US8426574B2 - Identification of astrovirus VA1 associated with gastroenteritis in humans - Google Patents

Identification of astrovirus VA1 associated with gastroenteritis in humans Download PDF

Info

Publication number
US8426574B2
US8426574B2 US12/790,618 US79061810A US8426574B2 US 8426574 B2 US8426574 B2 US 8426574B2 US 79061810 A US79061810 A US 79061810A US 8426574 B2 US8426574 B2 US 8426574B2
Authority
US
United States
Prior art keywords
astv
virus
astrovirus
nucleic acid
sample
Prior art date
Legal status (The legal status 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 status listed.)
Expired - Fee Related, expires
Application number
US12/790,618
Other versions
US20110008353A1 (en
Inventor
David Wang
Herbert Whiting Virgin, IV
Guoyan Zhao
Stacy Finkbeiner
Jan Vinje
Yan Li
Suxiang Tong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GOVERNMENT OF United States, SECRETARY OF DEPARTMENT OF HEALTH AND HUMAN SERVICES CENTERS FOR DISEASE CONTROL AND PREVENTION
Centers of Disease Control and Prevention CDC
Washington University in St Louis WUSTL
Original Assignee
Washington University in St Louis WUSTL
Centers of Disease Control and Prevention CDC
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 Washington University in St Louis WUSTL, Centers of Disease Control and Prevention CDC filed Critical Washington University in St Louis WUSTL
Priority to US12/790,618 priority Critical patent/US8426574B2/en
Assigned to THE GOVERNMENT OF THE UNITED STATES OF AMERICA, SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, CENTERS FOR DISEASE CONTROL AND PREVENTION reassignment THE GOVERNMENT OF THE UNITED STATES OF AMERICA, SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, CENTERS FOR DISEASE CONTROL AND PREVENTION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VINJE, JAN, LI, YAN, TONG, SUXIANG
Assigned to WASHINGTON UNIVERSITY reassignment WASHINGTON UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZHAO, GUOYAN, VIRGIN IV, HERBERT W., FINKBEINER, STACY, WANG, DAVID
Publication of US20110008353A1 publication Critical patent/US20110008353A1/en
Application granted granted Critical
Publication of US8426574B2 publication Critical patent/US8426574B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: WASHINGTON UNIVERSITY
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2770/12011Astroviridae
    • C12N2770/12022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)

Definitions

  • This invention relates to virology and infectious disease. More particularly, the invention relates to a new human astrovirus.
  • Astroviruses are known to infect a variety of avian and mammalian species and typically cause diarrhea. Clinical symptoms usually last 2-4 days and consist of watery diarrhea and, less commonly, vomiting, headache, fever, abdominal pains, and anorexia. They are thought to be host specific with little evidence for cross-species transmission. In humans, 8 serotypes of astroviruses have been described (Mendez, E., et al., Fields Virology. 5th ed., Knipe, D. M., Howley, P. M., editors, Lippincott Williams & Wilkins, Philadelphia, Pa. (2007) 981-1000. Recently, a novel astrovirus (Astrovirus MLB1) (Finkbeiner, S.
  • the present invention relates to an additional infectious agent associated with diarrhea which provides a further means to diagnose, prevent and treat this condition.
  • This agent designated Astrovirus VA1 (AstV-VA1)
  • Astroviruses are a family of small, single-stranded, positive-sense RNA viruses. Their genomes are organized into three open reading frames denoted ORF's 1a, 1b, and 2, which encode a serine protease, RNA-dependent RNA polymerase (RdRP), and a capsid precursor protein, respectively. At both the 5′ and 3′ ends, non-translated regions (NTR) flank the 6.1-7.3 kb sized genomes.
  • ORF's 1a, 1b, and 2 which encode a serine protease, RNA-dependent RNA polymerase (RdRP), and a capsid precursor protein, respectively.
  • RdRP RNA-dependent RNA polymerase
  • capsid precursor protein capsid precursor protein
  • the invention relates to a novel astrovirus (AstV-VA1), initially detected in fecal samples from an outbreak of acute gastroenteritis in a child care center by two sequence independent genome amplification and sequencing methods, high throughput pyrosequencing and low throughput Sanger sequencing. The identification and presence of the novel astrovirus was confirmed in both sequencing methods.
  • the invention is directed to nucleic acids and proteins associated with AstV-VA1 that are useful in the diagnosis, prevention, and in identification and production of methods of treatment of diarrhea caused by this virus.
  • the new virus is distantly related to the known astrovirus agents associated with diarrhea, and even more distantly related from other causative agents associated with this condition.
  • Nucleic acids associated with the genome of the AstV-VA1 virus and their complements are useful in methods to produce the proteins, and thus one aspect of the invention is recombinant materials for such production. These include recombinant expression systems wherein heterologous control sequences effect expression of a viral protein or a useful portion thereof. Thus, methods for recombinantly producing viral proteins are included in the invention. These proteins or fragments may then be used in several ways. They may be used to produce antibodies which are also included in the scope of the invention. They may also be used in vaccines for immunizing individuals against infection. The proteins may also be used as screening tools to identify small molecules that may be used as therapeutic agents, somewhat analogous to the development of protease inhibitors in controlling HIV.
  • the antibodies themselves may be used for passive immunization and for detection of the presence of AstV-VA1 in biological samples or in the environment. They also may be used as targeting agents to couple cytotoxic agents (or detection reagents to seek out the virus itself).
  • nucleic acid of the AstV-VA1 or its complement, or, typically, portions thereof may be used as probes or primers for detection of the virus.
  • nucleic acids may be used to design ribosomes or antisense nucleic acids that inhibit the replication of the virus or production of viral proteins. Small interfering RNA or other methods of gene silencing may also be used.
  • Methods for detecting AstV-VA1 either at a protein or nucleic acid level are within the scope of the invention, as are methods to reduce the susceptibility of a subject to such infection or to ameliorate the symptoms by prophylactically administering an immunogenic portion of the virus protein or the attenuated virus itself or by generating antibodies intracellularly using nucleic acids isolated from the invention antibodies.
  • FIGS. 1A-1C illustrate the phylogenetic analysis of AstV-VA1 Virus. Amino acid based phylogenetic trees were generated using PAUP with 1000 bootstrap replicates. Significant bootstrap values are shown. A) ORF1a serine protease; B) ORF1b polymerase; C) ORF2 capsid precursor.
  • FIG. 2 shows the sequence of the AstV-VA1 virus genome (SEQ ID NO:1).
  • FIG. 3 shows the nucleotide sequence of ORF1a of the AstV-VA1 virus (SEQ ID NO:2).
  • FIG. 4 shows the polypeptide sequence of ORF1a of the AstV-VA1 virus (SEQ ID NO:3).
  • FIG. 5 shows the nucleotide sequence of ORF1b of the AstV-VA1 virus (SEQ ID NO:4).
  • FIG. 6 shows the polypeptide sequence of ORF1b of the AstV-VA1 virus (SEQ ID NO:5).
  • FIG. 7 shows the nucleotide sequence of ORF2 of the AstV-VA1 virus (SEQ ID NO:6).
  • FIG. 8 shows the polypeptide sequence of ORF2 of the AstV-VA1 virus (SEQ ID NO:7).
  • AstV-VA1 permits identification and/or preparation of a therapeutic target, an immunogenic agent, a diagnostic agent, or a therapeutic agent previously unavailable to assess and counteract the infectious agents associated with cases of diarrhea that have not been previously characterized.
  • AstV-VA1 is in the family of astroviruses known to be associated with the symptoms of diarrhea, as shown in FIGS. 1A-1C this virus is relatively distantly related to the previously known astroviruses at the protein level.
  • FIG. 1A shows that the serine protease encoded by ORF1a contains hundreds of amino acid substitutions as compared to the previous family of eight human AstV's and even as compared to MLB1.
  • the polymerase as shown in FIG.
  • AstV-VA1 the availability of the protease and polymerase of AstV-VA1 permits these proteins to be used to screen for inhibitors of the activity of these proteins which would be useful as therapeutics once the subject is infected.
  • the availability of the nucleic acid of this virus also permits the design of ribozymes and antisense inhibitors of expression. Since the AstV-VA1 is only distantly related to other known astroviruses, and indeed other agents that cause diarrhea in humans, its materials are vital to complete the fabric of diagnostics and vaccines to detect the agent causing this condition as well as immunogenic compositions for its prevention.
  • Diagnosis of diarrhea per se is, of course, not problematic, but determination of the infectious agent associated with it is important since this permits the epidemiology of an infection to be traced.
  • an outbreak of diarrhea may, by tracking the causative agent, be traced to a particular source such as a well or food manufacturing plant. Detection of the causative agent is significant in controlling outbreaks of this condition.
  • the complete nucleotide sequence of AstV-VA1 is shown in FIG. 2 .
  • the invention includes useful portions of this sequence and complements thereto. It further includes nucleic acid molecules that hybridize under stringent conditions to SEQ ID NO:1 or its complement or to a significant portion thereof, comprising at least 100 nucleotides, preferably 200 or 300 nucleotides.
  • under stringent conditions refers to hybridization and washing conditions under which nucleotide sequences having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% identity to each other remain hybridized to each other.
  • stringent hybridization conditions are hybridization in 6 ⁇ sodium chloride/sodium citrate (SSC), 0.5% SDS at about 68° C.
  • stringent hybridization conditions are hybridization in 6 ⁇ SSC at about 45° C. followed by one or more washes (e.g., about 5 to 30 min each) in 0.1 ⁇ SSC, 0.1% SDS at about 45-65° C.
  • the nucleic acid molecules may be analogous to those occurring in nature, or may have modified linkages such as thioester or phosphoramidate linkages or may be peptide nucleic acids or nucleic acids with other modified backbones so long as the binding specificity of the sequence of bases is retained.
  • the invention further includes the proteins encoded by the open reading frames of AstV-VA1 or homologs thereof comprising at least 80%, or 85% or 90% or 95% or 98% identity to these sequences.
  • the proteins or polypeptides may also include characteristic immunogenic portions of these sequences and their homologs as well as means to produce them.
  • a “characteristic immunogenic portion” is meant a significant portion of the peptide which will elicit antibodies that are specifically reactive with the protein, as opposed to other proteins likely to be present in the same context, such as the proteins encoded by the genomes of other astroviruses or other viruses that cause diarrhea.
  • the portion will be selected from regions of diversity in the amino acid sequences of the AstV-VA1 protein as compared to other proteins associated with viruses causing diarrhea.
  • peptide As used herein, “peptide”, “protein”, and “polypeptide” are used interchangeably without regard to the length of the amino acid chain.
  • compositions comprising a polypeptide of SEQ ID NO:7 or a characteristic immunogenic portion thereof are preferred, as these represent the capsid proteins that are exposed on the virus.
  • Attenuated virus vaccines comprising the AstV-VA1 virus itself may also be employed. “Attenuated” virus includes killed or inactivated virus, as well as virus that is merely weakened.
  • Chimeric AstV-VA1 viruses are also part of the invention. These are recombinant AstV-VA1 viruses which further comprise a heterologous nucleotide sequence.
  • the genome of a chimeric virus contains a nucleotide sequence heterologous to the AstV-VA1 genome.
  • heterologous refers to a portion of a nucleic acid or a protein that is not natively found coupled to the nucleic acid or protein referred to.
  • control sequences that are heterologous to a coding sequence are those which are not found bound to said coding sequence in nature.
  • the invention includes expression systems where heterologous control sequences are coupled operably to sequences encoding the proteins encoded by AstV-VA1 virus or portions thereof.
  • Such expression systems may be included in vectors and transfected into suitable host cells for production of the proteins.
  • Cell types include prokaryotic cells, insect cells, mammalian cells, yeast cells, plant cells and the like.
  • Primers and probes useful for the amplification and detection of the human astrovirus AstV-VA1 are also included in the invention, as well as methods to detect AstV-VA1 virus employing them.
  • Probes typically contain AstV-VA1 virus (SEQ ID NO:1), sequences substantially identical thereto, or their complements including portions comprising at least 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, or 500 consecutive bases.
  • a primer or probe comprises an oligonucleotide comprising at least about 20 to 50 consecutive bases of the sequence.
  • any suitable method for detecting the presence of AstV-VA1 may be employed but methods that can be adapted to detect the presence of AstV-VA1 virus are well known in the art.
  • a sample or nucleic acids isolated from the sample are contacted with a probe which can be detected in a variety of ways such as fluorescent or radioactive labels.
  • the nucleic acids in the sample may be first amplified and then identified typically using suitable probes.
  • the presence of viral proteins in the sample can be detected by a wide variety of immunological assays using antibodies of the invention.
  • the samples for detection may include any tissue or bodily fluid or excretum which is expected to contain an infectious agent.
  • AstV-VA1 is associated with diarrhea, typically the sample is a fecal sample.
  • primers useful to amplify the virus bind specifically to AstV-VA1.
  • Exemplary primers include forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) (SEQ ID NO:8) and reverse primer (5′ GTC TAT TGT TTT GGG CGT CTG C 3′) (SEQ ID NO:9) as well as forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) (SEQ ID NO:10) and reverse primer (5′ CGG GGG TGG TGC GAC AT 3′) (SEQ ID NO:11).
  • forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′)
  • reverse primer 5′ AGG GGT CGC TGG GAG TTT G 3′
  • SEQ ID NO:11 reverse primer
  • These primer pairs may be used in nested PCR.
  • real time PCR may be used to detect the samples. See Example 3 for an exemplary method of detecting AstV-VA1 in a sample that may be used in some embodiments.
  • Amplification reactions can also quantify the amount of nucleic acid in a sample, label the nucleic acid (e.g., to apply it to an array or a blot), or detect the nucleic acid.
  • Amplification methods are well known in the art, and include, e.g., polymerase chain reaction (PCR); ligase chain reaction (LCR); transcription amplification; self-sustained sequence replication; Q-Beta replicase amplification; automated Q-Beta replicase amplification assay; and other RNA polymerase mediated techniques.
  • Antibodies that specifically bind a polypeptide sequence encoded by ORF1a, ORF1b, and ORF2 or portions thereof are included in the invention.
  • An antibody or an antibody fragment is specific for a polypeptide of the AstV-VA1 virus if it permits one of skill in the art to discern the presence of the virus or a protein or peptide encoded by the virus in a sample, and is not cross-reactive with non-AstV-VA1 viral antigens.
  • For detection antibodies include polyclonal, monoclonal, bi-specific, multi-specific, single chain antibodies, diabodies, nanobodies, single domain antibodies (e.g., camel antibodies), Fab, F(ab′) 2 , Fvs, intrabodies and fragments containing either a V L or V H domain or even a complementary determining region (CDR) that specifically binds to a polypeptide of the AstV-VA1 virus disclosed herein.
  • CDR complementary determining region
  • Human, humanized and chimeric forms are also included in the invention as these are most useful for passive immunization. Particularly preferred for this application are neutralizing antibodies that can be identified using culture testing or other means.
  • Antibodies useful in the detection of AstV-VA1 virus can be labeled with any suitable detectable label and employed in assays such as enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence.
  • ELISAs enzyme linked immunosorbent assays
  • Western blots immunoprecipitations and immunofluorescence.
  • compositions comprising the virus or portions thereof and a pharmaceutically acceptable carrier.
  • some compositions can comprise one or more isolated proteins from the AstV-VA1 virus, live AstV-VA1 virus, attenuated AstV-VA1 virus, or inactivated AstV-VA1 virus.
  • Methods for detecting the presence or absence of a polypeptide or nucleic acid from the AstV-VA1 virus in a biological sample comprises contacting the sample with a compound or an agent capable of detecting an epitope on a protein or nucleic acid (e.g., mRNA, genomic DNA) of the AstV-VA1 virus such that the presence of AstV-VA1 virus is detected in the sample.
  • a compound or an agent capable of detecting an epitope on a protein or nucleic acid (e.g., mRNA, genomic DNA) of the AstV-VA1 virus such that the presence of AstV-VA1 virus is detected in the sample.
  • Another aspect provides for methods of detecting an antibody, which immunospecifically binds to the AstV-VA1 virus, in a biological sample, for example blood, plasma or serum.
  • the method comprising contacting the sample with the polypeptides or protein encoded by the nucleotide sequence of AstV-VA1 virus, or a characteristic portion thereof, directly immobilized on a substrate and detecting the virus-bound antibody directly or indirectly by a labeled heterologous anti-isotype antibody.
  • Kits for detecting the presence of AstV-VA1 virus, or a nucleic acid polypeptide thereof or antibody thereto are included. Kits can also include instructions for use.
  • the kit can comprise, for example: (1) a first antibody (e.g., attached to a solid support) which binds to a polypeptide or epitope encoded by the AstV-VA1 virus; and, optionally, (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
  • a first antibody e.g., attached to a solid support
  • a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
  • the kit can comprise, for example: (1) an oligonucleotide, e.g., a detectably labeled oligonucleotide, which hybridizes to a nucleic acid sequence contained within SEQ ID NO:1 or to a sequence within the AstV-VA1 viral genome and/or (2) a pair of primers useful for amplifying a nucleic acid molecule containing an AstV-VA1 viral sequence.
  • the kit can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent.
  • the kit can also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate).
  • the kit can also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained. Each component of the kit is usually enclosed within an individual container and all of the various containers are within a single package along with instructions for use.
  • Yet another aspect provides a method of screening for anti-viral agents useful in reducing the symptoms of AstV-VA1 infections comprising: contacting a cell infected with the human astrovirus AstV-VA1 with a candidate anti-viral agent; assaying the anti-viral agent activity by determining the effect of the agent upon viral titer in the cell, and identifying the agent as an anti-viral agent if it inhibits viral replication, expression, or activity.
  • the methods can be designed to screen for agents in in vitro assays against cell lines infected with the virus, against cells producing an enzyme from a virus or against a purified viral enzyme. Alternatively, the agents may be screened in in vivo assays where the virus is hosted by a mammal.
  • Identified anti-viral agent can prevent or inhibit the binding of the virus or viral proteins to a host cell under a physiological condition, thereby preventing or inhibiting the infection of the host cell by the virus.
  • Anti-viral agents may prevent or inhibit replication of the viral nucleic acid molecules in the host cell under a physiological condition by interacting with the viral nucleic acid molecules or its transcription mechanisms.
  • the antiviral agent may also inhibit the activity of essential viral proteins as set forth above.
  • Test viral inhibitory molecules also can be obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs.
  • fecal specimens Five fecal specimens (labeled A, B, C, D and E) were collected from a gastroenteritis outbreak at a child care center in Virginia. Symptoms included vomiting and/or diarrhea.
  • the five fecal specimens (A-E) (Table 1) tested negative for enteric parasites, enteric bacteria by standard microscopy and culture, and negative for enteric viruses including rotavirus (RotaClone EIA), norovirus, sapovirus, human astrovirus, and adenovirus gp F by (RT)-PCR.
  • Sequences from each sample were identified by the unique barcodes introduced during the Rd B amplification. Primer and barcode sequences were then trimmed off prior to analysis of the sequences. Sequences were clustered using CD-HIT (Li, W., et al., Bioinformatics (2006) 22:1658-1659) to reduce redundancy with the requirement that they had to be 98% identical over 98% of their lengths. The longest sequence from each cluster was selected for inclusion in the pool of unique sequences to be analyzed. Unique sequences were filtered for repetitive sequences and compared with the human genome using BLASTn with an e-value cutoff of 1e-10.
  • Sequences without significant similarity to the human genome were then compared to the GenBank nucleic acid database using BLASTn (cutoff: 1e-10) and tBLASTx (cutoff: 1e-5), and remaining sequences without significant hits to sequences in the database were then compared to the NCBI All Viral Genome database located on the World Wide Web using tBLASTx (cutoff: 1e-5) (Altschul, S. F., Nucleic Acids Res . (1997) 25:3389-3402). Overlapping sequences with significant sequence identity were assembled into contigs using Newbler (454 Life Sciences) or CAP3 (Huang, X., et al., Genome Res . (1999) 9:969-877).
  • TNA Total nucleic acid
  • the 300-800 bp amplicons were then cloned using the TOPO TA CLONING® kit (Invitrogen, Carlsbad, Calif.) and plasmids were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit on an ABI PRISM® 3130 automated sequencer (Applied Biosystems, Foster City, Calif.). Sequence analysis and generation of contigs were performed using Sequencher software (Ann Arbor, Mich., USA). Sequence identification was performed through NCBI nucleotide-nucleotide BLASTn similarity searches.
  • a set of eight overlapping RT-PCR products with an average size of 900 bp which cover the entire genome including the 3′ end poly A tail were generated by primer pairs designed from clone sequences as described above, using the SUPERSCRIPTTM III First-Strand Synthesis System for RT-PCR and ACCUPRIMETM High Fidelity Taq DNA polymerase (Invitrogen, Carlsbad, Calif., USA). Both strands of each amplicon were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit as described above. The 5′ end genome sequence was amplified and determined using the 5′/3′ RACE Kit (Roche, Mannheim, Germany) following the manufacturer's instructions. The complete genome sequence of AstV-VA1 has been deposited in Genbank and is shown in FIG. 2 as SEQ ID NO:1.
  • RNA and DNA extracted from samples A, B, C and D were 313 unique high quality sequence reads were found in sample B and 1,017 unique high quality reads were found in sample C most closely related to astroviruses.
  • a 6,376 nucleotide (nt) contig was assembled from the astrovirus-like sequences detected in sample B and 4 contigs totaling 6,026 nucleotides were assembled from sample C.
  • the translated contigs had only limited sequence similarity (37-71% aa identity) to proteins from mink and ovine astroviruses. The five original contigs were then assembled to generate a larger contig of 6,581 nucleotides in length.
  • Primers were then designed to generate a series of eight overlapping RT-PCR amplicons with an average size of ⁇ 900 bp that yielded a genomic sequence of 6,537 nt.
  • three independent 5′RACE reactions were performed and a total of 23 clones from these reactions were sequenced. All clones extended the genome by 49 nt and yielded the identical 5′end sequence, suggesting that the genome was complete with a total length of 6,586 nt, excluding the poly-A tail.
  • ORF 1a Open Reading Frame Analysis.
  • the genome of AstV-VA1 had three predicted open reading frames (ORF 1a, 1b, and 2) as well as non-translated regions (NTRs) at both the 5′ and 3′ ends of the genome.
  • Open reading frames (ORFs) 1a and 2 were predicted by the NCBI ORF Finder located on the World Wide Web.
  • the end of ORF1b was also predicted by NCBI ORF Finder, however the start of ORF1b was predicted based on the location of the heptameric slippery sequence found in other astroviruses. Protein motifs were identified by conserved domain searches using BlastX and Pfam.
  • ORFs 1a and 2 were predicted by the NCBI ORF Finder program; however the full coding region for ORF1b was not predicted by the program because translation of ORF1b is dependent on a ⁇ 1 ribosomal frameshift that occurs during translation. This frameshift is thought to be mediated by the presence of a heptameric ‘slippery sequence’ (AAAAAAAC) near the end of ORF1a, which was also conserved in the AstV-VA1 sequence, suggesting that this new virus follows the same paradigm.
  • AAAAAAAC heptameric ‘slippery sequence’
  • the 5′ non-translated region (NTR) of AstV-VA1 is 38 nt in length, which is between the lengths of the 5′ NTRs of mink astrovirus (26 nt) and ovine astrovirus (45 nt).
  • the 3′ NTR is 98 nt in length, which again is intermediate between the length of the NTRs of ovine astrovirus (59 nt) and mink astrovirus (108 nt).
  • the 3′ NTR of nearly all astroviruses contains a highly conserved RNA secondary structure called the stem-loop II-like motif (s2m), which has also been identified in several coronaviruses and in equine rhinovirus 2.
  • ClustalX (1.83) was used to carry out multiple sequence alignments of the protein sequences associated with all three of the open reading frames of astroviruses for which sequences were available.
  • Maximum parsimony trees were generated using PAUP with 1,000 bootstrap replicates (Swofford, D. L., PAUP*. Phylogenetic Analysis Using Parsimony (* and Other Methods ), Sunderland, Mass.: Sinauer Associates (1998)).
  • the maximum parsimony trees confirmed that AstV-VA1 was highly divergent from, but most closely related to mink and ovine astrovirus in all three ORFs ( FIGS. 1A-1C ). Furthermore, the greatest sequence identity between AstV-VA1 and mink and ovine astroviruses is in ORF1b with 61% amino acid identity to mink astrovirus and 62% to ovine astrovirus. The ORF1a (serine protease) coding region was more divergent with 39% and 40% amino acid identity with ovine astrovirus and mink astrovirus, respectively. In ORF2, AstV-VA1 virus shared 41% amino acid identity to mink astrovirus and 42% to ovine astrovirus.
  • AstV-VA1 was highly divergent from all previously described astroviruses including the 8 human astrovirus serotypes and recently described astrovirus MLB1 (AstV-MLB1). AstV-VA1 appears to have diverged from a common ancestor of the mink and ovine astroviruses following their separation from the branch containing human astroviruses 1-8 and astrovirus MLB1. The discovery of AstV-VA1 following the recent identification of AstV-MLB1 clearly demonstrates that a much greater diversity of astroviruses exists in humans than is commonly recognized.
  • Example 1 The samples collected in Example 1 were screened in order to assess the presence of AstV-VA1 in each of the samples. High throughput pyrosequencing yielded many AstV-VA1 sequences in samples B and C, but none were detected in samples A or D. Sample E was not analyzed by pyrosequencing due to technical problems with the sample preparation. Similarly, Sanger sequencing detected AstV-VA1 positive reads in samples B and C, but not in samples A and E (sample D was not initially tested). To determine whether low levels of AstV-VA1 might be present in samples A, D and E, real time RT-PCR and semi-nested RT-PCR assays were developed targeting regions in ORF1b and ORF2, respectively. Using these assays, sample D tested positive and sequencing of the 250 bp amplicon confirmed the presence of AstV-VA1.
  • the real-time RT-PCR assay was performed using the SUPERSCRIPTTM III One-Step RT-PCR kit (Invitrogen Corp., Carlsbad, Calif.) and the MX4000® system (Stratagene, La Jolla, Calif.).
  • Each 50 ⁇ l reaction mixture contained 900 pmol of forward primer (5′ TAT CCA TAG TTG TGG ATA TTT GTC CA 3′), 1,000 pmol of reverse primer (5′ TGT CTT AGG GGA GAC TTG CAA A 3′) and 100 pmol of probe (5′ TT CC CCCT GTC CTG GAT TGT CAC TTC 3′), 1 ⁇ buffer, 6.0 mM MgSO4 (final concentration), 20 units of RNase inhibitor, a 5 pJ aliquot of RNA extracts, and 1 unit of SUPERSCRIPTTM III RT/PLATINUM® TAQ MIX. Water was added to achieve a final volume of 50 ⁇ l. The RT-PCR reaction mixture was incubated at 60° C.
  • CT threshold cycle
  • the first round RT-PCR in the semi-nested assay was performed according to the protocol described previously (Tong, S., et al., J. Clin. Microbiol . (2008) 46:2652-2658) using forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) and reverse primer (5′ GTC TAT TGT TTT GGG CGT CTG C 3′).
  • the 2nd round PCR in the semi-nested assay PCR assay in 50 ⁇ l reaction mixture contained 1 ⁇ buffer (PLATINUM® Taq kit; Invitrogen), 2 mM MgCl2, 200 ⁇ M (each) of deoxynucleoside triphosphates, 50 pmol (each) of forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) and reverse primer (5′ CGG GGG TGG TGC GAC AT 3′) 1 U PLATINUM® Taq, one 2- ⁇ l aliquot from the first reaction, and water to achieve a final volume of 50 ⁇ l. The mixture was first heated to 94° C. for 2 min.
  • the cycling conditions were 40 cycles with the same conditions as for the first amplification: 94° C. for 15 s, primer annealing at 55° C. for 30 s, and 72° C. for 30 s. A final extension was carried out at 72° C. for 7 min.
  • the final semi-nested PCR products were visualized by UV light after electrophoresis on a 2% agarose gel containing 0.5 ug/ml ethidium bromide in 0.5 ⁇ Tris-borate buffer. Amplicons from the final round of PCR were purified using the QIAQUICK® PCR purification kit (Qiagen, Inc., Valencia, Calif.). Both strands of the amplicons were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit as described above.

Abstract

Provided herein is a novel human astrovirus, its nucleic acid sequence, as well as methods to detect and diagnose the presence of the astrovirus.

Description

CROSS-REFERENCE TO RELATED APPLICATION
This application claims benefit of U.S. Ser. No. 61/182,008 filed 28 May 2009. The contents of this document are incorporated herein by reference.
STATEMENT OF RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH
This work was supported in part by a grant from the National Institutes of Health to the Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research (2 U54 AI057160-06). The U.S. government has certain rights in this invention.
REFERENCE TO SEQUENCE LISTING SUBMITTED VIA EFS-WEB
The entire content of the following electronic submission of the sequence listing via the USPTO EFS-WEB server, as authorized and set forth in MPEP §1730 II.B.2(a)(C), is incorporated herein by reference in its entirety for all purposes. The sequence listing is identified on the electronically filed text file as follows:
File Name Date of Creation Size (bytes)
295002007900Seqlist.txt Jul. 28, 2010 37,002 bytes
TECHNICAL FIELD
This invention relates to virology and infectious disease. More particularly, the invention relates to a new human astrovirus.
BACKGROUND ART
Astroviruses are known to infect a variety of avian and mammalian species and typically cause diarrhea. Clinical symptoms usually last 2-4 days and consist of watery diarrhea and, less commonly, vomiting, headache, fever, abdominal pains, and anorexia. They are thought to be host specific with little evidence for cross-species transmission. In humans, 8 serotypes of astroviruses have been described (Mendez, E., et al., Fields Virology. 5th ed., Knipe, D. M., Howley, P. M., editors, Lippincott Williams & Wilkins, Philadelphia, Pa. (2007) 981-1000. Recently, a novel astrovirus (Astrovirus MLB1) (Finkbeiner, S. R., et al., Virol J. (2008) 5:117; Finkbeiner, S. R., et al., PLoS Pathog (2008) 4:e1000011) and a novel picornavirus (Cosavirus E1) (Holtz, L. R., et al., Virol J. (2008) 5:159) were identified in diarrhea patients by the present inventors.
Human astroviruses have been associated with up to ˜10% of sporadic cases of viral diarrhea in children and with 0.5-15% of outbreaks. Significantly, in some reports the etiologies of 12-41% of the outbreaks remain undetermined even after extensive testing. Similarly, on average, approximately 40% of the cases of sporadic diarrhea are unexplained. It is therefore evident that additional infectious agents associated with diarrhea remain undiscovered.
The present invention relates to an additional infectious agent associated with diarrhea which provides a further means to diagnose, prevent and treat this condition. This agent, designated Astrovirus VA1 (AstV-VA1), was identified and sequenced as associated with a gastroenteritis outbreak at a child care center.
Astroviruses are a family of small, single-stranded, positive-sense RNA viruses. Their genomes are organized into three open reading frames denoted ORF's 1a, 1b, and 2, which encode a serine protease, RNA-dependent RNA polymerase (RdRP), and a capsid precursor protein, respectively. At both the 5′ and 3′ ends, non-translated regions (NTR) flank the 6.1-7.3 kb sized genomes. Two characteristic features of astroviruses are the dependency on a ribosomal frameshift for the translation of ORF1b and the generation of a sub-genomic RNA from which ORF2 is translated.
DISCLOSURE OF THE INVENTION
The invention relates to a novel astrovirus (AstV-VA1), initially detected in fecal samples from an outbreak of acute gastroenteritis in a child care center by two sequence independent genome amplification and sequencing methods, high throughput pyrosequencing and low throughput Sanger sequencing. The identification and presence of the novel astrovirus was confirmed in both sequencing methods.
Thus, in one aspect, the invention is directed to nucleic acids and proteins associated with AstV-VA1 that are useful in the diagnosis, prevention, and in identification and production of methods of treatment of diarrhea caused by this virus. As further explained below, the new virus is distantly related to the known astrovirus agents associated with diarrhea, and even more distantly related from other causative agents associated with this condition.
While treatment of diarrhea is generally directed to simply alleviating the symptoms, more sophisticated methods of prevention and treatment may be desirable. Similarly, while diagnosis of diarrhea as a condition is self-evident, identification of the causative agent may permit control of epidemiology. These aspects of diagnosis and treatment are specific to the causative agent and thus the identification of this new virus opens up these avenues of diagnosis and control.
Nucleic acids associated with the genome of the AstV-VA1 virus and their complements are useful in methods to produce the proteins, and thus one aspect of the invention is recombinant materials for such production. These include recombinant expression systems wherein heterologous control sequences effect expression of a viral protein or a useful portion thereof. Thus, methods for recombinantly producing viral proteins are included in the invention. These proteins or fragments may then be used in several ways. They may be used to produce antibodies which are also included in the scope of the invention. They may also be used in vaccines for immunizing individuals against infection. The proteins may also be used as screening tools to identify small molecules that may be used as therapeutic agents, somewhat analogous to the development of protease inhibitors in controlling HIV.
The antibodies themselves may be used for passive immunization and for detection of the presence of AstV-VA1 in biological samples or in the environment. They also may be used as targeting agents to couple cytotoxic agents (or detection reagents to seek out the virus itself).
In addition to their use as a tool for recombinant production of viral proteins, the nucleic acid of the AstV-VA1 or its complement, or, typically, portions thereof, may be used as probes or primers for detection of the virus. In addition, the nucleic acids may be used to design ribosomes or antisense nucleic acids that inhibit the replication of the virus or production of viral proteins. Small interfering RNA or other methods of gene silencing may also be used.
Methods for detecting AstV-VA1 either at a protein or nucleic acid level are within the scope of the invention, as are methods to reduce the susceptibility of a subject to such infection or to ameliorate the symptoms by prophylactically administering an immunogenic portion of the virus protein or the attenuated virus itself or by generating antibodies intracellularly using nucleic acids isolated from the invention antibodies.
Methods of identifying therapeutics and designing them are also included.
BRIEF DESCRIPTION OF THE DRAWINGS
FIGS. 1A-1C illustrate the phylogenetic analysis of AstV-VA1 Virus. Amino acid based phylogenetic trees were generated using PAUP with 1000 bootstrap replicates. Significant bootstrap values are shown. A) ORF1a serine protease; B) ORF1b polymerase; C) ORF2 capsid precursor.
FIG. 2 shows the sequence of the AstV-VA1 virus genome (SEQ ID NO:1).
FIG. 3 shows the nucleotide sequence of ORF1a of the AstV-VA1 virus (SEQ ID NO:2).
FIG. 4 shows the polypeptide sequence of ORF1a of the AstV-VA1 virus (SEQ ID NO:3).
FIG. 5 shows the nucleotide sequence of ORF1b of the AstV-VA1 virus (SEQ ID NO:4).
FIG. 6 shows the polypeptide sequence of ORF1b of the AstV-VA1 virus (SEQ ID NO:5).
FIG. 7 shows the nucleotide sequence of ORF2 of the AstV-VA1 virus (SEQ ID NO:6).
FIG. 8 shows the polypeptide sequence of ORF2 of the AstV-VA1 virus (SEQ ID NO:7).
MODES OF CARRYING OUT THE INVENTION
The isolation of AstV-VA1 permits identification and/or preparation of a therapeutic target, an immunogenic agent, a diagnostic agent, or a therapeutic agent previously unavailable to assess and counteract the infectious agents associated with cases of diarrhea that have not been previously characterized. Although AstV-VA1 is in the family of astroviruses known to be associated with the symptoms of diarrhea, as shown in FIGS. 1A-1C this virus is relatively distantly related to the previously known astroviruses at the protein level. Thus, FIG. 1A shows that the serine protease encoded by ORF1a contains hundreds of amino acid substitutions as compared to the previous family of eight human AstV's and even as compared to MLB1. Similarly, the polymerase as shown in FIG. 1B contains hundreds of such substitutions and the capsid protein, as shown in FIG. 1C, contains thousands of such substitutions. It is apparent that antibodies that would successfully detect the known astroviruses would not necessarily be crossreactive with the antibodies that would successfully detect AstV-VA1, nor would nucleic acid probes directed against these prior art viruses be successful in detecting the presence of AstV-VA1. Importantly, immunological compositions which would enhance the resistance of an individual to AstV-VA1 are made available as administering immunogenic portions of the capsid protein of the prior art viruses would not successfully result in such enhancement of the infective agent is AstV-VA1. In addition, antibodies detected in a subject that are immunoreactive with the prior art viruses or their proteins would not offer the opportunity to diagnose infection with AstV-VA1.
Further, the availability of the protease and polymerase of AstV-VA1 permits these proteins to be used to screen for inhibitors of the activity of these proteins which would be useful as therapeutics once the subject is infected. The availability of the nucleic acid of this virus also permits the design of ribozymes and antisense inhibitors of expression. Since the AstV-VA1 is only distantly related to other known astroviruses, and indeed other agents that cause diarrhea in humans, its materials are vital to complete the fabric of diagnostics and vaccines to detect the agent causing this condition as well as immunogenic compositions for its prevention.
Diagnosis of diarrhea per se is, of course, not problematic, but determination of the infectious agent associated with it is important since this permits the epidemiology of an infection to be traced. Thus, an outbreak of diarrhea may, by tracking the causative agent, be traced to a particular source such as a well or food manufacturing plant. Detection of the causative agent is significant in controlling outbreaks of this condition.
The complete nucleotide sequence of AstV-VA1 is shown in FIG. 2. The invention includes useful portions of this sequence and complements thereto. It further includes nucleic acid molecules that hybridize under stringent conditions to SEQ ID NO:1 or its complement or to a significant portion thereof, comprising at least 100 nucleotides, preferably 200 or 300 nucleotides. The term “under stringent conditions” refers to hybridization and washing conditions under which nucleotide sequences having at least 80%, at least 85%, at least 90%, at least 95% or at least 98% identity to each other remain hybridized to each other. In one example, stringent hybridization conditions are hybridization in 6× sodium chloride/sodium citrate (SSC), 0.5% SDS at about 68° C. followed by one or more washes (e.g., about 5 to 30 min each) in 2×SSC, 0.5% SDS at room temperature. In another example, stringent hybridization conditions are hybridization in 6×SSC at about 45° C. followed by one or more washes (e.g., about 5 to 30 min each) in 0.1×SSC, 0.1% SDS at about 45-65° C.
The nucleic acid molecules may be analogous to those occurring in nature, or may have modified linkages such as thioester or phosphoramidate linkages or may be peptide nucleic acids or nucleic acids with other modified backbones so long as the binding specificity of the sequence of bases is retained.
The invention further includes the proteins encoded by the open reading frames of AstV-VA1 or homologs thereof comprising at least 80%, or 85% or 90% or 95% or 98% identity to these sequences. The proteins or polypeptides may also include characteristic immunogenic portions of these sequences and their homologs as well as means to produce them.
By a “characteristic immunogenic portion” is meant a significant portion of the peptide which will elicit antibodies that are specifically reactive with the protein, as opposed to other proteins likely to be present in the same context, such as the proteins encoded by the genomes of other astroviruses or other viruses that cause diarrhea. Thus, the portion will be selected from regions of diversity in the amino acid sequences of the AstV-VA1 protein as compared to other proteins associated with viruses causing diarrhea.
As used herein, “peptide”, “protein”, and “polypeptide” are used interchangeably without regard to the length of the amino acid chain.
The polypeptides of the invention are useful for the generation of antibodies directed against them or directed against the virus. For use as a vaccine, compositions comprising a polypeptide of SEQ ID NO:7 or a characteristic immunogenic portion thereof are preferred, as these represent the capsid proteins that are exposed on the virus. Attenuated virus vaccines comprising the AstV-VA1 virus itself may also be employed. “Attenuated” virus includes killed or inactivated virus, as well as virus that is merely weakened.
Chimeric AstV-VA1 viruses are also part of the invention. These are recombinant AstV-VA1 viruses which further comprise a heterologous nucleotide sequence. The genome of a chimeric virus contains a nucleotide sequence heterologous to the AstV-VA1 genome.
As used here, “heterologous” refers to a portion of a nucleic acid or a protein that is not natively found coupled to the nucleic acid or protein referred to. Thus, for example control sequences that are heterologous to a coding sequence are those which are not found bound to said coding sequence in nature.
Thus, the invention includes expression systems where heterologous control sequences are coupled operably to sequences encoding the proteins encoded by AstV-VA1 virus or portions thereof. Such expression systems may be included in vectors and transfected into suitable host cells for production of the proteins. Cell types include prokaryotic cells, insect cells, mammalian cells, yeast cells, plant cells and the like.
Primers and probes useful for the amplification and detection of the human astrovirus AstV-VA1 are also included in the invention, as well as methods to detect AstV-VA1 virus employing them. Probes typically contain AstV-VA1 virus (SEQ ID NO:1), sequences substantially identical thereto, or their complements including portions comprising at least 20, 25, 30, 35, 40, 50, 75, 100, 150, 200, 300, 400, or 500 consecutive bases. In one embodiment, a primer or probe comprises an oligonucleotide comprising at least about 20 to 50 consecutive bases of the sequence.
Any suitable method for detecting the presence of AstV-VA1 may be employed but methods that can be adapted to detect the presence of AstV-VA1 virus are well known in the art. For detection of nucleic acids associated with the virus, typically a sample or nucleic acids isolated from the sample are contacted with a probe which can be detected in a variety of ways such as fluorescent or radioactive labels. Alternatively, the nucleic acids in the sample may be first amplified and then identified typically using suitable probes. The presence of viral proteins in the sample can be detected by a wide variety of immunological assays using antibodies of the invention.
The samples for detection may include any tissue or bodily fluid or excretum which is expected to contain an infectious agent. As AstV-VA1 is associated with diarrhea, typically the sample is a fecal sample.
Thus, prior to detection one may amplify the virus or nucleotides of the virus in order to improve sensitivity. Primers useful to amplify the virus bind specifically to AstV-VA1. Exemplary primers include forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) (SEQ ID NO:8) and reverse primer (5′ GTC TAT TGT TTT GGG CGT CTG C 3′) (SEQ ID NO:9) as well as forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) (SEQ ID NO:10) and reverse primer (5′ CGG GGG TGG TGC GAC AT 3′) (SEQ ID NO:11). These primer pairs may be used in nested PCR. Optionally, real time PCR may be used to detect the samples. See Example 3 for an exemplary method of detecting AstV-VA1 in a sample that may be used in some embodiments.
Amplification reactions can also quantify the amount of nucleic acid in a sample, label the nucleic acid (e.g., to apply it to an array or a blot), or detect the nucleic acid. Amplification methods are well known in the art, and include, e.g., polymerase chain reaction (PCR); ligase chain reaction (LCR); transcription amplification; self-sustained sequence replication; Q-Beta replicase amplification; automated Q-Beta replicase amplification assay; and other RNA polymerase mediated techniques.
Antibodies that specifically bind a polypeptide sequence encoded by ORF1a, ORF1b, and ORF2 or portions thereof are included in the invention. An antibody or an antibody fragment is specific for a polypeptide of the AstV-VA1 virus if it permits one of skill in the art to discern the presence of the virus or a protein or peptide encoded by the virus in a sample, and is not cross-reactive with non-AstV-VA1 viral antigens. For detection antibodies include polyclonal, monoclonal, bi-specific, multi-specific, single chain antibodies, diabodies, nanobodies, single domain antibodies (e.g., camel antibodies), Fab, F(ab′)2, Fvs, intrabodies and fragments containing either a VL or VH domain or even a complementary determining region (CDR) that specifically binds to a polypeptide of the AstV-VA1 virus disclosed herein. Human, humanized and chimeric forms are also included in the invention as these are most useful for passive immunization. Particularly preferred for this application are neutralizing antibodies that can be identified using culture testing or other means.
Antibodies useful in the detection of AstV-VA1 virus can be labeled with any suitable detectable label and employed in assays such as enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence.
Furthermore, provided herein are immunological compositions comprising the virus or portions thereof and a pharmaceutically acceptable carrier. Thus, some compositions can comprise one or more isolated proteins from the AstV-VA1 virus, live AstV-VA1 virus, attenuated AstV-VA1 virus, or inactivated AstV-VA1 virus.
Methods for detecting the presence or absence of a polypeptide or nucleic acid from the AstV-VA1 virus in a biological sample comprises contacting the sample with a compound or an agent capable of detecting an epitope on a protein or nucleic acid (e.g., mRNA, genomic DNA) of the AstV-VA1 virus such that the presence of AstV-VA1 virus is detected in the sample.
Another aspect provides for methods of detecting an antibody, which immunospecifically binds to the AstV-VA1 virus, in a biological sample, for example blood, plasma or serum. In one embodiment, the method comprising contacting the sample with the polypeptides or protein encoded by the nucleotide sequence of AstV-VA1 virus, or a characteristic portion thereof, directly immobilized on a substrate and detecting the virus-bound antibody directly or indirectly by a labeled heterologous anti-isotype antibody.
Kits for detecting the presence of AstV-VA1 virus, or a nucleic acid polypeptide thereof or antibody thereto are included. Kits can also include instructions for use.
For antibody-based kits, the kit can comprise, for example: (1) a first antibody (e.g., attached to a solid support) which binds to a polypeptide or epitope encoded by the AstV-VA1 virus; and, optionally, (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
For oligonucleotide-based kits, the kit can comprise, for example: (1) an oligonucleotide, e.g., a detectably labeled oligonucleotide, which hybridizes to a nucleic acid sequence contained within SEQ ID NO:1 or to a sequence within the AstV-VA1 viral genome and/or (2) a pair of primers useful for amplifying a nucleic acid molecule containing an AstV-VA1 viral sequence. The kit can also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent. The kit can also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate). The kit can also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained. Each component of the kit is usually enclosed within an individual container and all of the various containers are within a single package along with instructions for use.
Yet another aspect provides a method of screening for anti-viral agents useful in reducing the symptoms of AstV-VA1 infections comprising: contacting a cell infected with the human astrovirus AstV-VA1 with a candidate anti-viral agent; assaying the anti-viral agent activity by determining the effect of the agent upon viral titer in the cell, and identifying the agent as an anti-viral agent if it inhibits viral replication, expression, or activity. The methods can be designed to screen for agents in in vitro assays against cell lines infected with the virus, against cells producing an enzyme from a virus or against a purified viral enzyme. Alternatively, the agents may be screened in in vivo assays where the virus is hosted by a mammal.
The availability of the protease and polymerase encoded by AstV-VA1 also offers a screening tool since the inhibitors of the activity of these proteins may be useful in controlling infection.
Identified anti-viral agent can prevent or inhibit the binding of the virus or viral proteins to a host cell under a physiological condition, thereby preventing or inhibiting the infection of the host cell by the virus. Anti-viral agents may prevent or inhibit replication of the viral nucleic acid molecules in the host cell under a physiological condition by interacting with the viral nucleic acid molecules or its transcription mechanisms. The antiviral agent may also inhibit the activity of essential viral proteins as set forth above.
Test viral inhibitory molecules also can be obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs.
The following examples are offered to illustrate but not to limit the invention.
Example 1 Genome Sequencing and Analysis
Five fecal specimens (labeled A, B, C, D and E) were collected from a gastroenteritis outbreak at a child care center in Virginia. Symptoms included vomiting and/or diarrhea. The five fecal specimens (A-E) (Table 1) tested negative for enteric parasites, enteric bacteria by standard microscopy and culture, and negative for enteric viruses including rotavirus (RotaClone EIA), norovirus, sapovirus, human astrovirus, and adenovirus gp F by (RT)-PCR.
TABLE 1
Epidemiologic Data of the 5 Specimens from a Child
Care Center Outbreak of Acute Gastroenteritis.
Sample ID Sex Age Onset Date Sample Date Symptoms
A M  2 years Aug. 19, 2008 Aug. 19, 2008 Diarrhea,
vomiting
B F 36 years Aug. 26, 2008 Aug. 28, 2008 Diarrhea,
vomiting
C M  6 months Aug. 25, 2008 Aug. 25, 2008 Diarrhea
D M 19 months Aug. 5, 2008 Aug. 26, 2008 Diarrhea
E Unknown 20 months Aug. 5, 2008 Aug. 27, 2008 Diarrhea
Shotgun sequencing of fecal specimens. The fecal specimens were sequenced in parallel at Washington University and at the CDC. At Washington University, the specimens were diluted in PBS at a 1:6 ratio (w/v) and total nucleic acid was extracted from 200 μL of each fecal suspension using the MAGNAPURE™ LC Automated Nucleic Acid Extraction System (Roche). Total nucleic acid was randomly amplified using the Round AB protocol as previously described with the exception that each sample was independently amplified with a different modified primer B containing a unique 6-nucleotide barcode at the 5′ end of the primer (Wang, D., et al., PLoS Biol (2003) 1:E2). Amplification products from multiple samples were pooled, adaptor-ligated, and sequenced using the Roche GS-FLX™ titanium platform (Roche) at the Washington University Genome Sequencing Center.
Sequences from each sample were identified by the unique barcodes introduced during the Rd B amplification. Primer and barcode sequences were then trimmed off prior to analysis of the sequences. Sequences were clustered using CD-HIT (Li, W., et al., Bioinformatics (2006) 22:1658-1659) to reduce redundancy with the requirement that they had to be 98% identical over 98% of their lengths. The longest sequence from each cluster was selected for inclusion in the pool of unique sequences to be analyzed. Unique sequences were filtered for repetitive sequences and compared with the human genome using BLASTn with an e-value cutoff of 1e-10. Sequences without significant similarity to the human genome were then compared to the GenBank nucleic acid database using BLASTn (cutoff: 1e-10) and tBLASTx (cutoff: 1e-5), and remaining sequences without significant hits to sequences in the database were then compared to the NCBI All Viral Genome database located on the World Wide Web using tBLASTx (cutoff: 1e-5) (Altschul, S. F., Nucleic Acids Res. (1997) 25:3389-3402). Overlapping sequences with significant sequence identity were assembled into contigs using Newbler (454 Life Sciences) or CAP3 (Huang, X., et al., Genome Res. (1999) 9:969-877).
At CDC, 10% fecal suspensions were first clarified by centrifugation at 6,000×g for 10 minutes and the supernatant was then filtered through a 0.22-um filter (ULTRAFREE® MC; Millipore, Bedford, Mass.). Total nucleic acid (TNA) was extracted from 200 μl of the cleared supernatant fluid with the QIAAMP® MINELUTE™ Virus Spin kit (QIAGEN, Valencia, Calif.) according to the manufacturer's instructions. After elution from the column in 50 μl of RNase-free water, TNA was randomly amplified using the Round AB protocol as previously described. The 300-800 bp amplicons were then cloned using the TOPO TA CLONING® kit (Invitrogen, Carlsbad, Calif.) and plasmids were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit on an ABI PRISM® 3130 automated sequencer (Applied Biosystems, Foster City, Calif.). Sequence analysis and generation of contigs were performed using Sequencher software (Ann Arbor, Mich., USA). Sequence identification was performed through NCBI nucleotide-nucleotide BLASTn similarity searches. In addition, a set of eight overlapping RT-PCR products with an average size of 900 bp which cover the entire genome including the 3′ end poly A tail were generated by primer pairs designed from clone sequences as described above, using the SUPERSCRIPT™ III First-Strand Synthesis System for RT-PCR and ACCUPRIME™ High Fidelity Taq DNA polymerase (Invitrogen, Carlsbad, Calif., USA). Both strands of each amplicon were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit as described above. The 5′ end genome sequence was amplified and determined using the 5′/3′ RACE Kit (Roche, Mannheim, Germany) following the manufacturer's instructions. The complete genome sequence of AstV-VA1 has been deposited in Genbank and is shown in FIG. 2 as SEQ ID NO:1.
Following high throughput pyrosequencing of RNA and DNA extracted from samples A, B, C and D (average of 12,730 reads per sample), 313 unique high quality sequence reads were found in sample B and 1,017 unique high quality reads were found in sample C most closely related to astroviruses. A 6,376 nucleotide (nt) contig was assembled from the astrovirus-like sequences detected in sample B and 4 contigs totaling 6,026 nucleotides were assembled from sample C. The translated contigs had only limited sequence similarity (37-71% aa identity) to proteins from mink and ovine astroviruses. The five original contigs were then assembled to generate a larger contig of 6,581 nucleotides in length.
Complete genome sequencing and genome analysis. Independently, four of the five fecal samples (stool samples A, B, C and E) were analyzed by Sanger sequencing. Three out of 96 clones from sample B and 69 out of 152 clones from sample C contained sequence signatures that were most closely related to previously known astroviruses by BLASTn similarity searches. Sequencing of 100 clones each from samples A and E yielded no clones with detectable similarity to astroviruses. The sequences of the 69 clones from sample C were assembled into 4 contigs. Primers were then designed to generate a series of eight overlapping RT-PCR amplicons with an average size of ˜900 bp that yielded a genomic sequence of 6,537 nt. In order to define the 5′ end of the genome, three independent 5′RACE reactions were performed and a total of 23 clones from these reactions were sequenced. All clones extended the genome by 49 nt and yielded the identical 5′end sequence, suggesting that the genome was complete with a total length of 6,586 nt, excluding the poly-A tail. Comparison of the genome sequences generated by the two sequencing methods yielded nearly identical sequences, with the exception of 5 missing nucleotides at the 5′ end of the contig generated by pyrosequencing and 3 nucleotide substitution differences. These were resolved by direct PCR sequencing to generate the final, corrected sequence. This virus has been provisionally named Astrovirus VA1 (AstV-VA1).
Open Reading Frame Analysis. The genome of AstV-VA1 had three predicted open reading frames (ORF 1a, 1b, and 2) as well as non-translated regions (NTRs) at both the 5′ and 3′ ends of the genome. Open reading frames (ORFs) 1a and 2 were predicted by the NCBI ORF Finder located on the World Wide Web. The end of ORF1b was also predicted by NCBI ORF Finder, however the start of ORF1b was predicted based on the location of the heptameric slippery sequence found in other astroviruses. Protein motifs were identified by conserved domain searches using BlastX and Pfam.
Several conserved protein motifs were identified including a serine protease in ORF1a, an RNA dependent RNA polymerase in ORF1b, and capsid protein in ORF 2. ORFs 1a and 2 were predicted by the NCBI ORF Finder program; however the full coding region for ORF1b was not predicted by the program because translation of ORF1b is dependent on a −1 ribosomal frameshift that occurs during translation. This frameshift is thought to be mediated by the presence of a heptameric ‘slippery sequence’ (AAAAAAAC) near the end of ORF1a, which was also conserved in the AstV-VA1 sequence, suggesting that this new virus follows the same paradigm. The sequence AUUUGGAGNGGNGGACCNAAN5-8AUGNC (SEQ ID NO:12) located upstream of ORF2, which has been proposed as the promoter for subgenomic RNA synthesis in all previously known astroviruses, is also present in AstV-VA1 with only 2 nt differences. The predicted size for each of the open reading frames is 2,661 nt, 1,575 nt, and 2,277 nt for ORFs 1a, 1b, and 2, respectively. These sizes are similar to the ORF sizes of mink and ovine astroviruses (Table 2).
TABLE 2
Genome Comparison of AstV-VA1 to Other Astroviruses.
Genome 5′ NTR ORF1a ORF1b ORF2 3′ NTR
Virus (bp) (nt) (nt) (nt) (nt) (nt)
Chicken AstV-1 6,927 15 3,017 1,533 2,052 305
Turkey AstV-1 7,003 11 3,300 1,539 2,016 130
Turkey AstV-2 7,325 21 3,378 1,584 2,175 196
Mink AstV 6,610 26 2,648 1,620 2,328 108
Ovine AstV 6,440 45 2,580 1,572 2,289 59
Human AstV-1 6,813 85 2,763 1,560 2,361 80
Human AstV-2 6,828 82 2,763 1,560 2,392 82
Human AstV-4a 6,723 84 2,763 1,548 2,316 81
Human AstV-5a 6,762 83 2,763 1,548 2,352 86
Human AstV-8 6,759 83 2,766 1,557 2,349 85
AstV-MLB1 6,172 58 2,364 1,536 2,271 58
AstV-VA1 6,586 38 2,661 1,575 2,277 98
aNumbers were deduced from the full length sequences
The 5′ non-translated region (NTR) of AstV-VA1 is 38 nt in length, which is between the lengths of the 5′ NTRs of mink astrovirus (26 nt) and ovine astrovirus (45 nt). The 3′ NTR is 98 nt in length, which again is intermediate between the length of the NTRs of ovine astrovirus (59 nt) and mink astrovirus (108 nt). The 3′ NTR of nearly all astroviruses contains a highly conserved RNA secondary structure called the stem-loop II-like motif (s2m), which has also been identified in several coronaviruses and in equine rhinovirus 2. An alignment of the 150 nt just upstream of the poly-A tail of AstV-VA1 along with the 3′ terminal sequences of other astroviruses known to contain the s2m motif indicated that AstV-VA1 contains the highly conserved ˜33 nucleotide core of the s2m motif, with 100% identity to other astroviruses in this region. The exact role of this motif is not understood; however its presence in multiple viral families suggests it may play an important role in the replication of these viruses.
Example 2 Phylogenetic Analysis of the Astrovirus VA1 Open Reading Frames
ClustalX (1.83) was used to carry out multiple sequence alignments of the protein sequences associated with all three of the open reading frames of astroviruses for which sequences were available. Maximum parsimony trees were generated using PAUP with 1,000 bootstrap replicates (Swofford, D. L., PAUP*. Phylogenetic Analysis Using Parsimony (*and Other Methods), Sunderland, Mass.: Sinauer Associates (1998)). Available nucleotide or protein sequences of the following astroviruses were obtained: Human Astrovirus 1 [GenBank: NC001943]; Human Astrovirus 2 [GenBank: L13745]; Human Astrovirus 3 [GenBank: AAD17224]; Human Astrovirus 4 [GenBank: DQ070852]; Human Astrovirus 5 [GenBank: DQ028633]; Human Astrovirus 6 [EMBL: CAA86616]; Human Astrovirus 7 [Gen Bank: AAK31913]; Human Astrovirus 8 [GenBank: AF260508]; Turkey Astrovirus 1 [GenBank: Y15936]; Turkey Astrovirus 2 [GenBank: NC005790]; Turkey Astrovirus 3 [GenBank: AY769616]; Chicken Astrovirus [GenBank: NC003790]; Ovine Astrovirus [GenBank: NC002469]; Mink Astrovirus [GenBank: NC004579], Astrovirus MLB1 [GenBank: NC011400], and Bat Astrovirus [GenBank: EU847155]. Bioedit was used to determine the percent identity between sequences as determined by pair-wise alignments.
The maximum parsimony trees confirmed that AstV-VA1 was highly divergent from, but most closely related to mink and ovine astrovirus in all three ORFs (FIGS. 1A-1C). Furthermore, the greatest sequence identity between AstV-VA1 and mink and ovine astroviruses is in ORF1b with 61% amino acid identity to mink astrovirus and 62% to ovine astrovirus. The ORF1a (serine protease) coding region was more divergent with 39% and 40% amino acid identity with ovine astrovirus and mink astrovirus, respectively. In ORF2, AstV-VA1 virus shared 41% amino acid identity to mink astrovirus and 42% to ovine astrovirus.
Detailed analysis of the viral DNA sequence and genomic organization confirmed the novelty of AstV-VA1. Complete genome sequencing and phylogenetic analysis demonstrated that AstV-VA1 was highly divergent from all previously described astroviruses including the 8 human astrovirus serotypes and recently described astrovirus MLB1 (AstV-MLB1). AstV-VA1 appears to have diverged from a common ancestor of the mink and ovine astroviruses following their separation from the branch containing human astroviruses 1-8 and astrovirus MLB1. The discovery of AstV-VA1 following the recent identification of AstV-MLB1 clearly demonstrates that a much greater diversity of astroviruses exists in humans than is commonly recognized.
Example 3 Real Time-PCR Screening for AstV-VA1
The samples collected in Example 1 were screened in order to assess the presence of AstV-VA1 in each of the samples. High throughput pyrosequencing yielded many AstV-VA1 sequences in samples B and C, but none were detected in samples A or D. Sample E was not analyzed by pyrosequencing due to technical problems with the sample preparation. Similarly, Sanger sequencing detected AstV-VA1 positive reads in samples B and C, but not in samples A and E (sample D was not initially tested). To determine whether low levels of AstV-VA1 might be present in samples A, D and E, real time RT-PCR and semi-nested RT-PCR assays were developed targeting regions in ORF1b and ORF2, respectively. Using these assays, sample D tested positive and sequencing of the 250 bp amplicon confirmed the presence of AstV-VA1.
Real Time Assay:
The real-time RT-PCR assay was performed using the SUPERSCRIPT™ III One-Step RT-PCR kit (Invitrogen Corp., Carlsbad, Calif.) and the MX4000® system (Stratagene, La Jolla, Calif.). Each 50 μl reaction mixture contained 900 pmol of forward primer (5′ TAT CCA TAG TTG TGG ATA TTT GTC CA 3′), 1,000 pmol of reverse primer (5′ TGT CTT AGG GGA GAC TTG CAA A 3′) and 100 pmol of probe (5′ TT CC CCCT GTC CTG GAT TGT CAC TTC 3′), 1× buffer, 6.0 mM MgSO4 (final concentration), 20 units of RNase inhibitor, a 5 pJ aliquot of RNA extracts, and 1 unit of SUPERSCRIPT™ III RT/PLATINUM® TAQ MIX. Water was added to achieve a final volume of 50 μl. The RT-PCR reaction mixture was incubated at 60° C. for 1 minute for denaturing, 50° C. for 30 minutes (for RT), 94° C. for 2 minutes (for hot start), then 40 cycles at 94° C. for 15 seconds; 55° C. for 30 seconds; 72° C. for 30 seconds and a final extension at 72° C. for 7 minutes. Fluorescence measurements were taken and the threshold cycle (CT) value for each sample was calculated by determining the point at which fluorescence exceeded a threshold limit set at the mean plus 10 standard deviations above the baseline.
Semi-Nested RT-PCR Assay:
The first round RT-PCR in the semi-nested assay was performed according to the protocol described previously (Tong, S., et al., J. Clin. Microbiol. (2008) 46:2652-2658) using forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) and reverse primer (5′ GTC TAT TGT TTT GGG CGT CTG C 3′). The 2nd round PCR in the semi-nested assay PCR assay in 50 μl reaction mixture contained 1× buffer (PLATINUM® Taq kit; Invitrogen), 2 mM MgCl2, 200 μM (each) of deoxynucleoside triphosphates, 50 pmol (each) of forward primer (5′ AGG GGT CGC TGG GAG TTT G 3′) and reverse primer (5′ CGG GGG TGG TGC GAC AT 3′) 1 U PLATINUM® Taq, one 2-μl aliquot from the first reaction, and water to achieve a final volume of 50 μl. The mixture was first heated to 94° C. for 2 min. The cycling conditions were 40 cycles with the same conditions as for the first amplification: 94° C. for 15 s, primer annealing at 55° C. for 30 s, and 72° C. for 30 s. A final extension was carried out at 72° C. for 7 min. The final semi-nested PCR products were visualized by UV light after electrophoresis on a 2% agarose gel containing 0.5 ug/ml ethidium bromide in 0.5×Tris-borate buffer. Amplicons from the final round of PCR were purified using the QIAQUICK® PCR purification kit (Qiagen, Inc., Valencia, Calif.). Both strands of the amplicons were sequenced with a BIGDYE® Terminators v3.1 ready reaction cycle sequencing kit as described above.
The detection of AstV-VA1 in three out of five samples of this gastroenteritis outbreak suggests a potential association between AstV-VA1 and symptomatic infection. The fact that AstV-VA1 was only detected in sample D by targeted PCR assays and not by either of the mass sequencing methods may be due to the late timing of sample acquisition relative to the onset of symptoms (Table 1). Further studies defining the frequency of detection of AstV-VA1 in additional samples from individuals with and without acute gastroenteritis are needed to define the role of AstV-VA1 in human diarrhea. It is likely that the application of sequence independent amplification and sequencing methods to other outbreaks of gastroenteritis of unknown etiology will identify other novel viruses and expand our ability to determine the cause of diarrheal disease.

Claims (6)

The invention claimed is:
1. An isolated nucleic acid molecule comprising a nucleotide sequence at least 95% identical to SEQ ID NO:1, or a complement to the entire said nucleotide sequence.
2. The isolated nucleic acid molecule of claim 1 wherein said nucleotide sequence is at least 98% identical to SEQ ID NO:1, or is a complement to the entire said nucleotide sequence.
3. The isolated nucleic acid molecule of claim 1 wherein said nucleotide sequence is identical to SEQ ID NO:1, or is a complement to the entire said nucleotide sequence.
4. A vector comprising the nucleotide sequence of claim 3 or a complement to the entire said nucleotide sequence.
5. An isolated cell comprising the vector of claim 4.
6. A method to detect the presence of AstV-VA1 in a sample which method comprises contacting the sample with a probe that specifically hybridizes under stringent conditions to the nucleic acid molecule of claim 5, or
which method comprises contacting the sample with amplification primers, amplifying nucleic acid in the sample, contacting the amplified nucleic acid with a probe that specifically hybridizes under stringent conditions to the nucleic acid molecule of claim 5,
wherein said stringent conditions are 6×SSC at about 45° C. followed by one or more washes of about 5-30 min each in 0.1×SSC, 0.1% SDS at 45-65° C.
US12/790,618 2009-05-28 2010-05-28 Identification of astrovirus VA1 associated with gastroenteritis in humans Expired - Fee Related US8426574B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/790,618 US8426574B2 (en) 2009-05-28 2010-05-28 Identification of astrovirus VA1 associated with gastroenteritis in humans

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18200809P 2009-05-28 2009-05-28
US12/790,618 US8426574B2 (en) 2009-05-28 2010-05-28 Identification of astrovirus VA1 associated with gastroenteritis in humans

Publications (2)

Publication Number Publication Date
US20110008353A1 US20110008353A1 (en) 2011-01-13
US8426574B2 true US8426574B2 (en) 2013-04-23

Family

ID=43427640

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/790,618 Expired - Fee Related US8426574B2 (en) 2009-05-28 2010-05-28 Identification of astrovirus VA1 associated with gastroenteritis in humans

Country Status (1)

Country Link
US (1) US8426574B2 (en)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Finkbeiner et al., PLoS Pathog. (2008) 4:e1000011.
Finkbeiner et al., Virol. J. (2008) 5:117.
Holtz et al., Virol. J. (2008) 5:159.
Mendez et al., Fields Virology, 5th ed., (2007) Knipe and Howley, (eds.) Philadelphia: Lippincott Williams & Wilkins, pp. 981-1000.

Also Published As

Publication number Publication date
US20110008353A1 (en) 2011-01-13

Similar Documents

Publication Publication Date Title
Jiang et al. Comparison of novel MLB-clade, VA-clade and classic human astroviruses highlights constrained evolution of the classic human astrovirus nonstructural genes
Günther et al. Imported lassa fever in Germany: molecular characterization of a new lassa virus strain.
Thoelen et al. Molecular typing and epidemiology of enteroviruses identified from an outbreak of aseptic meningitis in Belgium during the summer of 2000
Okada et al. Molecular epidemiology and phylogenetic analysis of Sapporo-like viruses
Pecora et al. First finding of genetic and antigenic diversity in 1b-BVDV isolates from Argentina
Candresse et al. Association of Little cherry virus 1 (LChV1) with the Shirofugen stunt disease and characterization of the genome of a divergent LChV1 isolate
Abozeid et al. Complete genome sequences of two avian infectious bronchitis viruses isolated in Egypt: evidence for genetic drift and genetic recombination in the circulating viruses
Sozzi et al. Molecular characterization and phylogenetic analysis of VP1 of porcine enteric picornaviruses isolates in Italy
Wolf et al. Discovery and genetic characterization of novel caliciviruses in German and Dutch poultry
Diab et al. Foot-and-mouth disease outbreaks in Egypt during 2013-2014: Molecular characterization of serotypes A, O, and SAT2
US7320857B2 (en) Characterization of the earliest stages of the severe acute respiratory syndrome (SARS) virus and uses thereof
Giordano et al. Evidence of closely related picobirnavirus strains circulating in humans and pigs in Argentina
De Mia et al. Genetic characterization of a caprine pestivirus as the first member of a putative novel pestivirus subgroup
Mirosław et al. Increased genetic variation of bovine viral diarrhea virus in dairy cattle in Poland
Agostini et al. Molecular evolution and epidemiology of JC virus
Marais et al. Characterization of the virome of shallots affected by the shallot mild yellow stripe disease in France
Nguyen et al. Genetic diversity and molecular characterization of classical swine fever virus envelope protein genes E2 and Erns circulating in Vietnam from 2017 to 2019
Dey et al. Prevalence of sapovirus infection among infants and children with acute gastroenteritis in Dhaka City, Bangladesh during 2004–2005
Lu et al. Complete sequence of a Kyrgyzstan swine hepatitis E virus (HEV) isolated from a piglet thought to be experimentally infected with human HEV
Ozoemena et al. Comparative evaluation of measles virus specific TaqMan PCR and conventional PCR using synthetic and natural RNA templates
Hudu et al. Hepatitis E virus isolated from chronic hepatitis B patients in Malaysia: Sequences analysis and genetic diversity suggest zoonotic origin
US8426574B2 (en) Identification of astrovirus VA1 associated with gastroenteritis in humans
EP3119429A1 (en) A non-naturally occuring porcine reproductive and respiratory syndrome virus (prrsv) and methods of using
Desport et al. Sequence analysis of Jembrana disease virus strains reveals a genetically stable lentivirus
Cabral et al. Simple protocol for population (Sanger) sequencing for Zika virus genomic regions

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GOVERNMENT OF THE UNITED STATES OF AMERICA, SE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TONG, SUXIANG;VINJE, JAN;LI, YAN;SIGNING DATES FROM 20090707 TO 20090805;REEL/FRAME:025023/0749

Owner name: WASHINGTON UNIVERSITY, MISSOURI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WANG, DAVID;ZHAO, GUOYAN;VIRGIN IV, HERBERT W.;AND OTHERS;SIGNING DATES FROM 20090317 TO 20090513;REEL/FRAME:025023/0760

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:WASHINGTON UNIVERSITY;REEL/FRAME:042125/0708

Effective date: 20170323

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20210423