,585; CDS (2661 aa)
Bases 8,586-8,713; 3’UTR
Real Time PCR
To validate if the tentative virus, in the following referred to as PV, could be disease causing we designed a RT-PCR-assay in the presumptive polymerase region of the sequence using forward and reverse primers (PV1-F and PV1-R table 4 and 5), and a minor groove binding (MGB) probe (PV1-P) labelled with 6-FAM (table 3). The primers were designed using the software Primer Express 3.0.1 (Thermo Fischer Scientific). Secondary structures and the possibility of primer dimers were tested using the online software IDT OligoAnalyzer 3.1, and the specificity of the primers and the probe were checked using NCBI’s Blastn. The primers and the probe were found not to form secondary structures or primer dimers, nor to hybridize to any other known sequence. The primers and the probe were manufactured by Thermo Fischer Scientific.
The RT-PCR assay was performed using TaqMan® Fast Virus 1-Step Master Mix. Amplifications were done on a QuantStudio 5 Real Time PCR systemThermo Fischer Scientific) with the following conditions: 5 min at 50 °C, 20 sec at 95 °C followed by 45 cycles of 95 °C/3 sec and 60 °C/30 seconds.
Results of the qPCR
To validate if the tentative virus could be disease causing, 11 fish with clinical symptoms, and 16 fish with no symptoms were tested with the PV1- qPCR assay (Table 2). Extracted total RNA from gill, kidney or heart samples were used in the test.
-----" means negative result of detection.
The PV1 Real Time assay detected relatively high amounts of the gene sequence the PV assay targeted. There are multiple factors that may have affected the fish, however the overall trends is clear positive, and samples from the gills of all fish having clinical symptoms, were positive with Ct values ranging from 16,2 to 29,8. The gill tissue from fish with no symptoms of disease were in general negative to PV1 by Real Time assay, except for two fish that had Ct values of 35,4 and 37 which show that there are very small amount of virus detected in these fish. This confirms that the virus detected causes the observed symptoms of anaemia.
Additional qPCR assays
We have designed 3 quantitative Real Time TaqMan assays targeting various regions of the genome, as described above for PV1.
Virus stability
The virus was passed through cell lines as described 10 times, and the virus did not change.
Virulence of the virus
Juvenile Atlantic salmon were challenged to a suspension of the virus, in freshwater, under standard conditions. High mortality was observed.

Claims (17)

Claims
1. An isolated nucleic acid sequence originating from a virus in Atlantic salmon having a sequence selected from the group consisting of SEQ ID No 2 and 3, sequences being complementary to SEQ ID NO 2 and 3, and variants thereof being at least 70 % identical thereof.
2. An isolated nucleic acid sequence according to claim 1, characterized in that the sequence is at least 80 %, preferably 90%, more preferably 95 % identical with any of the sequences SEQ ID No 2 or 3, or any sequences being complementary to SEQ ID No 2 or 3.
3. A primer and/or probe having a sequence selected from the group consisting of SEQ ID NO 4-12, sequences being complementary to SEQ ID NO 4-12, and variants being at least 90 % identical with any of the sequences SEQ ID NO 4-12 and sequences being complementary to variants of SEQ ID NO 4-12.
4. A method for detection of a virus in a biological sample, characterized by comprising the following steps:
a) preparing a sample comprising nucleic acid sequences isolated from the biological sample for a reverse transcription reaction,
b) subjecting the mixture of a) to a polymerase chain reaction with a pair of primers according to claim 3, and
c) determining whether the binding of the primers to nucleotide sequences in the sample and amplification of the sequence between them have occurred indicating the presence of the virus in the sample tested.
5. A method for detection of a virus in a biological sample, characterized by comprising the following steps:
a) preparing a sample comprising nucleic acid sequences isolated from the biological sample for a reverse transcription reaction,
b) sequencing the mixture of a), and
c) comparing the resulting sequence with the sequence selected of the group consisting of SEQ ID No 2 and 3 and sequences being complementary to SEQ ID NO 2 and 3, wherein at least 70 % identity verifies the presence of the virus in the biological sample tested.
6. A method according to claim 5, wherein the sequencing is performed by a method selected from the group consisting in Illumina (Solexa) sequencing, Roche 454 sequencing, Ion Torrent and SOLiD sequencing.
7. A method according to 5 or 6, wherein the sequence is at least 80 %, preferably 90%, more preferably 95 % identical with any of the sequences SEQ ID No 2 and 3 or sequences being complementary to SEQ ID No 2 and 3.
8. Use of a nucleotide sequence comprising at the least 10 contiguous nucleotides of any of the sequences SEQ ID No 2-12, or 10 contiguous nucleotides being complementary of any of the sequences SEQ ID No 2-12, for establishing the existence of virus in a biological sample.
9. Use according to claim 8, wherein the nucleic acid sequence has a sequence selected from the group of SEQ ID No 2 and 3, or a sequence being complementary to the any of the sequences SEQ ID No 2 and 3.
10. A vector comprising a nucleic acid sequence according to any one of claims 1-2.
11. A host cell comprising a vector according to claim 10.
12. A DNA-vaccine comprising a nucleic acid sequence selected from the group consisting of SEQ ID NO 2 and 3, and variants thereof being at least 70 % identical with any of the sequences of SEQ ID NO 2 and 3.
13. A recombinant protein encoded by a nucleic acid sequence selected from the group consisting of SEQ ID NO 2 and 3, and variants thereof being at least 70 % identical with any of the sequences of SEQ ID NO 2 and 3.
14. A recombinant protein having an amino acid sequence according to SEQ ID No 1.
15. A recombinant vaccine comprising at least one recombinant protein according to claim 13 or 14.
16. An antibody specifically recognizing and specifically binding to a recombinant protein according to claim 12 or 13.
17. Diagnostic kit comprising at least one primer sequence according to claim 3.
NO20180836A 2018-06-15 2018-06-15 Novel fish virus NO344698B1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
NO20180836A NO344698B1 (en) 2018-06-15 2018-06-15 Novel fish virus
PCT/NO2019/050124 WO2019240596A1 (en) 2018-06-15 2019-06-14 Novel fish virus
EP19737262.6A EP3807403A1 (en) 2018-06-15 2019-06-14 Novel fish virus
CA3103433A CA3103433A1 (en) 2018-06-15 2019-06-14 Novel fish virus
CL2020003238A CL2020003238A1 (en) 2018-06-15 2020-12-14 New fish virus

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
NO20180836A NO344698B1 (en) 2018-06-15 2018-06-15 Novel fish virus

Publications (2)

Publication Number Publication Date
NO20180836A1 NO20180836A1 (en) 2019-12-16
NO344698B1 true NO344698B1 (en) 2020-03-09

Family

ID=67211790

Family Applications (1)

Application Number Title Priority Date Filing Date
NO20180836A NO344698B1 (en) 2018-06-15 2018-06-15 Novel fish virus

Country Status (5)

Country Link
EP (1) EP3807403A1 (en)
CA (1) CA3103433A1 (en)
CL (1) CL2020003238A1 (en)
NO (1) NO344698B1 (en)
WO (1) WO2019240596A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004045543A2 (en) * 2002-11-14 2004-06-03 Dharmacon, Inc. Functional and hyperfunctional sirna
WO2012066481A1 (en) * 2010-11-15 2012-05-24 Pharmaq As New ethiological agent
US20130045873A1 (en) * 2005-01-27 2013-02-21 Institute For Systems Biology Methods for identifying and using organ-specific proteins in blood

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003035680A1 (en) * 2001-10-19 2003-05-01 Akzo Nobel N.V. Infectious salmon anaemia virus vaccine
WO2016075277A1 (en) * 2014-11-14 2016-05-19 Patogen Analyse As Novel fish virus and method for detection

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004045543A2 (en) * 2002-11-14 2004-06-03 Dharmacon, Inc. Functional and hyperfunctional sirna
US20130045873A1 (en) * 2005-01-27 2013-02-21 Institute For Systems Biology Methods for identifying and using organ-specific proteins in blood
WO2012066481A1 (en) * 2010-11-15 2012-05-24 Pharmaq As New ethiological agent

Also Published As

Publication number Publication date
EP3807403A1 (en) 2021-04-21
NO20180836A1 (en) 2019-12-16
WO2019240596A1 (en) 2019-12-19
CL2020003238A1 (en) 2021-06-11
CA3103433A1 (en) 2019-12-19

Similar Documents

Publication Publication Date Title
Roohani et al. Characterisation of genotype VII Newcastle disease virus (NDV) isolated from NDV vaccinated chickens, and the efficacy of LaSota and recombinant genotype VII vaccines against challenge with velogenic NDV
Gong et al. Genome-wide characterization of Toll-like receptor gene family in common carp (Cyprinus carpio) and their involvement in host immune response to Aeromonas hydrophila infection
Lei et al. Difference between hemocyanin subunits from shrimp Penaeus japonicus in anti-WSSV defense
Whang et al. Characterization and expression analysis of a goose-type lysozyme from the rock bream Oplegnathus fasciatus, and antimicrobial activity of its recombinant protein
Gjerde Sarcocystis species in red deer revisited: with a re-description of two known species as Sarcocystis elongata n. sp. and Sarcocystis truncata n. sp. based on mitochondrial cox1 sequences
Nguyen et al. Identification and expression analysis of two pro-inflammatory cytokines, TNF-α and IL-8, in cobia (Rachycentron canadum L.) in response to Streptococcus dysgalactiae infection
Bellière et al. Genetic comparison among dolphin morbillivirus in the 1990–1992 and 2006–2008 Mediterranean outbreaks
WO2009098789A1 (en) Method of detecting pathogenic virus in crustacean by lamp method and reagent kit for detection
Fellahi et al. Phylogenetic analysis of avian infectious bronchitis virus S1 glycoprotein regions reveals emergence of a new genotype in Moroccan broiler chicken flocks
WO2018203757A1 (en) Novel virus in fish and a method for detection
Mao et al. Avian leukosis virus contamination in live vaccines: a retrospective investigation in China
Chen et al. Full-length transcriptome sequencing and identification and immune response of TRIM genes in Dabry's sturgeon (Acipenser dabryanus)
Shen et al. Molecular cloning, characterization and expression of the complement component Bf/C2 gene in grass carp
Thakor et al. Molecular characterization of avian adeno virus causing inclusion body hepatitis–hydropericardium syndrome in broiler chickens of Anand, Gujarat, India
Tang et al. Expression and functional characterization of interferon regulatory factors 4, 8, and 9 in large yellow croaker (Larimichthys crocea)
GB2377017A (en) Detection of foot and mouth disease virus
Abernathy et al. Gene expression profiling of a fish parasite Ichthyophthirius multifiliis: Insights into development and senescence-associated avirulence
NO344698B1 (en) Novel fish virus
Yeh et al. EcVig, a novel grouper immune-gene associated with antiviral activity against NNV infection
Wang et al. Identification of immune‐related genes from kidney and spleen of turbot, Psetta maxima (L.), by suppression subtractive hybridization following challenge with Vibrio harveyi
Godahewa et al. Complement factor D homolog involved in the alternative complement pathway of rock bream (Oplegnathus fasciatus): Molecular and functional characterization and immune responsive mRNA expression analysis
KR20150110531A (en) High resolution melt genotyping od ibv, csfv and ndv
Barghash Molecular changes in Trypanosoma evansi after treatment against trypanosomosis
KR101617142B1 (en) Nucleic acid test based avian influenza virus detection kit with improved detection accuracy
NO20201187A1 (en) Novel fish virus