Murine calicivirus

Virgin, Herbert W.

Patent Application Summary

U.S. patent application number 10/757832 was filed with the patent office on 2005-02-17 for murine calicivirus. Invention is credited to Virgin, Herbert W..

Application Number20050037016 10/757832
Document ID /
Family ID34794769
Filed Date2005-02-17

United States Patent Application 20050037016
Kind Code A1
Virgin, Herbert W. February 17, 2005

Murine calicivirus

Abstract

The invention disclosed herein relates to a newly discovered murine norovirus, and compositions and methods related thereto.


Inventors: Virgin, Herbert W.; (Clayton, MO)
Correspondence Address:
    THOMPSON COBURN, LLP
    ONE US BANK PLAZA
    SUITE 3500
    ST LOUIS
    MO
    63101
    US
Family ID: 34794769
Appl. No.: 10/757832
Filed: January 14, 2004

Related U.S. Patent Documents

Application Number Filing Date Patent Number
60440016 Jan 14, 2003

Current U.S. Class: 424/186.1 ; 435/235.1; 435/320.1; 435/325; 435/456; 435/69.3; 530/350; 536/23.72
Current CPC Class: A61K 2039/5258 20130101; C07K 14/005 20130101; C12N 2770/16021 20130101; C12N 2770/16022 20130101; C12N 7/00 20130101; A61K 39/00 20130101; C12N 2770/16023 20130101
Class at Publication: 424/186.1 ; 435/069.3; 435/320.1; 435/325; 435/456; 435/235.1; 530/350; 536/023.72
International Class: A61K 039/12; C07H 021/04; C07K 014/005; C12N 015/86

Goverment Interests



[0002] This invention was made with government support under Grant No. RO1 A149286. The United States government may have certain rights in the invention.
Claims



What is claimed is:

1. An isolated polynucleotide comprising a nucleotide sequence that is at least 80% identical to SEQ ID NO: 1, and wherein a virus encoded by said polynucleotide infects mice.

2. The polynucleotide of claim 1 wherein said sequence is at least 95% identical to SEQ ID NO:1.

3. The polynucleotide of claim 1, wherein said sequence is identical to SEQ ID NO: 1.

4. The polynucleotide of claim 1, wherein said polynucleotide consists of SEQ ID NO: 1.

5. An isolated polynucleotide comprising a sequence that is completely complementary to the sequence of claim 1.

6. An amino acid sequence encoded by the nucleotide sequence of claim 1.

7. A peptide of at least 20 amino acids encoded by the polynucleotide of claim 1.

8. A peptide of at least 20 amino acids encoded by the polynucleotide of claim 2.

9. An isolated polypeptide comprising an amino acid sequence at least 80% identical to SEQ ID NO:2, and wherein said amino acid sequence is a murine norovirus sequence.

10. The isolated polypeptide of claim 9 wherein the sequence is at least 95% identical to SEQ ID NO:2.

11. The isolated polypeptide of claim 9 wherein the sequence is identical to SEQ ID NO:2.

12. An isolated polypeptide comprising an amino acid sequence at least 80% identical to SEQ ID NO:3, and wherein said amino acid sequence is a murine norovirus sequence.

13. The isolated polypeptide of claim 12, wherein the sequence is at least 95% identical to SEQ ID NO:3.

14. The isolated polypeptide of claim 12 wherein the sequence consists of SEQ ID NO:3.

15. An isolated polypeptide comprising an amino acid sequence at least 80% identical to SEQ ID NO:4, and wherein said amino acid sequence is a murine norovirus sequence.

16. The isolated polypeptide of claim 15 wherein the sequence is at least 95% identical to SEQ ID NO:4.

17. The isolated polypeptide of claim 15 wherein the sequence consists of SEQ ID NO:4.

18. An isolated nucleotide sequence comprising at least 10 contiguous nucleotides completely complementary to the polynucleotide of claim 1, wherein said nucleotide sequence hybridizes to SEQ ID NO: 1 under highly stringent conditions.

19. The isolated nucleotide sequence of claim 18 wherein said at least 10 contiguous nucleotides are completely complementary to SEQ ID NO: 1.

20. An isolated nucleotide sequence comprising at least 10 contiguous nucleotides identical to the polynucleotide of claim 1, wherein said nucleotide sequence hybridizes to the complement of SEQ ID NO: 1 under highly stringent conditions.

21. The isolated nucleotide sequence of claim 20 wherein said at least 10 contiguous nucleotides are identical to SEQ ID NO: 1.

22. A host cell transfected with the polynucleotide of claim 1, or a portion thereof that encodes a polypeptide of at least 20 amino acids.

23. A vector comprising the polynucleotide of claim 1, or a portion thereof that encodes a polypeptide of at least 20 amino acids.

24. A host cell comprising the vector of claim 23.

25. A method for detecting the presence of murine calicivirus in a sample, comprising a) contacting the sample with the isolated nucleotide sequence of claim 18; b) providing conditions that allow specific hybridization of the nucleotide sequence of step a) to the murine calicivirus; and c) detecting whether the nucleotide sequence hybridizes to murine calicivirus.

26. The method of claim 25 wherein said detecting step further comprises quantitating the presence of murine calicivirus.

27. A method for detecting the presence of murine calicivirus in a sample, comprising a) contacting the sample with a nucleotide sequence capable of specifically hybridizing to murine calicivirus; b) providing conditions that allow specific hybridization of the nucleotide sequence of step a) to the murine calicivirus; and c) detecting whether the nucleotide sequence hybridizes to murine calicivirus.

28. The method of claim 27 wherein said detecting step further comprises quantitating the presence of murine calicivirus.

29. An isolated polynucleotide encoding a peptide of a murine calicivirus, said polynucleotide consisting essentially of: a) a nucleotide sequence selected from the group consisting of nucleotides 147-5021 of SEQ ID NO:1; nucleotides 5057-6679 of SEQ ID NO:1; and nucleotides 6682-7302 of SEQ ID NO: 1; b) a nucleotide sequence that is at least 80% identical to the polynucleotide of a); c) the complement of a) or b).

30. A vector comprising the polynucleotide of claim 29.

31. A host cell transfected with the polynucleotide of claim 30.

32. An antibody capable of selectively binding to a murine calicivirus amino acid sequence, wherein said amino acid sequence is selected from the group consisting of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, the amino acid sequence of the peptide of claim 7, and the amino acid sequence of the peptide of claim 8.

33. The antibody of claim 32, wherein said antibody is a monoclonal antibody.

34. The antibody of claim 32, wherein said antibody is a polyclonal antibody.

35. A method for identifying an agent capable of modulating or preventing murine calicivirus (MNV-1) infection, said method comprising a) providing a mouse infected with MNV-1; b) administering said agent to the mouse of a); c) monitoring the outcome of the MNV-1 infection.
Description



CROSS REFERENCE TO RELATED APPLICATIONS

[0001] This application claims benefit of priority to Provisional U.S. Patent Application Ser. No. 60/440,016, filed Jan. 14, 2003, which application is hereby incorporated herein by reference in its entirety.

BACKGROUND

[0003] The Caliciviridae are a family of positive-sense, single-stranded RNA viruses with a 7-8 kb genome that are divided into 4 distinct genera and further subdivided into genogroups. The genera Norwalk-like viruses, together with the closely related Sapporo-like viruses, recently renamed Noroviruses and Sapoviruses (Mayo, M. A., Arch. Virol. 147:1655-1656, 2002), make up human caliciviruses (Kapikian, A. Z. et al., J. Virol. 10: 1075-1081, 1972; Jiang, X. et al., Science 250:1580-1583, 1990; Jiang, X. et al., Virol. 195:51-61, 1993; Hardy, M. E. et al., Virus Genes 12:287-290, 1996). Noroviruses are responsible for more than 90% of all cases of non-bacterial epidemic gastroenteritis (Kapikian et al., 1972; Kapikian, A. Z. et al., Chapter 25 in Fields Virology, Fields, B. N. et al., Eds., 1996; Pang, X. L. et al., Pediatr. Infect. Dis. J. 18:420-426, 1999; Pang, X. L. et al., J. Infect. Dis. 181(Supp. 2): S288-S294, 2000; Fankhauser, R. L. et al., J. Infect. Dis. 178:1571-1578, 1998; Glass, R. I. et al., J. Infect. Dis. 181(Supp. 2): S254-S261, 2000; Hedlund, K. O. et al., J. Infect. Dis. 181(Supp. 2): S275-S280, 2000; Koopmans, M. et al., J. Infect. Dis. 181(Supp. 2): S262-S269, 2000; Inouye, S. et al., J. Infect. Dis. 181(Supp. 2): S270-S274, 2000). There are no current therapeutic drugs or vaccines for these important human pathogens. Sapoviruses are typically associated with sporadic cases of pediatric gastroenteritis (Pang et al., 1999; Pang et al., 2000). Two other calicivirus genera, Vesiviruses and Lagoviruses, contain animal viruses exclusively. Calicivirus genomes typically contain a large 5' open reading frame (ORF1) encoding a nonstructural polyprotein, followed by ORF2 encoding a single capsid protein. ORF2 is either in frame with ORF1 or present as an independent ORF. While the 5' end of ORF1 shows extensive sequence diversity, the remainder of ORF 1 contains motifs arranged in a specific order conserved between caliciviruses and picornaviruses. ORF3, encoding a basic protein, is present at the 3' end of the genome preceding a poly-A tract (Clarke, I. N. et al., J. Infect. Dis. 181(Supp. 2): S309-S316, 2000).

DESCRIPTION OF THE FIGURES

[0004] FIG. 1: Passage of a new pathogen by intracranial inoculation in RAG/STAT-/- and IFN.alpha..beta..gamma.R-/- mice.

[0005] The unknown pathogen was passaged into RAG/STAT-/- and IFN.alpha..beta..gamma.R-/- mice and caused lethal disease within 30 days of inoculation (A), characterized histologically by meningitis (C), vasculitis of the cerebral vessel (D), and encephalitis (E) compared to mock-infected brain (B). (B, C)RAG/STAT-/- mice; (D, E) IFN.alpha..beta..gamma.R-/- mice. Brain homogenate from an infected RAG/STAT-/- mouse was passed into 129 wild-type mice (A) and sera of these mice harvested 35 days later tested negative for mycoplasma, Sendai virus, reovirus type 3, Theiler's mouse encephalomyelitis virus (GDVII strain), lymphocytic choriomeningitis virus, pneumonia virus of mice, minute virus of mice, mouse hepatitis virus, ectromelia virus, epizootic diarrhea of infant mice, mouse cytomegalovirus, polyoma virus, K virus, orphan parvovirus, and mouse adenovirus.

[0006] FIG. 2: Sequencing and phylogenetic analysis of the MNV-1 genome.

[0007] A) Double-stranded cDNA (dsDNA) from the brain of an infected IFN.alpha..beta..gamma.R-/- mouse at passage 2 (FIG. 1) was prepared, digested with restriction enzymes, and ligated to adaptors containing PCR primer sequences to generate "tester" nucleic acids. dsDNA lacking linkers was prepared concurrently from a control brain to generate "driver" nucleic acids. Serial rounds of subtractive hybridization of tester in the presence of excess driver followed by PCR amplification of tester-specific sequences were performed to generate difference products (DP) one through four (DP1-DP4). DP3 and DP4 were cloned into pGEMT (Promega, Madison, Wis.), sequenced, analyzed using BLAST, and clones (1-8, FIG. 2A) homologous, but not identical, to calicivirus sequences were identified that spanned the Norwalk virus genome. Sequences within RDA clones (indicated by asterisks) were used to clone and sequence five fragments (a', b', c', d', e', FIG. 2A) of the MNV-1 genome after PCR or 5' and 3' RACE (Marathon cDNA amplification kit, Clontech, Palo Alto, Calif.). The 5' end of the genome was difficult to clone and consequently the first 15 nucleotides are based on a single sequence, while the remaining sequence has at least a 10-fold redundancy. This may explain why there is no start codon close to the 5' end as is expected based on comparison with other Noroviruses. B) Schematic of the final 7726 bp MNV-1 genome sequence with predicted open reading frames (ORFs). The locations of amino acid motifs in ORF1 are indicated: 2C helicase: GXXGXGKT (SEQ ID NO: 50); 3C protease: GDCG (SEQ ID NO: 51); 3D polymerase: KDEL (SEQ ID NO: 52), GLPS(SEQ ID NO: 53), YGDD (SEQ ID NO: 54). The putative S and P domains of the ORF2 encoded capsid protein were identified based on sequence alignments with Norwalk virus. AAA: 3' poly-A tail. C) Alignment of the complete MNV-1 genome with complete genomes of representative members of the four Caliciviridae genera and members of the most closely related virus family, the Picornaviridae. Specific members were chosen based on the 2000 taxonomy study by Green et al. (J Infect Dis '00 v. 81 p. S322). D) Alignment of the capsid protein sequence of MNV-1, done as in C. Note that the alignments in C and D were confirmed using other algorithms (data not presented).

[0008] FIG. 3: Sequence variability of MNV-1. A) All variable nucleotides within ORF1 and ORF2, based on sequence analysis of multiple clones of the entire MNV-1 genome, are depicted. These nucleotides had 20% or less variability between clones. B) Sequences of individual clones spanning nucleotides 1767 to 1893 (solid box on ORF1 in panel A), with variable positions highlighted with arrowheads SEQ ID Nos: 21-48). The consensus sequence of MNV-1 is shown at the bottom (bold type) (SEQ ID NO:49), with variable nucleotides highlighted by arrowheads.

[0009] FIG. 4: Purification and pathogenicity of MNV-1.

[0010] MNV-1 was purified from an infected IFN.alpha..beta..gamma.R-/- mouse brain homogenate by CsCl density gradient centrifugation. As a control, mock-infected mouse brain homogenates were processed similarly. (A) Determination of the average buoyant density of genome-containing MNV-1 particles. Dialyzed gradient fractions were analyzed by MNV-1 specific RT-PCR (Titanium one-step RT-PCR kit, Clontech, Palo Alto, Calif.) and products were separated on a 1% agarose gel. Primers were chosen in ORF 1 to yield an expected product of 184 bp (indicated by the asterisk). (B) MNV-1 virions visualized by EM. Samples were absorbed onto formvar/carbon-coated grids for 1 min. The grids were washed in dH.sub.2O, stained with 2% aqueous uranyl acetate (Ted Pella Inc., Redding, Calif.) for 1 min, and air dried prior to viewing on a JEOL 1200EX transmission electron microscope (JEOL USA, Peabody, Mass.). (C) Survival of RAG/STAT-/- mice infected i.c. with unpurified, or purified MNV-1, as well as gradient fractions from mock-infected brain. The P values for mock versus infected mice are indicated. Statistical analyses were performed using GraphPad Prism software.

[0011] FIG. 5: IFN.alpha..beta. or IFN.gamma. receptors and STAT1 are required to protect from lethal MNV-1 challenge.

[0012] A MNV-1 stock was prepared as a brain homogenate from 17 IFN.alpha..beta..gamma.R-/- mice inoculated i.c. three days previously with brain homogenate from a passage 2 (FIG. 1) mouse. Infected brains were homogenized in sterile PBS and filtered through a 0.2 .mu.m filter. Brains from five IFN.alpha..beta..gamma.R-/- mice inoculated i.c. with uninfected brain tissue were used to generate a mock virus stock. Mice of various strains were inoculated with MNV-1 or mock-inoculated using 10 .mu.l intracerebrally (ic), 25 .mu.l intranasally (in), or 25 .mu.l perorally (po). A number of mouse strains did not show increased mortality compared to wild-type 129 controls (A). The survival after inoculation with MNV-1 or mock virus is shown for IFN.alpha..beta..gamma.- R-/- mice (B), STAT1-/- mice (C), RAG/STAT-/- mice (D), and STAT1/PKR-/- mice (E). All p values for mock versus infected groups were.ltoreq.0.0001 except: IFN.alpha..beta..gamma.R-/- i.n.: p=0.002; STAT1-/- i.n.: p=0.097; and STAT1-/- p.o.: p=0.034. Statistical analyses were performed with GraphPad Prism software.

[0013] FIG. 6: Generation of MNV-1 virus-like particles.

[0014] A) Western blot analysis of cell lysates from High-Five cells infected with recombinant baculovirus expressing the MNV-1 capsid protein (see Example 9) or a control baculovirus expressing the LacZ cassette (negative control). Proteins were detected by ECL Plus after incubation with serum from a MNV-1 infected mouse followed by a HRP-labeled secondary antibody. The size of the molecular weight marker is indicated on the right. B)-D) Electron microscopy of negatively stained VLPs. Supernatants of High-Five cells infected with a control baculovirus expressing LacZ (B), recombinant baculovirus expressing the MNV-1 capsid protein (C), or VLPs purified from these supernatants (D) were stained with uranyl acetate and photographed at a magnification of 50,000.times..

[0015] FIG. 7: Reactivity of mouse serum against MNV-1 VLP supernatants or cell lysates by ELISA.

[0016] Supernatants of High-Five cells infected with recombinant baculovirus expressing the MNV-1 capsid protein or LacZ expressing control were coated on ELISA plates. A) Analysis of half-log serial dilutions of serum from MNV-1 infected mice or 129 wild type mice. B) Analysis of 1:10 dilution of several cages of STAT-/- mice. Each dot represents one mouse. Reactivity was assessed after incubation with a HRP-coupled secondary antibody and colorimetric detection at 405 nm. Cages 1, 3, 4, 5 and 6 contained seronegative mice. Cages 2, 7, 8, and 9 contained seropositive mice.

[0017] FIG. 8: Tissue MNV-1 RNA levels after infection via different routes.

[0018] Four IFN.alpha..beta..gamma.R-/- mice were inoculated with MNV-1 i.c. (10 .mu.l), p.o. (25 .mu.l), or i.n. (25 .mu.l). Two mice were sacrificed at both 2 and 7 dpi and lung (Lu), intestine (Int), brain (Br) and feces were collected. RNA was extracted from each organ, and cDNA was synthesized and used (5 ng) in triplicate real time PCR reactions. Primers specific to a 131 nucleotide region of ORF 1 were used (sense=cagtgccagccctcttat (SEQ ID NO:19); antisense=gtcccttgatgaggagga (SEQ ID NO:20)). Signal was compared to a standard curve generated using a plasmid containing target sequences. Triplicate reactions were performed using GAPDH primers to verify equivalent amounts of starting template (not shown). The levels of virus RNA as log.sub.10 MNV-1 genome copies are shown (open bars=2 dpi, solid bars=7 dpi, *=undetectable levels).

[0019] FIG. 9: Immunohistochemical staining of spleen secti ns from MNV-1 infected mouse. Formalin-fixed spleen sections from a STAT1-/- animal 3 days after p.o. inoculation with MNV-1 were stained with either immune polyclonal rabbit serum inoculated with bacterially expressed MNV-1 capsid protein (left panel), or with the preimmune serum from the same rabbit (right panel). Immunohistochemistry was performed with the PerkinElmer.TM. TSA.TM.-Plus DNP (HRP) System, according to the supplied protocol. Primary antibodies were used at a 1:25 dilution. Positive cells are indicated by arrows.

[0020] FIG. 10: Single copy sensitivity of MNV-1 cDNA detection by nested PCR assay. Nested PCR primers specific to a region of MNV-1 ORF2 were designed (outer-sense=gcgcagcgccaaaagccaat (SEQ ID NO: 15); outer-antisense=gagtcctttggcatgctacccagg (SEQ ID NO: 16); inner-sense=gccgccgggcaaattaacca (SEQ ID NO: 17); and inner-antisense=ggcttaacccctaccttgccca (SEQ ID NO: 18)). A) Multiple PCR reactions with either 1 or 10 copies of a plasmid containing the appropriate region of MNV-1 were performed. 3/4 and 4/4 reactions were positive for 1 and 10 copies, respectively. The expected size of the PCR product is 153 bp. B) cDNA was generated from spleen tissue of 10 IFN.alpha..beta..gamma.R-/- mice and 1 .mu.g of each was used in nested PCR reactions (7/10 samples were positive). All water controls are negative.

DESCRIPTION

[0021] It has been discovered that mice doubly deficient in STAT1 and RAG2 (RAG/STAT) contained an infectious pathogen that caused severe encephalitis and could be serially passaged by intracerebral (i.c.) inoculation (FIG. 1). Lethal infection was associated with encephalitis, vasculitis of the cerebral vessels, meningitis, hepatitis, and pneumonia (FIG. 1 and data not shown). Disease was passed by filtered samples, suggesting the presence of a virus (FIG. 1A). Sera of 129 mice infected with the putative virus tested negative for an extensive panel of mouse pathogens (see legend of FIG. 1). Brain homogenate from an infected RAG/STAT-/- mouse was passed into 129 wild-type or IFN.alpha..beta..gamma.R-/- mice before and after filtration. A full work-up was performed on mice from passages 1 and 2, including histopathology, electron microscopy, standard clinical virology and microbiology work-ups, as well as special stains of histology sections (GMS, AFB [acid-fast bacilli], Gram stain). All of these failed to reveal the nature of the pathogen.

[0022] The pathogen is more virulent in mice lacking both the interferon .alpha..beta.(IFN.alpha..beta.) and the interferon .gamma.(IFN.gamma.) receptors (IFN.alpha..beta..gamma.R-/-, .sup.2) than in wild-type mice (see below) and it passes through a 0.2 .mu.m filter (see above and FIG. 1A). The pathogen does not appear to cause cytopathic effect on HeLa cells, Vero cells or murine embryonic fibroblasts (including those lacking IFN receptors or STAT1). These data suggest that a previously unknown IFN-- sensitive but non-cultivatable pathogen that was <0.2 .mu.m in size was present in diseased mice.

[0023] Identification and Sequencing

[0024] To identify the new pathogen a previously published representational difference analysis protocol (RDA) was used (See Pastorian et al., Anal. Bicochem. 283:89-98 (2000), which is hereby incorporated in its entirety). Double-stranded cDNA (dsDNA) from the brain of an infected IFN.alpha..beta..gamma.R-/- mouse at passage 2 (FIG. 1) was prepared, digested with restriction enzymes, and ligated to adaptors containing PCR primer sequences (tester) (see Pastorian protocol for sequences of RDA primers). Control dsDNA lacking linkers was prepared concurrently from a control brain (driver). Serial rounds of subtractive hybridization of tester in the presence of excess driver followed by PCR amplification of tester-specific sequences were performed to generate difference products (DP) one through four (DP1-DP4). DP3 and DP4 were cloned and sequenced. Three of 24 clones from DP3 and ten of 48 clones derived from DP4 had significant homology to multiple caliciviruses (data not shown). These RDA clones spanned the Norwalk virus genome (FIG. 2A), but were not identical to any known full or partial calicivirus sequence, demonstrating that we had identified a novel calicivirus. This new virus is referred to herein as murine Norovirus-1 (MNV-1).

[0025] To determine the relationship of MNV-1 to other caliciviruses, the MNV-1 genome was cloned and sequenced from cDNA of an infected mouse brain using a combination of 5' and 3' RACE and PCR (FIG. 2A). Sequencing was performed in both directions with 10-fold redundancy to obtain a consensus sequence with the exception that the 5' 15 nucleotides were obtained from a single clone. The assembled genome included 7726 bp of unique sequence plus a 3' polyA tail, and contained the expected three ORFs conserved across the Caliciviridae (FIG. 2B). Phylogenetic analysis using the CLUSTAL W algorithm, and other algorithms including PAUP, aligning either the complete genome sequence (FIG. 2C) or the capsid protein sequence (FIG. 2D) of MNV-1 with corresponding sequences from members of the four calicivirus genera and several members of the Picornaviridae family revealed that MNV-1 is a Norovirus that does not cluster within previously identified genogroups (FIGS. 2C, D)(Green KY JID 181 S322-330). Therefore, it is proposed that MNV-1 is exemplary of a new Norovirus genogroup.

[0026] Thus, disclosed herein is a pathogen that infects mice, referred to herein as Murine Norovirus-1 (MNV-1). MNV-1 is both a unique norovirus, and is the first member of a new genogroup of Noroviruses. An exemplary sequence for the MNV-1 virus and genogroup is provided as SEQ ID NO: 1, which is a consensus sequence representative of the full length MNV-1 genome as determined from a series of clones derived by PCR or RACE analysis from RNA derived from the brain of an infected mouse. Thus, one embodiment comprises an isolated RNA sequence as shown in SEQ ID NO: 1. An additional embodiment comprises sequences of MNV-1 isolates that vary from the sequence in SEQ ID NO: 1 by an amount determined by both sequence analysis and current understanding of the relatedness of different caliciviruses (see below). One embodiment comprises the viruses related directly to MNV-1 as viral quasispecies. Another embodiment comprises other members of the MNV-1 genogroup of which MNV-1 is the defining member. The criteria for viral quasispecies and viral genogroup are defined below, and serve to specifically set criteria for the MNV-1 embodiments described herein.

[0027] RNA viruses vary during infection due to errors made by the viral RNA-dependent RNA polymerase. Thus, MNV-1 (a positive-strand RNA virus) may be expected to vary during replication into a quasispecies comprising multiple viruses with sequences closely related to, but not identical to, the sequence of the original infecting virus. Thus, some embodiments of MNV-1 include viruses with sequences that vary from the sequence provided in SEQ ID NO: 1 by an amount consistent with variation within a calicivirus quasispecies. The level of variation from the MNV-1 consensus SEQ ID NO: 1 that still is considered by those skilled in the art to be the same virus (since these viruses always exist as quasispecies) is 5-7% (Radford et. Al. Veterinary rewcord Jan. 29, 2000 pp 117 on, Radford et al Vet Record Oct. 20, 2001 pp 477 on). Thus, an embodiment comprises the MNV-1 viral quasispecies of sequences that vary from our initial consensus sequence (SEQ ID NO: 1) by no more than 5%. It has been confirmed that there is significant variance in MNV-1 nucleotide sequence even within a single infected animal (FIG. 3). To show this, a portion of the primary data from which the 10-fold redundant consensus sequence SEQ ID NO: 1 was derived is presented. It was found that over the highly conserved ORF2 region, there are multiple sites at which there is sequence variation (FIG. 3A). A portion of the sequence data is presented in FIG. 3B for a region within which sequence variation was found. The frequency of variation at the sites shown in boxes is greater than that observed at multiple other sites (e.g. the remainder of the sequence in FIG. 3B), showing that these variations represent true biological variation rather than PCR artifacts or sequencing errors. Thus, MNV-1 does exist as a quasispecies.

[0028] Further embodiments comprise viruses with an amount of variance from SEQ ID NO: 1 that is consistent with variation within a genogroup, and less than the variation observed between genogroups. For caliciviruses, genogroup and genus definition has been developed and officially set by the International Committee on the Taxonomy of viruses (ICTV) and research in the field has led to definitions of the amount of variation in sequence that is expected within a single genogroup as opposed to between viruses of different genogroups (K. Y. Green et al JID 2000 S322-330). Because nucleotide sequences can vary without causing variation in amino acid sequence, relatedness at the nucleotide level is a preferred method for distinguishing between genogroups or within a quasispecies (see above). Nucleotide identity within a genogroup of Noroviruses has been established as greater than 80% within the highly conserved capsid protein (ORF 2) gene (J. Vinje et al Arch Virol (2000) 145:223-241). Thus, viruses that differ by more than 20% at the nucleotide level from a member of a genogroup (in this case from the MNV-1 sequence in SEQ ID NO:1) are not members of the genogroup. Nucleotide identity between genogroups is 64%-78% or less. Therefore, the genogroup to which MNV-1 belongs comprises viruses that vary by no more than 20% from the MNV-1 sequence within the capsid region. Similar reasoning applies to other conserved regions of the genome including the RNA dependent RNA polymerase gene. Therefore, our use of the capsid sequence for the definition of genogroup is standard.

[0029] Further embodiments include RNA sequences that are at least about 80% identical to SEQ ID NO: 1, where the % identity is determined using Vector NTI AlignX program. Other embodiments include an isolated DNA sequence, or fragments thereof, identical to or complementary to SEQ ID NO: 1, and isolated DNA sequences at least about 80% identical to or complementary to SEQ ID NO: 1. Further embodiments comprise sequences between 80% and 100% identical to SEQ ID NO: 1, and sequences complementary thereto.

[0030] Additional embodiments comprise fragments of any of the above mentioned sequences, such as may be used, for example, as primers or probes. Examples of such sequences include the primers listed in legends to FIGS. 8 and 10 that were used to detect virus infection in animals by nested PCR (FIG. 10) or to determine the amount of MNV-1 genome in a tissue by the use of real time PCR (FIG. 8). These primers will be useful for detection of MNV-1 infection in commercially bred mice and for quantitation of MNV-1 in tissues after trials of antiviral agents or vaccines. Such primers and probes are selected such that they are substantially complementary to a target sequence, wherein the target sequence consists of coding sequence of MNV-1. Here, substantially complementary means that the primer or probe is sufficiently complementary to the target sequence that it will hybridize to the target sequence under highly stringent conditions. As used herein, highly stringent conditions are as defined in the nested and real time PCR protocols exemplified in FIGS. 8 and 10. For hybridization in blots as opposed to PCR reactions, stringent refers to: hybridization at 68 degrees C. in 5.times.SSC/5.times. Denhardt's solution/1.0% SDS, and washing in 0.2.times.SSC/1.0% SDS at room temperature. Such probes and primers are useful, for example in various assays for detecting the presence of MNV-1 (FIG. 10) and determining how much MNV-1 is in a particular sample (FIG. 8). Other assays for which such primers or portions of MNV-1 sequence would be useful include Northern and Southern hybridization blot assays, additional PCR assays (e.g. degenerate PCR using primers with degenerate nucleotides at specific sites within the PCR primer to detect viruses within the MNV-1 genogroup but not identical to the MNV-1 sequence in SEQ ID NO: 1), transcription-mediated amplification assays and the like, and as positive controls and internal standards for commercial assays to detect the presence of MNV-1 in mice or after treatment with anti-viral agents or vaccines.

[0031] A feature that distinguishes the human Noroviruses from the Sapoviruses are the cup-shaped depressions on the virion surface that gave the calicivirus family its name (calyx=cup in Latin). Sapovirus capsids show these characteristic cup-shaped depressions by electron microscopy (EM), while Norovirus capsids have a feathery appearance. To visualize MNV-1 virions, MNV-1 was purified from the brain of an infected IFN.alpha..beta..gamma.R-/- mouse on CsCl gradients (FIG. 4). Gradient fractions containing MNV-1 genome were identified by RT-PCR (FIG. 4A), revealing a buoyant density of MNV-1 of 1.36 g/cm.sup.3+/-0.04 g/cm.sup.3 (n=3 experiments), in close agreement with the published buoyant densities of Noroviruses (1.33-1.41 g/cm.sup.3). Analysis of these gradient fractions by EM revealed particles with a diameter of 28-35 nm (FIG. 4B), similar to the known size (26-32 nm) of Norovirus particles in negatively stained preparations. The particles were icosahedral and had the same feathery surface morphology as Noroviruses but lacked the cup-like depressions characteristic of Sapoviruses. Gradient fractions prepared from mock-infected brain did not contain these particles (data not shown).

[0032] To test the pathogenicity of MNV-1, mice were infected i.c. with CsCl gradient purified MNV-1. These virions were infectious since 18/18 RAG/STAT mice inoculated with them died, while 18 of 18 mice inoculated with gradient fractions prepared from a mock-infected brain survived (FIG. 4C). Mice inoculated with gradient-purified virions showed encephalitis, meningitis, cerebral vasculitis, pneumonia, and hepatitis (data not shown). This mortality rate and pathology was similar to that observed previously in mice inoculated with unpurified brain homogenate (FIG. 4C and data not shown). The presence of disease in mice inoculated with CsCl-purified MNV-1 demonstrates that MNV-1 is the causative agent of the disease initially detected and passed (FIG. 1).

[0033] The MNV-1 genome comprises three open reading frames (ORFs). Analysis of the predicted coding sequence of ORF1 indicated a polyprotein with a molecular weight (MW) of 180.7 kDa and revealed the presence of multiple conserved motifs shared by caliciviruses and picornaviruses (FIG. 2B). ORF2 is separated from ORF1 by 32 nt and starts in the -1 frame relative to ORF1. It encodes a 542 aa protein with a calculated MW of 58.9 kDa and an isoelectric point of 5.19. When this gene was expressed in a recombinant baculovirus, virus-like particles (VLPs) were found in the supernatant of infected cells, demonstrating ORF2 encodes the capsid protein (FIG. 6). These VLPs provide a reagent for analysis of MNV-1 infection (see below). The predicted ORF3 encodes a basic protein (pI of 10.22) with a calculated MW of 22.1 kDa that overlaps by 2 nt with ORF2 and is expressed in the +I frame relative to ORF 1 but the -2 frame relative to ORF2.

[0034] Thus, further embodiments comprise the amino acid sequences encoded by ORF1, ORF2 and ORF3. These amino acid sequences are shown in SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO:4, respectfully. Additional embodiments comprise amino acid sequences that are encoded by viruses that vary from SEQ ID NO: 1 by no more than 20% at the nucleotide level as defined above. The protein translation of such sequences will vary on a percentage basis depending on the placement of nucleotides within codons and the frequency of amino acids coded for by single versus multiple three base pair codons used by the translational machinery. Therefore the extent of variation of these embodiments is properly determined by defining the extent of total nucleotide variation accepted as defining the MNV-1 genogroup. Some embodiments comprise the nucleotide sequences that encode each of the amino acid sequences of SEQ ID NO:2, SEQ ID NO:3 and SEQ ID NO:4, including degenerate variants that encode those amino acid sequences. Additional embodiments comprise the nucleotide sequences of ORF 1, ORF2 and ORF3 of MNV-1

[0035] Additional embodiments include vectors capable of expression of any of the proteins encoded by MNV-1 or their variants as defined above. Examples of suitable vectors include baculovirus vectors, alphavirus vectors (e.g. Sindbis virus vectors, VEEV replicons), retroviral vectors, plasmids within which expression is driven from eukaryotic promoters, plasmids that generate short RNA sequences suitable for gene inactivation by RNAi technology, plasmids in which the presence of an RNA polymerase transcribes MNV-1 sequences (including the entire sequence) in order to provide RNA (including up to full length infectious RNA) for analysis or transfection into cells. Infectious RNA is defined as RNA, which, on transfection into eukaryotic cells, gives rise to intact infectious virus. Portions of the genome may also be expressed in this fashion for the generation of viral proteins or for analysis of the processing of MNV-1 viral proteins for the purpose of developing assays for identification of steps in viral replication that may serve as drug targets. Additional uses of expression vectors include generation of cells expressing viral proteins in a stable fashion for the purpose of screening anti-viral antibodies or for providing positive controls for assay for detection of anti-viral antibody in the serum.

[0036] As discussed above, expression of the capsid protein, i.e., the protein encoded by the sequence of ORF2, results in the formation of virus-like particles (VLPs). Thus, some embodiments comprise methods of producing VLPs. Such methods comprise transfecting a cell or animal with a vector that encodes the MNV-1 capsid protein, and recovering VLPs, or expression of the capsid protein from within the baculovirus genome such that the capsid protein is produced in insect cells infected with the baculovirus expressing the capsid protein. Further embodiments comprise MNV-1 VLPs. VLPs are useful, for example, for isolation of antibodies, analysis of the epitopes that antibodies recognize, and for cryo-EM and X-ray crystallography and other methods for determining the three dimensional structure of the MNV-1 capsid. VLPs may also be studied for potential use as a vaccine. In this setting they may be useful for mapping the specific conformational epitopes recognized by anti-viral antibodies and the specific peptides recognized by antiviral CD4 and CD8 T cells.

[0037] Antibodies

[0038] Some embodiments comprise antibodies that bind specifically to any of the various proteins encoded by the MNV-1 genome. Methods for producing antibodies are known in the art. Such antibodies may be either monoclonal or polyclonal. Antibodies can be used in various assays, such as for example ELISA assays, to detect the presence of MNV-1 in a sample. Samples include for example serum, saliva, feces, and tissues. In addition, antibodies may be utilized in methods for preventing lethal MNV-1 infection and studied for potential use as vaccines or anti-viral therapeutics.

[0039] An example of the use of antibodies and antibody detection assays is the demonstration that seroconversion can be detected by ELISA of serum using MNV-1 VLPs as the target antigen bound to the ELISA plate (FIG. 7). A further example is the demonstration that MNV-1 infection can be detected in specific cells by using immunohistochemistry to detect the binding of MNV-1 specific antibodies to infected cells (FIG. 9). This type of use may also be employed for detecting binding of virus to cells by FACS analysis. This in turn will provide an opportunity to identify the receptor for MNV-1. Identification of the MNV-1 receptor on the cell surface may provide important targets for anti-viral drug development. In addition, antibodies will be used for immunofluorescence and in-situ detection of virus infected cells.

[0040] Small Animal Model

[0041] The discovery of the first murine Norovirus provides the first small animal model for development and testing of pharmaceuticals and vaccines for treatment and prevention of a major human disease. This presents an opportunity to answer important questions regarding the pathogenesis of Norovirus infections, to determine the role and mechanisms of immunity in either protection against infection or immunopathology, to identify novel therapeutic targets for treatment of human calicivirus disease, and to better understand how innate immunity can control enteric virus infection. The mouse model can also be used in methods to identify agents that alter calicivirus infection and disease.

[0042] The course of human Norovirus infection strongly suggests that symptoms are caused by acute infection. Prominent amongst the clinical manifestations are vomiting and diarrhea with a mean incubation period of 24 hours and duration of 24-48 hours. Understanding of the viral and host mechanisms involved in the induction and clearance of human Norovirus disease is rudimentary. Acquired immunity can play a role in Norovirus resistance, but may not explain why certain individuals get severe disease while others do not. It may be that long-term immunity can be achieved, and the use of the MNV-1 virus in a small animal model provides the first opportunity to define such possible mechanisms. Infected individuals can develop short-term immunity to homologous virus, but the development of long-term immunity is questionable. An unexpected inverse relationship between pre-challenge antibody levels and susceptibility to infection has been reported in some studies (Parrino, T. S., et al., N. Engl. J. Med. 297:86-89, 1977; Johnson, P. C. et al., J. Infect. Dis. 161:18-21, 1990; Okhuysen, P. C. et al., J. Infect. Dis. 171:566-569, 1995), while others have reported that circulating antibody does correlate with resistance to calicivirus infection (Lew, J. F. et al., J. Infect. Dis. 169:1364-1367, 1994; Ryder, R. W. et al., J. Infect. Dis. 151:99-105, 1985; Nakata, S. et al., J. Infect. Dis. 152:274-279, 1985). This controversy has led to studies showing that non-immune host factors, such as blood groups, influence susceptibility to infection (Hutson, A. M. et al., J. Infect. Dis. 185:1335-1337, 2002). The discovery of MNV-1 provides a small animal model for the study of Noroviruses.

[0043] One embodiment is therefore the use of mice infected with MNV-1 as an approach to identifying the efficacy of vaccines or therapeutic agents. Mice would be infected with the newly discovered virus, and then treated with candidate agents and the outcome of the infection monitored using ELISA (FIG. 7), quantitative real time PCR for the viral RNA (FIG. 8), immunohistochemistry (FIG. 9), lethality (FIG. 5), or in situ hybridization to monitor the course of infection. Similarly, another embodiment is the use of mice infected with MNV-1 to test the efficacy of vaccination protocols against the virus. In this case, different vaccine preparations (including vectors expressing portions of the new virus genome or proteins from the virus or from human noroviruses that cross react with proteins from the mouse virus) would be administered to infected mice and the effect of vaccination on the course of the infection monitored using ELISA (FIG. 7), quantitative real time PCR for the viral RNA (FIG. 8), immunohistochemistry (FIG. 9), lethality (FIG. 5), or in situ hybridization. As it is not practical to perform such experiments in humans, the capacity to perform these types of screens for in vivo efficacy of therapeutics or vaccines is not possible without the use of this newly described virus.

[0044] In addition, the discovery of MNV-1 and the generation of a consensus sequence will enable construction of an infectious clone for MNV-1. One embodiment is therefore generation of such an infectious clone from the current cloned and sequenced genome or from sequences that vary within the limits described above for the MNV-1 quasispecies or MNV-1 genogroup. Such a clone can be used to develop various screening assays for MNV-1 antiviral agents and targets for antiviral drug development and vaccines for prevention of infection or antibodies for therapy of disease, and also may be used to study certain aspects of the viruses infection cycle including binding, entry, uncoating, negative strand RNA synthesis, positive strand RNA synthesis, subgenomic RNA synthesis, synthesis of structural and non-structural proteins, viral assembly and viral egress to be studied and used to develop screens for antiviral drugs that might have uses in preventing or treating Norovirus induced disease. In addition, placement of portions of human Noroviruses into an infectious clone for MCV-1 (e.g. substituting proteins such as the capsid of RNA polymerase of the human virus into the mouse virus infectious clone) will allow the murine virus to be humanized and potentially still used in mice. This will allow screening of therapeutic agents that target the functions of human norovirus proteins in an animal model. This is possible only through the combined use of an infectious MNV-1 clone as a vector for expressing functional proteins and a small animal model which allows assessment of the effects of therapeutic agents or vaccines on the course of infection with such "humanized" forms of the mouse calicivirus MNV-1.

[0045] In addition, the use of the newly discovered MCV-1 virus in mice with different immune deficiencies will allow identification of host proteins that play a role in control of Norovirus infection. An example of this is the detection of the critical role of STAT-1 in resistance to MNV-1 infection (Working Example 14, FIG. 5). Identification of such host proteins could allow development of targeted therapeutic agents that enhance specific parts of the host immune response as a way to treat or prevent Norovirus disease. Such embodiments include, for example, use of the virus in mice with deficiencies in specific parts of the immune system in order to identify mice that have increased susceptibility or increased resistance to infection by MCV-1. Such embodiments would be useful for identifying immune protective or immunopathologic aspects of the host response and thereby inform searches for vaccines or therapeutic agents that could prevent or treat Norovirus infection. An example would be targeting enhanced STAT-1 function, based on the experiments in FIG. 5, for prevention of Norovirus disease in humans.

WORKING EXAMPLES

Example 1

Generation of MNV-1 Stock

[0046] After identification of MNV-1 in RAG/STAT and IFN.alpha..beta..gamma.R-deficient mice, a brain homogenate from an IFN.alpha..beta..gamma.R-deficient mouse at passage 3 was used for i.c. inoculations of 17 additional IFN.alpha..beta..gamma.R-deficient mice. Brains of infected mice were harvested 3 days post-infection and homogenized in PBS. Homogenates were centrifuged at low speed and filtered through a 0.2 .mu.m filter and the resulting supernatant was used in subsequent infections. For control experiments, brain homogenates of mock-infected mice were prepared similarly after injection of mice with uninfected mouse brain homogenate. (See FIG. 5).

Example 2

Purification of MNV-1 Virions

[0047] Homogenate from one MNV-1 infected brain was used for purification of MNV-1 virions while a mock-infected mouse brain was used as a control (FIG. 4). Homogenized brain was subjected to a cycle of freeze/thaw and two low speed centrifugations before filtration through 0.22 .mu.m filter. Supernatants were centrifuged at 90,000.times.g for 2 hrs and the resulting pellets were incubated for 30 min at 37 C in 1 ml 1% Na-deoxycholate. The resulting material was mixed with CsCl to a final density of 1.36 g/cm.sup.3 and centrifuged for 40 hrs at 150,000.times.g. Gradients were fractionated, their density determined with a refractometer, and dialyzed against a buffer containing 0.01M Tris-HCl, 0.15M NaCl, 1 mM CaCl.sub.2, and 0.05M MgCl.sub.2. (See FIG. 4).

Example 3

RNA Isolation, cDNA Synthesis, and RDA

[0048] Total RNA was isolated from a MNV-1 infected mouse brain using Trizol (Invitrogen, Carlsbad, Calif.) following the manufacturer's instructions. Double-stranded cDNA for use in RDA was synthesized from total RNA using the Superscript Choice System for cDNA synthesis (Invitrogen, Carlsbad, Calif.) and a combination of random hexamers and oligo dT primers. Single-stranded cDNA for quantitative PCR was generated using Supercript II (Invitrogen, Carlsbad, Calif.) following the manufacturer's recommendations. RDA was performed as described by Pastorian et al. (Anal. Biochem. 283:89-98, 2000) with the following modification. The QIAquick PCR purification kit (Quiagen, Valencia, Calif.) was used to remove unincorporated nucleotides and small cDNA species. Difference products from rounds 3 and 4 were cloned into the pGEM-T vector system (Promega, Madison, Wis.) following the manufacturer's instructions. Bacterial colonies were grown up and inserts were PCR amplified for sequencing. (See FIGS. 2, 3).

Example 4

RT-PCR and Quantitative PCR

[0049] RT-PCR was performed with primers 445/1/AS6 (TCCAGGATGACATAGTCCAGGG- GCG)(SEQ ID NO:5) and 445/1/S6 (TGGGATGATTTCGGCATGGACAACG) (SEQ ID NO:6) using the Titanium one-step RT-PCR kit (Clontech, Palo Alto, Calif.) following manufacturer's recommendations. Quantitative PCR (FIG. 8) was performed with primers ORF1/RT1/S (cagtgccagccctcttat) and ORF1/RT1/AS2 (tcctcctcatcaagggac) that amplify a 132 bp segment of ORF1 outside of the predicted subgenomic RNA. This assay has a sensitivity of 100 viral genomes/.mu.g cellular RNA or about 1 MNLV-1 genome per 1720 cell equivalents of RNA (estimating 1 .mu.g cellular RNA/I 72,000 cells). The assay linearly quantitates genome over at least a 6-log range. (See FIG. 8).

Example 5

Cloning of the MNV-1 Genome

[0050] A combination of PCR and RACE was used to clone the MNV-1 genome (FIG. 2A). For internal sequence information, primers were constructed based on sequence information obtained through RDA and used to amplify and clone larger pieces of MNV-1 from 1.sup.st strand cDNA from a RAG/STAT mouse brain (passage 3). These PCR products were cloned into the pGEMT vector (Promega, Madison, Wis.) and universal M13 forward and reverse primers used for sequencing. Primer walking was applied when necessary. For the 5' and 3' ends of MNV-1, RACE was performed with the Marathon cDNA Amplification Kit (Clontech, Palo Alto, Calif.) using total RNA from the same RAG/STAT mouse brain (passage 3) as starting template. These products were cloned and sequenced as outlined above. A consensus sequence with at least 10-fold redundancy (except for the 5'end, see below) was constructed using the VectorNTI contig program. The 5'end of the genome was difficult to clone and consequently the first 15 nucleotides are based on a single sequence, possibly explaining why there is no start codon close to the 5'end as is expected based on comparison with other Noroviruses. (See FIG. 2).

Example 6

Cloning and Expression of the MNV-1 Capsid Protein in Bacteria

[0051] The MNV-1 capsid protein was PCR amplified from first strand cDNA from a RAG/STAT mouse brain (passage 3). The following primers C-pET1 GTGGTGCTCGAGTGCGGCCGCAAGCTTTATTATTGTTTGAGCATTCGGCCTG) (SEQ ID NO:7) and N-pET 1 (ATCCGAATTCTAGATGCACCACCACCACCACCACATGAGGATGAGTGATGGCGCA G) (SEQ ID NO:8) containing HindIII and EcoRI restriction sites (underlined), respectively, and a 6 Histidine N-terminal tag (bold) were used in a 2 step PCR reaction (5 cycles 50 C.sub.1-30 cycles 60 C) in the presence of 5% DMSO. The resulting PCR product was ligated into a PCR blunt cloning vector (Zero Blunt PCR Cloning kit, Invitrogen, Carlsbad, Calif.) and transformed into DH5.alpha.CaCl.sub.2 competent cells (Invitrogen, Carlsbad, Calif.). DNA was isolated from the resulting clones and diagnostic restriction digests followed by DNA sequencing confirmed the presence and sequence of the insert. The insert was cloned into the bacterial expression vector pET-30a (+) (Novagen, Madison, Wis.) using the EcoRI and HindIII restriction sites. Next, BL21 (DE3) competent cells were transformed and protein was expressed following the manufacturer's protocol (Novagen, Madison, Wis.).

Example 7

Purification of Bacterially Expressed MNV-1 Capsid Protein

[0052] Following a 2 hour expression, protein was purified from inclusion bodies of bacterial cells using the BugBuster protein extraction reagent (Novagen, Madison, Wis.). His-tagged capsid protein was isolated from remaining protein by nickel column chromatography (Ni--NTA his Bind Resin, Novagen, Madison, Wis.) in the presence of 8M urea and protease inhibitors (protease inhibitor cocktail set III, Novagen, Madison, Wis.). Samples were dialyzed against 25 mM phosphate buffer (pH 6.0) and the purity of each preparation was assessed by SDS-PAGE and silver staining (Silver stain Plus kit, Biorad, Hercules, Calif.).

Example 8

Generation of Antisera in Rabbits

[0053] Rabbit sera was produced through Cocalico Biologicals, Inc. (Reamstown, Pa.). Basically, rabbits were injected with 100 .mu.g bacterially expressed capsid protein in CFA (complete Freund's adjuvant) and boosted after a month once every month with 501 g protein in IFA (incomplete Freund's adjuvant). Production bleeds were collected a week after each boost and before the start of injections. The same procedure is being used for generation of antibodies directed against virus-like MNV-1 particles.

Example 9

Cloning and Expression of the MNV-1 Capsid Protein in Baculovirus

[0054] The MNV-1 capsid protein was cloned into the baculovirus expression vector in an analogue way to the cloning into the bacterial expression vector. The following primers were used for initial 2 step PCR amplification (4 cycles at 50 C.sub.1-30 cycles at 64 C) of the MNV-1 capsid protein:

[0055] N-Bac1 (CGGAATTCGGATGAGGATGAGTGATGGCGCA)(SEQ ID NO:9), C-Bac 1 (TCTCGACAAGCTTTTATTGTTTGAGCATTCGGCCT)(SEQ ID NO: 10). The same restriction sites, EcoRI and HindIII (underlined) were used for cloning into the pFastBac1 vector (Invitrogen, Carlsbad, Calif.). Recombinant baculoviruses were made using the Bac-to-Bac Expression system (Invitrogen, Carlsbad, Calif.) following the manufacturer's instructions. Briefly, the recombinant vector plasmid containing the MNV-1 capsid protein was transformed into DH10Bac E. coli cells allowing for transposition of the gene of interest into the bacmid genome. Clones containing recombinant bacmids were identified by antibiotic selection and disruption of the lacZ gene. Recombinant bacmid DNA was then used for transfection of Sf9 insect cells. Recombinant baculoviruses were amplified for several rounds on Sf9 or Sf21 cells (Invitrogen, Carlsbad, Calif.) before infection of High-Five cells (Invitrogen, Carlsbad, Calif.) for protein expression. High-Five cells were infected for 5-7 days and supernatant were collected for purification of MNV-1 VLPs. Initial preparations were screened for the presence of VLPs by negative staining electron microscopy. VLPs were identified in the supernatants of several isolates (FIG. 6C). Two isolates with the highest amount of protein expression were chosen for further experiments. The amount of protein expression in each preparation was analyzed by SDS-PAGE and immunoblotting (FIG. 6A).

Example 10

Purification of MNV-1 VLPs

[0056] MNV-1 VLPs are purified from the supernatant of infected High-Five cells 7 days post-infection (FIG. 6D). The purification protocol is based on Leite et al. (Arch Virol, 1996, 141:865-875), which is hereby incorporated by reference. Briefly, protein in the cell supernatant is being precipitated using PEG 8000, and particles are purified using CsCl gradients. VLPs are dialyzed against 25 mM phosphate buffer, pH 6.0. Details of the protocol are being optimized at this point.

Example 11

Use of VLPs, Potential and Actual Targets of VLPs

[0057] VLP-containing insect cell supernatants are being used for ELISA to screen mouse sera (see ELISA below). VLPs will be used to generate rabbit antisera. Their role as potential vaccine will be investigated. They will also be used for three-dimensional structure determination of the MNV-1 capsid.

Example 12

ELISA Assay

[0058] This assay can be used to screen mice capable of eliciting an antibody response (FIG. 7). The assay was optimized for a maximal signal-to-background ratio. VLP-containing insect cell supernatants are used as antigens for coating ELISA plates over night at 4 C. Plates are blocked for two hours at 37 C with 3% BSA and washed with 0.1 SM NaCl+0.05% Tween 20. Sera from mice are diluted 1:100 and incubated for 1 hour at 37 C. after washing, wells are incubated for two hours at 37 C with a 1:1000 dilution of peroxidase-conjugated AffiniPure goat anti-mouse IgG (H+L) (Jackson ImmunoResearch Laboratories, Inc., West Grove, Pa.). Plates are developed after another round of washing by addition of the substrate 2,2'-Azinobis 3-ethylbenzthiazoline sulfonic acid (ABTS, Sigma-Aldrich Corp., St. Louis, Mo.) for 10 min, the reaction is stopped using 0.2N phosphoric acid, and quantified by reading the absorbance at 415 nm.

Example 13

Nested PCR Assay

[0059] This assay can be used to screen tissues of immunocompromised mice with no antibody response (FIG. 10). RNA is isolated from the tissue(s) of interest and 1.sup.st strand cDNA is being made (see above). To sets of primers were designed with the following sequences: outer sense primer CCAAAAGCCAATGGCTCTGA (SEQ ID NO: 11), outer antisense primer AGTTGAATGGGCTCCAGGGT (SEQ ID NO: 12), inner sense primer CCGCCGGGCAAATTAACCAA (SEQ ID NO: 13), inner antisense primer AGGTGGGCAAGGTAGGGGTTA (SEQ ID NO: 14). Each reaction contained 2 .mu.l of first strand cDNA and a final concentration of 1 .mu.M sense and antisense primer, 2.5 mM MgCl.sub.2, 0.2 mM dNTPs, and 1.25 unit Taq DNA Polymerase (Promega, Madison, Wis.) in 1.times. buffer (Taq DNA Polymerase 10.times. Reaction Buffer without MgCl.sub.2, Promega, Madison, Wis.). PCR was performed for 45 cycles for the 1 round, and 30 cycles for the 2.sup.nd round with the following setting: heating 2 min 94 C, cycle for 30 sec 94 C, 30 sec 60 C (annealing), and 30 sec 72 C (extension), with a final extension step of 10 min 72 C. Two .mu.l product from the 1.sup.st round are used in the 2.sup.nd round using the same overall set-up. Products are analyzed by agarose gel electrophoresis.

Example 14

Use of MNV-1 Infected Mice as Small Animal Model of Norovirus Infection

[0060] To determine whether T and B cell mediated immunity is required for resistance to MNV-1, wild-type and RAG1-/- mice were infected by the i.c. route and followed for 90 days (data not shown). Surprisingly, MNV-1 infection does not kill RAG1-/- mice (n=20) after direct i.c. inoculation even though these mice are typically highly susceptible to infection with a range of different viruses. The finding that RAG-/- mice are resistant to lethal disease argues that typical adaptive responses are not required for protection from lethal disease. This finding may explain in part contradictory conclusions as to the importance of antibody in resistance to Norovirus disease. While B and T cell responses are not required for resistance to lethal infection, it may be that pre-existing immunity influences the pathogenicity of MNV-1. Alternatively, the presence of immune cells may contribute to disease induced by MNV-1 as is seen for lymphocytic choriomeningitis virus.

[0061] Together with a course of clinical illness too brief to allow typical adaptive responses, these studies in RAG-/- mice beg the question of whether innate rather than acquired immunity is critical for resistance to calicivirus infection. We therefore inoculated a variety of mouse strains lacking components of the innate immune system with MNV-1. The peroral (p.o.) and intranasal (i.n.) routes were tested in addition to the i.c. route since the physiologic routes of infection for human caliciviruses are oral and respiratory. Mice lacking the IFN.alpha..beta. receptor or the IFN.gamma. receptor were no more susceptible to lethal infection than wild-type controls (FIG. 5A). In contrast, mice lacking both IFN.alpha..beta. and IFN.gamma. receptors were more susceptible to lethal infection than congenic controls after either i.c. or i.n. inoculation with MNV-1 (FIG. 5B). These data demonstrate that the IFN receptors can compensate for each other in resistance to MNV-1 infection such that only deficiency in both receptors leads to lethality. Mice deficient in inducible nitric oxide synthetase (iNOS) or in the RNA activated protein kinase PKR, two IFN regulated proteins with antiviral properties, were also resistant to lethal MNV-1 infection after i.c. or p.o. inoculation (FIG. 5A).

[0062] Since deficiency in both IFN receptors is required to predispose to lethal MNV-1 infection, we reasoned that a component of the innate immune system that can be activated by either the IFN.alpha..beta. or the IFN.gamma. receptor was critical for MNV-1 survival. We therefore tested the hypothesis that the latent cytoplasmic transcription factor STAT1, which is shared by both the IFN.alpha..beta. and IFN.gamma. signaling pathways, was critical for resistance to calicivirus infection. STAT1 deficiency resulted in lethal MNV-1 infection in mice with T and B cells (STAT1-/-, FIG. 5C), mice lacking T and B cells (RAG/STAT, FIG. 5D), and mice lacking PKR (PKR-/-STAT1-/-, FIG. 5E) by all routes analyzed. Thus STAT1 is the first host component identified as essential for resistance to lethal Norovirus infection, and is required for survival even when T and B cells are present.

[0063] Having identified STAT1 as essential for calicivirus resistance, we then investigated the relationship between the interferon receptors and STAT1. No statistically significant differences were found in the survival of IFN.alpha..beta..gamma.R-/- and STAT1-/- mice after either i.c. or i.n. inoculations. However after p.o. inoculation, deficiency of STAT1, but not deficiency in both IFN.alpha..beta. and IFN.gamma. receptors, led to lethal infection (see FIGS. 5B and C). This might suggest that STAT1 has IFN receptor-independent effects that are critical for Norovirus resistance. Supporting this are findings that the biological effects of STAT1 do not completely overlap with those of the IFN receptors during viral infection since there are both STAT1-independent antiviral effects of the IFN receptors, and IFN receptor-independent effects of STAT1.

Sequence CWU 1

1

54 1 7726 DNA Murine Norovirus type 1 misc_feature (147)..(5024) ORF1 1 gtgaattcta gaaggcaacg ccatcttctg cgccctctgt gcgcaacaca gagaaacgca 60 aaaacaagaa ggcttcgyct aaagctagtg tctcctttgg agcacctagc cccctctctt 120 cggagagcga agacgaartt aattacatga cccctcctga gcaggaagct cagcccggcg 180 cccttgcggc ccttcatgcg gaagggccgc ttgccgggct ccccgtgacg cgtagtgatg 240 cacgcgtgct gatcttcaat gagtgggagg agaggaagaa gtctgatccg tggctacggc 300 tggacatgtc tgataaggct atcttccgcc gttaccccca tctgcggcct aaggaggata 360 ggcctgacgc gccctcccat gcggaggacg ctatggatgc caaggagcct gtgatcggct 420 ctatcttgga gcaggatgat cacaagtttt accattactc tgtctacatc ggtggcggcc 480 ttgtgatggg ggtcaacaac cccagtgctg cggtctgcca ggcaacgatt gatgtggaga 540 agctacacct ctggtggcgg cctgtctggg agccccgcca wccccttgac tcggctgagt 600 tgaggaagtg cgtgggcatg actgtcccct acgtggccac caccgtcaac tgttatcagg 660 tctgctgctg gattgttggc atcaaggaca cctggctgaa gagggcgaag atctctagag 720 atctgccctt ctacagcccc gtccaggact ggaacgtcga cccccaggag cccttcattc 780 catccaagct caggatggtc tcggatggca tcctggtggc cttgtcggca gtgattggcc 840 ggccaattaa gaacctactg gcctcagtta agccgctcaa cattctcaac atcgtgctga 900 gctgtgattg gaccttttcg ggcattgtca atgccctgat cttgcttgct gagctctttg 960 acatcttttg gaccccccct gatgtracca rctggatgat ctctatcttc ggggaatggc 1020 aggccgaagg gcccttcgac cytgctcttg acgtggtgcc caccctgttg ggcgggatcg 1080 ggatggcttt tggcctcrcc tctgagacca tcgggcgcaa gctcdcttcc accaactcgg 1140 ctctcaaggc cgcccaagag atgggcaagt tcgccataga ggtcttcaag caaattatgg 1200 cctggatctg gccctctgag gacccagtgc cagccctctt atccaacatg gagcaggcca 1260 tcattaagaa tgagtgtcaa ctdgagaacc aactcacggc catgttgcgg gatcgcaacg 1320 caggggctga attcctvagg tcccttgatg aggaggagca ggaagtccgc aagatcgcag 1380 ctaagtgcgg caactcggcc accactggaa ccaccaacgc tctgctggcc aggatcagca 1440 tggcccgcgc ggcctttgag aaagctcgcg ctgaacagac ctcccgagtc cgccctgtgg 1500 tgdtcatggt ctcaggcagg cccgggatcg ggaaaacctg cttttgccaa aacctagcca 1560 agaggattgc tgcgtccctg ggtgatgaga cctctgttgg catcatacca cgcgctgatg 1620 tcgaccactg ggatgcttac aagggagcca gagtggttct ctgggatgat ttcggcatgg 1680 acaacgtggt gaaggatgca ctgaggcttc agatgcttgc cgacacgtgc ccagtgacac 1740 tcaattgtga caggattgag aacaagggaa agatgyttga ctctcaggtc attatcatca 1800 ccacaaatca acaaaccccc gygcccctgg actatgtcaa cctggaggcg gtctgccgcc 1860 gcatagattt cctggtttat gmtgagagcc ctgttgttga tgatgctcgg gccagagccc 1920 ctggcgatgt gaatgcagtg aaagctgcca tgaggcccga ttacagccac atcaatttca 1980 tcttggcacc gcagggcggc tttgaccgtc gggaaacacc ccctacggta agggcgtcac 2040 caagatcatt ggcgccactg ctctttgcgc gagagcggtt gctcttgtcc atgagcgcca 2100 tgatgatttc ggcctccaga acaaggtcya tgactttgat gcgcgcaarg tcaccgcctt 2160 caaagccatg gcggctgacg ccggcattcc atggtacaaa atggcagcta ttgggtgcaa 2220 agcaatgggg gtgcacctgt gtagaggagg ccatgcattt acttaaggat tatgaggtgg 2280 ctccctgtca ggtgatctac aatggtgcca cctataatgt gagctgcatc aagggtgccc 2340 caatggttga aaaggtcaag gagcctgaat tgcccaaaac acttgtcaac tgtgtcagaa 2400 ggataaagga ggcccgcctc cgctgctact gtaggatggc tgctgacgtc atcacgtcca 2460 ttctgcaggc ggccggcacg gccttctcta tttaccacca gattgagaag aggtctagac 2520 catcctttta ttgggatcat ggatacacct accgtgacgg acctggatcc tttgacatct 2580 ttgaggatga cgatgatggg tggtaccact ctgagggaaa gaagggcaag aacaagaagg 2640 gccgggggcg acccggagtc ttcagaaccc gtgggctcac ggatgaggag tacgatgaat 2700 tcaagaagcg ccgcgagtct aggggcggca agtactccat tgatgattac ctcgctgrcc 2760 gcgagcgaga agaagaactc ctggagcggg acgaggagga ggctatcttc ggggayggct 2820 tcgggttgaa ggccacccgc cgttcccgca aggcagagag agccaaactg ggcctggttt 2880 ctggtggcga catccgcgcc cgcaagccga tcgactggaa tgtggttggc ccctcctggg 2940 ctgacgatga ccgccaggtc gctacggcga gaagatcaac tttgaggccc cagtytccat 3000 ctggtcccgt gttgtgcagt tcggcacggg gtggggcttt tggggtgagc ggccacgtct 3060 tcatcaccgc caagcatgtg gcgcccccca agggcacgga gatctttggg cgcaagcccg 3120 gggacttcac tgtcrcttcc agcggggact tcttgaagta ctacttcacc agcgccgtca 3180 ggcctgacrt tcccgccatg gtcctggaga atgggtgcca ggagggcgtc gtcgcctcgg 3240 tccttgtcaa gagagcctcc ggcgagatgc ttgccctggc tgtcaggatg ggttcacagg 3300 ccgccatcaa gattggtagt gccgttgtgc atgggcaaac tggcatgctc ctgactggct 3360 ctaatgccaa ggcccaggac ctcgggacca tcccgggcga ctgtggctgt ccctatgttt 3420 ataagaaggg taacacctgg gttgtgattg gggtgcacgt ggcggccact aggtctggta 3480 acacagtcat tgccgccact cacggagaac ccacacttga ggctctggag ttccagggac 3540 cccccatgct tccccgcccc tcaggcacct atgcaggcct ccccatcgcc gattacggcg 3600 acgctccccc cttgagcacc aagaccatgt tctggcgtac ctcgccagag aagcttcccc 3660 ctggggcttg ggagccagcc tatctcggct ctaaagatga gagggtggac ggtccttccc 3720 ttcagcaggt catgcgagat cagcttaagc cctattcaga accacgcggt ctgcttcccc 3780 ctcaagaaat ccttgatgca gtctgcgacg ccattgagaa ccgccttgag aacacccttg 3840 aaccacagaa gccctggaca tttaagaagg cttgtgagag cttggacaag aacaccagya 3900 gygggtatcc ctatcacaag cagaagagca aggactggac ggggagcgct tttattggcg 3960 rtcttggtga ccaggccacc cacgccaaca acatgtatga gatgggtaaa tccatgcgac 4020 ccatttatac agctgccctc aaggatgaac tggttaagcc agacaagatc tacgggaaga 4080 taaagaagag gcttctctgg ggctctgacc ttgrcaccat gattcgcgct gcccgtgcyt 4140 ttggcccttt ctgtgatgct ctgaaagaar cctgcatttt caaccccatc agagtgggca 4200 tgtcgatgaa cgaagatggc cccttcatct tcgcaagaca cgccaatttc aggtaccaca 4260 tggatgctga ctataccagg tgggactcca cccaacagag agccatccta aagcgcgctg 4320 gygacatcat ggygcgcctc tcccctgagc cagacttggc tcgggttgtc atggatgatc 4380 tcctggcccc ctcgctgttg gacgtcggcg actrtaagat cgttgtcgag gaggggctcc 4440 catccggctg cccttgcacc acacagctga atagtttggc tcactggatt ttgacccttt 4500 gtgcaatggt tgaggtaacc cgagttgacc ctgacattgt gatgcaagaa tctgagttyt 4560 ccttctatgg tgatgacgag gtggtttcga ccaacctcga gttggatatg gttaagtaca 4620 ccatggcttt gaggcggtac ggtctcctcc cgactcgcgc ggacaaggag gagggacctc 4680 tggagcgtcg ccagacgctg cagggcatct ccttcctgcg ccgtgcgata gttggtgacc 4740 agtttgggtg gtacggtcgt cttgatcgtg ccagcatcga ccgccagctc ctctggacta 4800 aaggacctaa ccaccagaac ccctttgaga ctctccctgg acatgctcag agaccctccc 4860 aactaatggc cctgctcggt gaggctgcca tgcatggtga aaagtattac aggactgtgg 4920 cttcccgtgt ctccaaggag gccgcccaaa gtgggatara aatggtagtc cccacgccac 4980 cgatctgttt tgcgctgggt gcgctttgga acaatggatg ctgagacccc gcaggaacgc 5040 tcagcagtct ttgtgaatga ggatgagtga tggcgcagcg ccaaaagcca atggctctga 5100 ggccagcggc caggatcttg ttcctgccgc cgttgaacag gccgtcccca ytcaacccgt 5160 ggctggcgcg gctcttgccg cccccgccgc cgggcaaatt aaccaaattg rcccctggat 5220 cttccaaaat tttgtccagt gcccccttgg tgagttttcc atttcgcctc gaaacacccc 5280 aggtgaaata ctgtttgatt tggccctcgg gccagggctt aacccctacc ttgcccacct 5340 ctcagccatg tacaccggct gggttgggaa crtggaggtt cagctggtcc tcgccggcaa 5400 tgcctttact gctggcaagg tggttgttgc ccttgtacca ccctattttc ccaaggggtc 5460 actcaccact gcccagatca catgcttccc acatgtcatg tgtgatgtgc gcaccctgga 5520 gcccattcaa ctccctcttc ttgatgtgcg tcgagtcctt tggcatgcta cccaggatca 5580 agaggaatct atgcgcctgg tttgcatgct gtacacgcca ctccgcacaa acagcccggg 5640 tgatgagtct tttgtggtct ctggccgcct tctttctaag ccggcggctg atttcaattt 5700 tgtctaccta actcccccca tagagagaac catctaccgg atggtcgact tgcccgtgat 5760 acagccgcgg ctgtgcacgc acgcacgttg gcctgccccg gtctatggtc tcttggtgga 5820 cccatccctc ccctcaaatc cccagtggca gaatggaagg gtgcacgttg atgggaccct 5880 gcttggtacc accccaatct ccggttcatg ggtgtcctgc tttgcgkcgg aggctgccta 5940 taagttccaa tcgggcaccg gtgaggtggc gacattcacc ctgattgagc aggatggatc 6000 tgcctacgtc cccggtgaca gggcagcacc actcgggtta ccccgatttc tctgggcaac 6060 tggagatcga ggtccagacc gagaccacca agactggaga caagctcaag gtcaccactt 6120 tgagatgatt cttggcccaa cgaccaacgc ggaccaggcc ccctaccagg gcagggtgtt 6180 cgccagcgtc actgctgcgg cctctcttga cttggtggat ggcagggttc gtgcggtccc 6240 aagatccatc tacggttttc aggacaccat ccctgaatac aacgatgggc tactggttcc 6300 ccttgccccc ccaattggtc cctttctccc cggcgaggtc ctcctgaggt tccggaccta 6360 catgcgtcag atcgacaccg ctgacgccgc agcagaggcg atagactgtg cactccccca 6420 ggagtttgtc tcctggttcg cgtctaacgc gttcaccgtg cagtccgagg ccctgctcct 6480 tagatacagg aacaccctga ctgggcaact gctgttcgag tgcaagctct acaacgaagg 6540 ttacatcgcc ttgtcttatt ccggctcagg acccctcacc ttcccgaccg atggcatctt 6600 tgaggtcgtc agttgggttc ctcgccttta ccaattggcc tctgtgggaa gtttggcaac 6660 aggccgaatg ctcaaacaat aatggctggt gctctttttg gagcgattgg aggtggcctg 6720 atgggcataa ttggcaattc catctcaaat gttcaaaacc ttcaggcaaa caaacaattg 6780 gcagctcagc aatttggtta taattcttcc ctgcttgcaa cgcaaattca agcccagaag 6840 gatctcactc tgatggggca gcaattcaac cagcagctcc aaaccaactc tttcaagcac 6900 gacttggaaa tgcttggcgc tcaggtgcaa gcccaggcgc aggcccagga gaacgccatc 6960 aatatcaaaa cggcgcagct ccaggccgca ggcttttcaa agacagatgc cacacgcctt 7020 gccttggggc agcagcccac gagggccgtg gattggtctg ggacgcggta ctacaccgct 7080 aaccagccag tcacgggctt ctcgggtggc tttaccccaa cctacactcc aggtaggcaa 7140 gtgacatccc gccctgtgga cacatcccct ctaccgatct cgggtggacg cttgccctcc 7200 cttcgtggag gttcctggtc cccgcgcgac catacgccgg cgactcaagg cacctacacg 7260 aacggacggt tcgtgtctct ccctaagatc gggagtagca gggcataggt tggaagagaa 7320 accttttgtg aaaatgattt ctgcttactg ctttcttttc tttgtggtag ttagatgcat 7380 ttcgagggcc gtggattggt ctgggacgcg gtactacacc gctaaccagc cagtcacggg 7440 cttctcgggt ggctttaccc caacctacac tccaggtagg caagtgacat cccgccctgt 7500 ggacacatcc cctctaccga tctcgggtgg acgcttgccc tcccttcgtg gaggttcctg 7560 gtccccgcgc gaccatacgc cggcgactca aggcacctac acgaacggac ggttcgtgtc 7620 tctccctaag atcgggagta gcagggcata ggttggaaga gaaacctttt gtgaaaatga 7680 tttctgctta ctgctttctt ttctttgtgg tagttagatg catttc 7726 2 1625 PRT Murine Norovirus type 1 misc_feature (145)..(145) Variable amino acid 2 Met Thr Pro Pro Glu Gln Glu Ala Gln Pro Gly Ala Leu Ala Ala Leu 1 5 10 15 His Ala Glu Gly Pro Leu Ala Gly Leu Pro Val Thr Arg Ser Asp Ala 20 25 30 Arg Val Leu Ile Phe Asn Glu Trp Glu Glu Arg Lys Lys Ser Asp Pro 35 40 45 Trp Leu Arg Leu Asp Met Ser Asp Lys Ala Ile Phe Arg Arg Tyr Pro 50 55 60 His Leu Arg Pro Lys Glu Asp Arg Pro Asp Ala Pro Ser His Ala Glu 65 70 75 80 Asp Ala Met Asp Ala Lys Glu Pro Val Ile Gly Ser Ile Leu Glu Gln 85 90 95 Asp Asp His Lys Phe Tyr His Tyr Ser Val Tyr Ile Gly Gly Gly Leu 100 105 110 Val Met Gly Val Asn Asn Pro Ser Ala Ala Val Cys Gln Ala Thr Ile 115 120 125 Asp Val Glu Lys Leu His Leu Trp Trp Arg Pro Val Trp Glu Pro Arg 130 135 140 Xaa Pro Leu Asp Ser Ala Glu Leu Arg Lys Cys Val Gly Met Thr Val 145 150 155 160 Pro Tyr Val Ala Thr Thr Val Asn Cys Tyr Gln Val Cys Cys Trp Ile 165 170 175 Val Gly Ile Lys Asp Thr Trp Leu Lys Arg Ala Lys Ile Ser Arg Asp 180 185 190 Leu Pro Phe Tyr Ser Pro Val Gln Asp Trp Asn Val Asp Pro Gln Glu 195 200 205 Pro Phe Ile Pro Ser Lys Leu Arg Met Val Ser Asp Gly Ile Leu Val 210 215 220 Ala Leu Ser Ala Val Ile Gly Arg Pro Ile Lys Asn Leu Leu Ala Ser 225 230 235 240 Val Lys Pro Leu Asn Ile Leu Asn Ile Val Leu Ser Cys Asp Trp Thr 245 250 255 Phe Ser Gly Ile Val Asn Ala Leu Ile Leu Leu Ala Glu Leu Phe Asp 260 265 270 Ile Phe Trp Thr Pro Pro Asp Val Thr Xaa Trp Met Ile Ser Ile Phe 275 280 285 Gly Glu Trp Gln Ala Glu Gly Pro Phe Asp Xaa Ala Leu Asp Val Val 290 295 300 Pro Thr Leu Leu Gly Gly Ile Gly Met Ala Phe Gly Leu Xaa Ser Glu 305 310 315 320 Thr Ile Gly Arg Lys Leu Xaa Ser Thr Asn Ser Ala Leu Lys Ala Ala 325 330 335 Gln Glu Met Gly Lys Phe Ala Ile Glu Val Phe Lys Gln Ile Met Ala 340 345 350 Trp Ile Trp Pro Ser Glu Asp Pro Val Pro Ala Leu Leu Ser Asn Met 355 360 365 Glu Gln Ala Ile Ile Lys Asn Glu Cys Gln Leu Glu Asn Gln Leu Thr 370 375 380 Ala Met Leu Arg Asp Arg Asn Ala Gly Ala Glu Phe Leu Arg Ser Leu 385 390 395 400 Asp Glu Glu Glu Gln Glu Val Arg Lys Ile Ala Ala Lys Cys Gly Asn 405 410 415 Ser Ala Thr Thr Gly Thr Thr Asn Ala Leu Leu Ala Arg Ile Ser Met 420 425 430 Ala Arg Ala Ala Phe Glu Lys Ala Arg Ala Glu Gln Thr Ser Arg Val 435 440 445 Arg Pro Val Val Xaa Met Val Ser Gly Arg Pro Gly Ile Gly Lys Thr 450 455 460 Cys Phe Cys Gln Asn Leu Ala Lys Arg Ile Ala Ala Ser Leu Gly Asp 465 470 475 480 Glu Thr Ser Val Gly Ile Ile Pro Arg Ala Asp Val Asp His Trp Asp 485 490 495 Ala Tyr Lys Gly Ala Arg Val Val Leu Trp Asp Asp Phe Gly Met Asp 500 505 510 Asn Val Val Lys Asp Ala Leu Arg Leu Gln Met Leu Ala Asp Thr Cys 515 520 525 Pro Val Thr Leu Asn Cys Asp Arg Ile Glu Asn Lys Gly Lys Met Xaa 530 535 540 Asp Ser Gln Val Ile Ile Ile Thr Thr Asn Gln Gln Thr Pro Xaa Pro 545 550 555 560 Leu Asp Tyr Val Asn Leu Glu Ala Val Cys Arg Arg Ile Asp Phe Leu 565 570 575 Val Tyr Xaa Glu Ser Pro Val Val Asp Asp Ala Arg Ala Arg Ala Pro 580 585 590 Gly Asp Val Asn Ala Val Lys Ala Ala Met Arg Pro Asp Tyr Ser His 595 600 605 Ile Asn Phe Ile Leu Ala Pro Gln Gly Gly Phe Asp Arg Arg Glu Thr 610 615 620 Pro Pro Thr Val Arg Ala Ser Pro Arg Ser Leu Ala Pro Leu Leu Phe 625 630 635 640 Ala Arg Glu Arg Leu Leu Leu Ser Met Ser Ala Met Met Ile Ser Ala 645 650 655 Ser Arg Thr Arg Ser Met Thr Leu Met Arg Ala Xaa Ser Pro Pro Ser 660 665 670 Lys Pro Trp Arg Leu Thr Pro Ala Phe His Gly Thr Lys Trp Gln Leu 675 680 685 Leu Gly Ala Lys Gln Trp Gly Cys Thr Cys Val Glu Glu Ala Met His 690 695 700 Leu Leu Lys Asp Tyr Glu Val Ala Pro Cys Gln Val Ile Tyr Asn Gly 705 710 715 720 Ala Thr Tyr Asn Val Ser Cys Ile Lys Gly Ala Pro Met Val Glu Lys 725 730 735 Val Lys Glu Pro Glu Leu Pro Lys Thr Leu Val Asn Cys Val Arg Arg 740 745 750 Ile Lys Glu Ala Arg Leu Arg Cys Tyr Cys Arg Met Ala Ala Asp Val 755 760 765 Ile Thr Ser Ile Leu Gln Ala Ala Gly Thr Ala Phe Ser Ile Tyr His 770 775 780 Gln Ile Glu Lys Arg Ser Arg Pro Ser Phe Tyr Trp Asp His Gly Tyr 785 790 795 800 Thr Tyr Arg Asp Gly Pro Gly Ser Phe Asp Ile Phe Glu Asp Asp Asp 805 810 815 Asp Gly Trp Tyr His Ser Glu Gly Lys Lys Gly Lys Asn Lys Lys Gly 820 825 830 Arg Gly Arg Pro Gly Val Phe Arg Thr Arg Gly Leu Thr Asp Glu Glu 835 840 845 Tyr Asp Glu Phe Lys Lys Arg Arg Glu Ser Arg Gly Gly Lys Tyr Ser 850 855 860 Ile Asp Asp Tyr Leu Ala Xaa Arg Glu Arg Glu Glu Glu Leu Leu Glu 865 870 875 880 Arg Asp Glu Glu Glu Ala Ile Phe Gly Asp Gly Phe Gly Leu Lys Ala 885 890 895 Thr Arg Arg Ser Arg Lys Ala Glu Arg Ala Lys Leu Gly Leu Val Ser 900 905 910 Gly Gly Asp Ile Arg Ala Arg Lys Pro Ile Asp Trp Asn Val Val Gly 915 920 925 Pro Ser Trp Ala Asp Asp Asp Arg Gln Val Ala Thr Ala Arg Arg Ser 930 935 940 Thr Leu Arg Pro Gln Xaa Pro Ser Gly Pro Val Leu Cys Ser Ser Ala 945 950 955 960 Arg Gly Gly Ala Phe Gly Val Ser Gly His Val Phe Ile Thr Ala Lys 965 970 975 His Val Ala Pro Pro Lys Gly Thr Glu Ile Phe Gly Arg Lys Pro Gly 980 985 990 Asp Phe Thr Val Xaa Ser Ser Gly Asp Phe Leu Lys Tyr Tyr Phe Thr 995 1000 1005 Ser Ala Val Arg Pro Asp Xaa Pro Ala Met Val Leu Glu Asn Gly 1010 1015 1020 Cys Gln Glu Gly Val Val Ala Ser Val Leu Val Lys Arg Ala Ser 1025 1030 1035 Gly Glu Met Leu Ala Leu Ala Val Arg Met Gly Ser Gln Ala Ala 1040 1045 1050 Ile Lys Ile Gly Ser Ala Val Val His Gly Gln Thr Gly Met Leu 1055 1060 1065 Leu Thr Gly Ser Asn Ala Lys Ala Gln Asp Leu Gly Thr Ile Pro 1070 1075 1080 Gly Asp Cys Gly Cys Pro Tyr Val Tyr Lys Lys Gly Asn Thr Trp 1085 1090 1095 Val Val Ile Gly Val His Val Ala Ala Thr Arg Ser Gly Asn Thr 1100 1105 1110 Val Ile Ala Ala Thr His Gly Glu Pro Thr Leu Glu Ala Leu Glu 1115 1120 1125 Phe Gln Gly Pro Pro Met Leu Pro Arg Pro Ser Gly Thr Tyr Ala 1130 1135 1140 Gly Leu Pro Ile Ala Asp Tyr Gly Asp Ala Pro Pro Leu Ser Thr 1145 1150 1155 Lys Thr Met Phe Trp Arg Thr Ser Pro Glu Lys Leu Pro Pro Gly

1160 1165 1170 Ala Trp Glu Pro Ala Tyr Leu Gly Ser Lys Asp Glu Arg Val Asp 1175 1180 1185 Gly Pro Ser Leu Gln Gln Val Met Arg Asp Gln Leu Lys Pro Tyr 1190 1195 1200 Ser Glu Pro Arg Gly Leu Leu Pro Pro Gln Glu Ile Leu Asp Ala 1205 1210 1215 Val Cys Asp Ala Ile Glu Asn Arg Leu Glu Asn Thr Leu Glu Pro 1220 1225 1230 Gln Lys Pro Trp Thr Phe Lys Lys Ala Cys Glu Ser Leu Asp Lys 1235 1240 1245 Asn Thr Ser Ser Gly Tyr Pro Tyr His Lys Gln Lys Ser Lys Asp 1250 1255 1260 Trp Thr Gly Ser Ala Phe Ile Gly Xaa Leu Gly Asp Gln Ala Thr 1265 1270 1275 His Ala Asn Asn Met Tyr Glu Met Gly Lys Ser Met Arg Pro Ile 1280 1285 1290 Tyr Thr Ala Ala Leu Lys Asp Glu Leu Val Lys Pro Asp Lys Ile 1295 1300 1305 Tyr Gly Lys Ile Lys Lys Arg Leu Leu Trp Gly Ser Asp Leu Xaa 1310 1315 1320 Thr Met Ile Arg Ala Ala Arg Ala Phe Gly Pro Phe Cys Asp Ala 1325 1330 1335 Leu Lys Glu Xaa Cys Ile Phe Asn Pro Ile Arg Val Gly Met Ser 1340 1345 1350 Met Asn Glu Asp Gly Pro Phe Ile Phe Ala Arg His Ala Asn Phe 1355 1360 1365 Arg Tyr His Met Asp Ala Asp Tyr Thr Arg Trp Asp Ser Thr Gln 1370 1375 1380 Gln Arg Ala Ile Leu Lys Arg Ala Gly Asp Ile Met Xaa Arg Leu 1385 1390 1395 Ser Pro Glu Pro Asp Leu Ala Arg Val Val Met Asp Asp Leu Leu 1400 1405 1410 Ala Pro Ser Leu Leu Asp Val Gly Asp Xaa Lys Ile Val Val Glu 1415 1420 1425 Glu Gly Leu Pro Ser Gly Cys Pro Cys Thr Thr Gln Leu Asn Ser 1430 1435 1440 Leu Ala His Trp Ile Leu Thr Leu Cys Ala Met Val Glu Val Thr 1445 1450 1455 Arg Val Asp Pro Asp Ile Val Met Gln Glu Ser Glu Phe Ser Phe 1460 1465 1470 Tyr Gly Asp Asp Glu Val Val Ser Thr Asn Leu Glu Leu Asp Met 1475 1480 1485 Val Lys Tyr Thr Met Ala Leu Arg Arg Tyr Gly Leu Leu Pro Thr 1490 1495 1500 Arg Ala Asp Lys Glu Glu Gly Pro Leu Glu Arg Arg Gln Thr Leu 1505 1510 1515 Gln Gly Ile Ser Phe Leu Arg Arg Ala Ile Val Gly Asp Gln Phe 1520 1525 1530 Gly Trp Tyr Gly Arg Leu Asp Arg Ala Ser Ile Asp Arg Gln Leu 1535 1540 1545 Leu Trp Thr Lys Gly Pro Asn His Gln Asn Pro Phe Glu Thr Leu 1550 1555 1560 Pro Gly His Ala Gln Arg Pro Ser Gln Leu Met Ala Leu Leu Gly 1565 1570 1575 Glu Ala Ala Met His Gly Glu Lys Tyr Tyr Arg Thr Val Ala Ser 1580 1585 1590 Arg Val Ser Lys Glu Ala Ala Gln Ser Gly Ile Xaa Met Val Val 1595 1600 1605 Pro Thr Pro Pro Ile Cys Phe Ala Leu Gly Ala Leu Trp Asn Asn 1610 1615 1620 Gly Cys 1625 3 541 PRT Murine Norovirus type 1 misc_feature (32)..(32) Variable amino acid 3 Met Arg Met Ser Asp Gly Ala Ala Pro Lys Ala Asn Gly Ser Glu Ala 1 5 10 15 Ser Gly Gln Asp Leu Val Pro Ala Ala Val Glu Gln Ala Val Pro Xaa 20 25 30 Gln Pro Val Ala Gly Ala Ala Leu Ala Ala Pro Ala Ala Gly Gln Ile 35 40 45 Asn Gln Ile Xaa Pro Trp Ile Phe Gln Asn Phe Val Gln Cys Pro Leu 50 55 60 Gly Glu Phe Ser Ile Ser Pro Arg Asn Thr Pro Gly Glu Ile Leu Phe 65 70 75 80 Asp Leu Ala Leu Gly Pro Gly Leu Asn Pro Tyr Leu Ala His Leu Ser 85 90 95 Ala Met Tyr Thr Gly Trp Val Gly Asn Xaa Glu Val Gln Leu Val Leu 100 105 110 Ala Gly Asn Ala Phe Thr Ala Gly Lys Val Val Val Ala Leu Val Pro 115 120 125 Pro Tyr Phe Pro Lys Gly Ser Leu Thr Thr Ala Gln Ile Thr Cys Phe 130 135 140 Pro His Val Met Cys Asp Val Arg Thr Leu Glu Pro Ile Gln Leu Pro 145 150 155 160 Leu Leu Asp Val Arg Arg Val Leu Trp His Ala Thr Gln Asp Gln Glu 165 170 175 Glu Ser Met Arg Leu Val Cys Met Leu Tyr Thr Pro Leu Arg Thr Asn 180 185 190 Ser Pro Gly Asp Glu Ser Phe Val Val Ser Gly Arg Leu Leu Ser Lys 195 200 205 Pro Ala Ala Asp Phe Asn Phe Val Tyr Leu Thr Pro Pro Ile Glu Arg 210 215 220 Thr Ile Tyr Arg Met Val Asp Leu Pro Val Ile Gln Pro Arg Leu Cys 225 230 235 240 Thr His Ala Arg Trp Pro Ala Pro Val Tyr Gly Leu Leu Val Asp Pro 245 250 255 Ser Leu Pro Ser Asn Pro Gln Trp Gln Asn Gly Arg Val His Val Asp 260 265 270 Gly Thr Leu Leu Gly Thr Thr Pro Ile Ser Gly Ser Trp Val Ser Cys 275 280 285 Phe Ala Xaa Glu Ala Ala Tyr Lys Phe Gln Ser Gly Thr Gly Glu Val 290 295 300 Ala Thr Phe Thr Leu Ile Glu Gln Asp Gly Ser Ala Tyr Val Pro Gly 305 310 315 320 Asp Arg Ala Ala Pro Leu Gly Leu Pro Arg Phe Leu Trp Ala Thr Gly 325 330 335 Asp Arg Gly Pro Asp Arg Asp His Gln Asp Trp Arg Gln Ala Gln Gly 340 345 350 His His Phe Glu Met Ile Leu Gly Pro Thr Thr Asn Ala Asp Gln Ala 355 360 365 Pro Tyr Gln Gly Arg Val Phe Ala Ser Val Thr Ala Ala Ala Ser Leu 370 375 380 Asp Leu Val Asp Gly Arg Val Arg Ala Val Pro Arg Ser Ile Tyr Gly 385 390 395 400 Phe Gln Asp Thr Ile Pro Glu Tyr Asn Asp Gly Leu Leu Val Pro Leu 405 410 415 Ala Pro Pro Ile Gly Pro Phe Leu Pro Gly Glu Val Leu Leu Arg Phe 420 425 430 Arg Thr Tyr Met Arg Gln Ile Asp Thr Ala Asp Ala Ala Ala Glu Ala 435 440 445 Ile Asp Cys Ala Leu Pro Gln Glu Phe Val Ser Trp Phe Ala Ser Asn 450 455 460 Ala Phe Thr Val Gln Ser Glu Ala Leu Leu Leu Arg Tyr Arg Asn Thr 465 470 475 480 Leu Thr Gly Gln Leu Leu Phe Glu Cys Lys Leu Tyr Asn Glu Gly Tyr 485 490 495 Ile Ala Leu Ser Tyr Ser Gly Ser Gly Pro Leu Thr Phe Pro Thr Asp 500 505 510 Gly Ile Phe Glu Val Val Ser Trp Val Pro Arg Leu Tyr Gln Leu Ala 515 520 525 Ser Val Gly Ser Leu Ala Thr Gly Arg Met Leu Lys Gln 530 535 540 4 208 PRT Murine Norovirus type 1 4 Met Ala Gly Ala Leu Phe Gly Ala Ile Gly Gly Gly Leu Met Gly Ile 1 5 10 15 Ile Gly Asn Ser Ile Ser Asn Val Gln Asn Leu Gln Ala Asn Lys Gln 20 25 30 Leu Ala Ala Gln Gln Phe Gly Tyr Asn Ser Ser Leu Leu Ala Thr Gln 35 40 45 Ile Gln Ala Gln Lys Asp Leu Thr Leu Met Gly Gln Gln Phe Asn Gln 50 55 60 Gln Leu Gln Thr Asn Ser Phe Lys His Asp Leu Glu Met Leu Gly Ala 65 70 75 80 Gln Val Gln Ala Gln Ala Gln Ala Gln Glu Asn Ala Ile Asn Ile Lys 85 90 95 Thr Ala Gln Leu Gln Ala Ala Gly Phe Ser Lys Thr Asp Ala Thr Arg 100 105 110 Leu Ala Leu Gly Gln Gln Pro Thr Arg Ala Val Asp Trp Ser Gly Thr 115 120 125 Arg Tyr Tyr Thr Ala Asn Gln Pro Val Thr Gly Phe Ser Gly Gly Phe 130 135 140 Thr Pro Thr Tyr Thr Pro Gly Arg Gln Val Thr Ser Arg Pro Val Asp 145 150 155 160 Thr Ser Pro Leu Pro Ile Ser Gly Gly Arg Leu Pro Ser Leu Arg Gly 165 170 175 Gly Ser Trp Ser Pro Arg Asp His Thr Pro Ala Thr Gln Gly Thr Tyr 180 185 190 Thr Asn Gly Arg Phe Val Ser Leu Pro Lys Ile Gly Ser Ser Arg Ala 195 200 205 5 25 DNA Artificial Sequence Primer 5 tccaggatga catagtccag gggcg 25 6 25 DNA Artificial Sequence Primer 6 tgggatgatt tcggcatgga caacg 25 7 52 DNA Artificial Sequence Primer 7 gtggtgctcg agtgcggccg caagctttat tattgtttga gcattcggcc tg 52 8 56 DNA Artificial Sequence Primer 8 atccgaattc tagatgcacc accaccacca ccacatgagg atgagtgatg gcgcag 56 9 31 DNA Artificial Sequence Primer 9 cggaattcgg atgaggatga gtgatggcgc a 31 10 35 DNA Artificial Sequence Primer 10 tctcgacaag cttttattgt ttgagcattc ggcct 35 11 20 DNA Artificial Sequence Primer 11 ccaaaagcca atggctctga 20 12 20 DNA Artificial Sequence Primer 12 agttgaatgg gctccagggt 20 13 20 DNA Artificial Sequence Primer 13 ccgccgggca aattaaccaa 20 14 21 DNA Artificial Sequence Primer 14 aggtgggcaa ggtaggggtt a 21 15 20 DNA Artificial Sequence Primer 15 gcgcagcgcc aaaagccaat 20 16 24 DNA Artificial Sequence Primer 16 gagtcctttg gcatgctacc cagg 24 17 20 DNA Artificial Sequence Primer 17 gccgccgggc aaattaacca 20 18 22 DNA Artificial Sequence Primer 18 ggcttaaccc ctaccttgcc ca 22 19 18 DNA Artificial Sequence Primer 19 cagtgccagc cctcttat 18 20 18 DNA Artificial Sequence Primer 20 gtcccttgat gaggagga 18 21 41 DNA Murine Norovirus type 1 21 ggaaagatgt ttgactctca ggtcattatc atcaccacaa a 41 22 42 DNA Murine Norovirus type 1 22 ggaaagatgt ttgactctca ggtcattatc atcaccacaa at 42 23 54 DNA Murine Norovirus type 1 23 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac cccc 54 24 65 DNA Murine Norovirus type 1 misc_feature (30)..(30) n is a, c, g, or t 24 ggaaagatgt ttgactctca ggtcattatn atnaccacaa atnaacaaac ccccgcgccc 60 ctgga 65 25 64 DNA Murine Norovirus type 1 misc_feature (43)..(43) n is a, c, g, or t 25 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atnaacaaac ccccgcgccc 60 ctgg 64 26 70 DNA Murine Norovirus type 1 26 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg 70 27 73 DNA Murine Norovirus type 1 misc_feature (43)..(43) n is a, c, g, or t 27 ggaaagatgc ttgactctca ggtcattatc atcaccacaa atnaacaaac ccccgcgccc 60 ctgnnctatg tca 73 28 77 DNA Murine Norovirus type 1 28 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacct 77 29 77 DNA Murine Norovirus type 1 29 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacct 77 30 79 DNA Murine Norovirus type 1 30 ggaaagatgc ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacctgg 79 31 79 DNA Murine Norovirus type 1 31 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacctgg 79 32 79 DNA Murine Norovirus type 1 32 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacctgg 79 33 79 DNA Murine Norovirus type 1 33 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaacctgg 79 34 127 DNA Murine Norovirus type 1 34 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 35 127 DNA Murine Norovirus type 1 35 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 36 127 DNA Murine Norovirus type 1 36 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 37 127 DNA Murine Norovirus type 1 37 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 38 127 DNA Murine Norovirus type 1 38 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 39 127 DNA Murine Norovirus type 1 39 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgatgag 120 agccctg 127 40 127 DNA Murine Norovirus type 1 40 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 41 127 DNA Murine Norovirus type 1 41 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 42 127 DNA Murine Norovirus type 1 42 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 43 127 DNA Murine Norovirus type 1 43 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 44 127 DNA Murine Norovirus type 1 misc_feature (24)..(24) n is a, c, g, or t 44 ggaaagatgt ttgactctca ggtnattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgctgag 120 agccctg 127 45 127 DNA Murine Norovirus type 1 45 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgtgccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgatgag 120 agccctg 127 46 117 DNA Murine Norovirus type 1 misc_feature (73)..(73) n is a, c, g, or t 46 ggaaagatgc ttgactctca ggtcattatc ataccacaaa tcaacaaacc cccgcgccct 60 ggactatgtc aanctggagg cggtctgccg ccgcatagat ttcctggttt atgctga 117 47 124 DNA Murine Norovirus type 1 misc_feature (75)..(75) n is a, c, g, or t 47 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaanctgga ggcggtctgc cgccgcatag atttcgttta tgatgagagc 120 cctg 124 48 119 DNA Murine Norovirus type 1 misc_feature (75)..(75) n is a, c, g, or t 48 ggaaagatgt ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgcgccc 60 ctggactatg tcaanctgga ggcggtctgc cgccgcatag atttcctggt ttatgctga 119 49 127 DNA Artificial Sequence Consensus sequence 49 ggaaagatgy ttgactctca ggtcattatc atcaccacaa atcaacaaac ccccgygccc 60 ctggactatg tcaacctgga ggcggtctgc cgccgcatag atttcctggt ttatgmtgag 120 agccctg 127 50 8 PRT Artificial Sequence Illustrative MNV-1 ORF1 motif 50 Gly Xaa Xaa Gly Xaa Gly Lys Thr 1 5 51 4 PRT Artificial Sequence Illustrative MNV-1 ORF1 motif 51 Gly Asp Cys Gly 1 52 4 PRT Artificial Sequence Illustrative MNV-1 ORF1 motif 52 Lys Asp Glu Leu 1 53 4 PRT Artificial Sequence Illustrative MNV-1 ORF1 motif 53 Gly Leu Pro Ser 1 54 4 PRT Artificial Sequence Illustrative MNV-1 ORF1 motif 54 Tyr Gly Asp Asp 1

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed