WO2010110914A2 - Mammalian genes involved in infection - Google Patents

Mammalian genes involved in infection Download PDF

Info

Publication number
WO2010110914A2
WO2010110914A2 PCT/US2010/000916 US2010000916W WO2010110914A2 WO 2010110914 A2 WO2010110914 A2 WO 2010110914A2 US 2010000916 W US2010000916 W US 2010000916W WO 2010110914 A2 WO2010110914 A2 WO 2010110914A2
Authority
WO
WIPO (PCT)
Prior art keywords
virus
infection
pathogen
gene product
gene
Prior art date
Application number
PCT/US2010/000916
Other languages
French (fr)
Other versions
WO2010110914A3 (en
Inventor
Natalie Mcdonald
Original Assignee
Zirus, Inc.
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 Zirus, Inc. filed Critical Zirus, Inc.
Publication of WO2010110914A2 publication Critical patent/WO2010110914A2/en
Publication of WO2010110914A3 publication Critical patent/WO2010110914A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of one or more pathogens, such as a virus, a bacteria, a fungus or a parasite.
  • pathogens such as a virus, a bacteria, a fungus or a parasite.
  • viruses that interfere with normal cellular processes. These include influenza, poliomyelitis, smallpox, Ebola, yellow fever, measles and AIDS, to name a few. Viruses are also responsible for many cases of human disease including encephalitis, meningitis, pneumonia, hepatitis and cervical cancer, warts and the common cold. Furthermore, viruses causing respiratory infections, and diarrhea in young children lead to millions of deaths each year in less-developed countries. Also, a number of newly emerging human diseases such as SARS are caused by viruses. In addition, the threat of a bioterrorist-designed pathogen is ever present.
  • the present invention provides genes and gene products set forth in Tables 1, 2, 3 and 4 that are involved in infection by one or more pathogens such as a virus, a parasite, a bacteria or a fungus, or are otherwise associated with the life cycle of a pathogen. Also provided are methods of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more of these genes or gene products set forth in Tables 1, 2, 3 and 4. Also provided are methods of decreasing infection by a pathogen in a subject by administering an agent that decreases the expression and/or activity of of the genes or gene products set forth in Tables 1, 2, 3 and 4. Further provided are methods of identifying an agent that decreases infection by a pathogen.
  • pathogens such as a virus, a parasite, a bacteria or a fungus
  • Ranges may be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. "Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, the phrase “optionally obtained prior to treatment” means obtained before treatment, after treatment, or not at all.
  • subject is meant an individual.
  • the subject is a mammal such as a primate, and, more preferably, a human.
  • Non-human primates include marmosets, monkeys, chimpanzees, gorillas, orangutans, and gibbons, to name a few.
  • subject includes domesticated animals, such as cats, dogs, etc., livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, ferret, chinchilla, mouse, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, chickens, turkeys, ducks, pheasants, pigeons, doves, parrots, cockatoos, geese, etc.).
  • livestock for example, cattle, horses, pigs, sheep, goats, etc.
  • laboratory animals for example, ferret, chinchilla, mouse, rabbit, rat, gerbil, guinea pig, etc.
  • avian species for example, chickens, turkeys, ducks, pheasants, pigeons, doves, parrots, cockatoos, geese, etc.
  • the subjects of the present invention can also include, but are not limited
  • genes listed in Table 1 , 2, 3 or 4 are host genes involved in viral infection.
  • gene product is the RNA or protein resulting from the expression of a gene listed in Table 1, 2, 3 or 4.
  • the nucleic acids of these genes and their encoded proteins can be involved in all phases of the viral life cycle including, but not limited to, viral attachment to cellular receptors, viral infection, viral entry, internalization, disassembly of the virus, viral replication, genomic integration of viral sequences, transcription of viral RNA, translation of viral mRNA, transcription of cellular proteins, translation of cellular proteins, trafficking, proteolytic cleavage of viral proteins or cellular proteins, assembly of viral particles, budding, cell lysis and egress of virus from the cells.
  • genes set forth herein were identified as cellular genes involved in viral infection, as discussed throughout, the present invention is not limited to viral infection.
  • any of these nucleic acid sequences and the proteins encoded by these sequences can be involved in infection by any infectious pathogen such as a bacteria, a fungus or a parasite which includes involvement in any phase of the infectious pathogen's life cycle.
  • ABTBl when referring to any of the genes in Table 1, 2, 3 or 4, for example, and not to be limiting, ABTBl , this includes any ABTBl gene, ABTBl gene product, for example, an ABTBl nucleic acid (DNA or RNA) or ABTBl protein, from any organism that retains at least one activity of ABTBl and can function as an ABTBl nucleic acid or protein utilized by a pathogen.
  • the nucleic acid or protein sequence can be from or in a cell in a human, a non-human primate, a mouse, a rat, a cat, a dog, a chimpanzee, a horse, a cow, a pig, a sheep, a guinea pig, a rabbit, a zebrafish, a chicken, to name a few.
  • a gene is a nucleic acid sequence that encodes a polypeptide under the control of a regulatory sequence, such as a promoter or operator.
  • the coding sequence of the gene is the portion transcribed and translated into a polypeptide (in vivo, in vitro or in situ) when placed under the control of an appropriate regulatory sequence.
  • the boundaries of the coding sequence can be determined by a start codon at the 5' (amino) terminus and a stop codon at the 3' (carboxyl) terminus. If the coding sequence is intended to be expressed in a eukaryotic cell, a polyadenylation signal and transcription termination sequence can be included 3' to the coding sequence.
  • Transcriptional and translational control sequences include, but are not limited to, DNA regulatory sequences such as promoters, enhancers, and terminators that provide for the expression of the coding sequence, such as expression in a host cell.
  • a polyadenylation signal is an exemplary eukaryotic control sequence.
  • a promoter is a regulatory region capable of binding RNA polymerase and initiating transcription of a downstream (3' direction) coding sequence.
  • a gene can include a signal sequence at the beginning of the coding sequence of a protein to be secreted or expressed on the surface of a cell. This sequence can encode a signal peptide, N-terminal to the mature polypeptide, which directs the host cell to translocate the polypeptide.
  • Tables 1 , 2, 3 and 4 provide the Entrez Gene numbers for the human genes set forth herein.
  • the information provided under the Entrez Gene numbers listed in Tables 1 , 2, 3 and 4 is hereby incorporated entirely by this reference.
  • Entrez Gene By accessing Entrez Gene, one of skill in the art can readily obtain information about every gene listed in Table 1, 2, 3 or 4, such as the genomic location of the gene, a summary of the properties of the protein encoded by the gene, expression patterns, function, information on homologs of the gene as well as numerous reference sequences, such as the genomic, mRNA and protein sequences for each gene. Therefore, one of skill in the art can readily obtain sequences, such as genomic, mRNA and protein sequences by accessing information available under the Entrez Gene number provided for each gene. Thus, all of the information readily obtained from the Entrez Gene Nos. set forth herein is also hereby incorporated by reference in its entirety.
  • the proteins encoded by the genes are also listed in column 3 of Table 5. In addition to the function of being involved in pathogenic infection as provided herein, a function of the proteins is also provided, if available, in column 4 of Table 5.
  • the chromosomal location of the gene in the human genome (column 5) is also set forth.
  • the present invention identifies a genomic loci of genes associated with viral infection. By identifying the gene and its location in the genome, the invention provides both the gene and its product(s) as targets for therapies such as antiviral, antibacterial, antifungal and antiparasitic therapies, to name a few. Also provided in Table 5 are the GenBank Accession Nos. for the coding sequences
  • nucleic acid refers to single or multiple stranded molecules which may be DNA or RNA, or any combination thereof, including modifications to those nucleic acids.
  • the nucleic acid may represent a coding strand or its complement, or any combination thereof.
  • Nucleic acids may be identical in sequence to the sequences which are naturally occurring for any of the moieties discussed herein or may include alternative codons which encode the same amino acid as that which is found in the naturally occurring sequence. These nucleic acids can also be modified from their typical structure.
  • Such modifications include, but are not limited to, methylated nucleic acids, the substitution of a non-bridging oxygen on the phosphate residue with either a sulfur (yielding phosphorothioate deoxynucleotides), selenium (yielding phosphorselenoate deoxynucleotides), or methyl groups (yielding methylphosphonate deoxynucleotides), a reduction in the AT content of AT rich regions, or replacement of non-preferred codon usage of the expression system to preferred codon usage of the expression system.
  • the nucleic acid can be directly cloned into an appropriate vector, or if desired, can be modified to facilitate the subsequent cloning steps.
  • the sequence encoding the specific amino acids can be modified or changed at any particular amino acid position by techniques well known in the art.
  • PCR primers can be designed which span the amino acid position or positions and which can substitute any amino acid for another amino acid.
  • one skilled in the art can introduce specific mutations at any point in a particular nucleic acid sequence through techniques for point mutagenesis.
  • General methods are set forth in Smith, M. "In vitro mutagenesis” Ann. Rev. Gen., 19:423-462 (1985) and Zoller, M.J. "New molecular biology methods for protein engineering” Curr. Opin. Struct. Biol., 1 :605-610 (1991), which are incorporated herein in their entirety for the methods. These techniques can be used to alter the coding sequence without altering the amino acid sequence that is encoded.
  • sequences contemplated herein include full-length wild-type (or native) sequences, as well as allelic variants, variants, fragments, homologs or fusion sequences that retain the ability to function as the cellular nucleic acid or protein involved in viral infection.
  • a protein or nucleic acid sequence has at least 50% sequence identity, for example at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 98% sequence identity to a native sequences of the genes set forth in any of tables 1-4.
  • a nucleic acid sequence involved in viral infection has a sequence that hybridizes to a sequence of a gene set forth in Table 1, 2 3 or 4 and retains the activity of the sequence of the gene set forth in Table 1, 2 3, or 4.
  • a nucleic acid that hybridizes to an ABTBl nucleic acid sequence and encodes a protein that retains ABTBl activity is contemplated by the present invention.
  • sequences include the genomic sequence for the genes set forth in Table 1, 2, 3 or 4.
  • the examples set forth above for ABTBl are merely illustrative and should not be limited to ABTBl as the analysis set forth in this example applies to every nucleic acid and protein for the genes listed in Table 1 , 2, 3 or 4.
  • any reference to a nucleic acid molecule throughout this application includes the reverse complement of the nucleic acid.
  • any siRNA sequence set forth herein also includes the reverse complement of that sequence.
  • any nucleic acid written to depict only a single strand encompasses both strands of a corresponding double- stranded nucleic acid.
  • depiction of a plus-strand of a dsDN A also encompasses the complementary minus-strand of that dsDNA.
  • any reference to the nucleic acid molecule that encodes a specific protein, or a fragment thereof encompasses both the sense strand and its reverse complement.
  • fragments of the nucleic acids set forth in Table 1, 2, 3 or 4 are also contemplated. These fragments can be utilized as primers and probes to amplify, inhibit or detect any of the nucleic acids or genes set forth in Table 1 , 2, 3 or 4.
  • Hybridization conditions resulting in particular degrees of stringency will vary depending upon the nature of the hybridization method and the composition and length of the hybridizing nucleic acid sequences. Generally, the temperature of hybridization and the ionic strength (such as the Na+ concentration) of the hybridization buffer will determine the stringency of hybridization. Calculations regarding hybridization conditions for attaining particular degrees of stringency are discussed in Sambrook et al., (1989) Molecular Cloning, second edition, Cold Spring Harbor Laboratory, Plainview, NY (chapters 9 and 11). The following is an exemplary set of hybridization conditions and is not limiting:
  • Hybridization 5x SSC at 65°C for 16 hours Wash twice: 2x SSC at room temperature (RT) for 15 minutes each
  • Hybridization 5x-6x SSC at 65°C-70°C for 16-20 hours Wash twice: 2x SSC at RT for 5-20 minutes each Wash twice: Ix SSC at 55°C-70°C for 30 minutes each
  • Hybridization 6x SSC at RT to 55°C for 16-20 hours Wash at least twice: 2x-3x SSC at RT to 55°C for 20-30 minutes each.
  • a vector comprising a nucleic acid set forth herein.
  • the vector can direct the in vivo or in vitro synthesis of any of the proteins or polypeptides described herein.
  • the vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid.
  • These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene (See generally, Sambrook et al).
  • the vector for example, can be a plasmid.
  • the vectors can contain genes conferring hygromycin resistance, ampicillin resistance, gentamicin resistance, neomycin resistance or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification.
  • E. coli Esscherichia coli
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species.
  • bacilli such as Bacillus subtilis
  • enterobacteriaceae such as Salmonella, Serratia, and various Pseudomonas species.
  • prokaryotic hosts one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication).
  • any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (Trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • yeast expression can be used.
  • the invention provides a nucleic acid encoding a polypeptide of the present invention, wherein the nucleic acid can be expressed by a yeast cell. More specifically, the nucleic acid can be expressed by Pichia past or is or S. cerevisiae.
  • Mammalian cells also permit the expression of proteins in an environment that favors important post-translational modifications such as folding and cysteine pairing, addition of complex carbohydrate structures, and secretion of active protein.
  • Vectors useful for the expression of active proteins are known in the art and can contain genes conferring hygromycin resistance, genticin or G418 resistance, or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification.
  • a number of suitable host cell lines capable of secreting intact human proteins have been developed in the art, and include the CHO cell lines, HeLa cells, COS-7 cells, myeloma cell lines, Jurkat cells, etc.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • expression control sequences such as an origin of replication, a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, etc.
  • the expression vectors described herein can also include nucleic acids of the present invention under the control of an inducible promoter such as the tetracycline inducible promoter or a glucocorticoid inducible promoter.
  • the nucleic acids of the present invention can also be under the control of a tissue-specific promoter to promote expression of the nucleic acid in specific cells, tissues or organs.
  • Any regulatable promoter, such as a metal lothionein promoter, a heat-shock promoter, and other regulatable promoters, of which many examples are well known in the art are also contemplated.
  • a Cre-loxP inducible system can also be used, as well as a FIp recombinase inducible promoter system, both of which are known in the art.
  • Insect cells also permit the expression of mammalian proteins. Recombinant proteins produced in insect cells with baculovirus vectors undergo post-translational modifications similar to that of wild-type proteins.
  • the invention also provides for the vectors containing the contemplated nucleic acids in a host suitable for expressing the nucleic acids.
  • the host cell can be a prokaryotic cell, including, for example, a bacterial cell. More particularly, the bacterial cell can be an E. coli cell.
  • the cell can be a eukaryotic cell, including, for example, a Chinese hamster ovary (CHO) cell, a COS-7 cell, a HELA cell, an avian cell, a myeloma cell, a Pichia cell, or an insect cell.
  • CHO Chinese hamster ovary
  • COS-7 COS-7
  • HELA HELA
  • avian cell avian
  • myeloma cell a cell line suitable for infection by a pathogen
  • tumor cell lines such as melanoma cell lines.
  • the vectors containing the nucleic acid segments of interest can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host.
  • the present invention provides isolated polypeptides comprising the polypeptide or protein sequences for the genes set forth in Table 1, 2, 3 or 4.
  • the present invention also provides fragments of these polypeptides. These fragments can be of sufficient length to serve as antigenic peptides for the generation of antibodies.
  • the present invention also contemplates functional fragments that possess at least one activity of a gene or gene product listed in Table 1, 2, 3 or 4, for example, involved in viral infection.
  • isolated polypeptide or “purified polypeptide” is meant a polypeptide that is substantially free from the materials with which the polypeptide is normally associated in nature or in culture.
  • the polypeptides of the invention can be obtained, for example, by extraction from a natural source if available (for example, a mammalian cell), by expression of a recombinant nucleic acid encoding the polypeptide (for example, in a cell or in a cell-free translation system), or by chemically synthesizing the polypeptide.
  • a polypeptide can be obtained by cleaving full length polypeptides. When the polypeptide is a fragment of a larger naturally occurring polypeptide, the isolated polypeptide is shorter than and excludes the full-length, naturally-occurring polypeptide of which it is a fragment.
  • polypeptide comprising an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the native polypeptide sequence for any gene set forth in Table 1, 2, 3 or 4.
  • variants of nucleic acids and polypeptides herein disclosed typically have at least, about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent homology to the stated sequence or the native sequence.
  • Those of skill in the art readily understand how to determine the homology of two polypeptides or nucleic acids.
  • the present invention provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is not HIV.
  • the present invention provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of, BCR, CA2, CHRNA7, CXCR4, EDNRA, EGFR, F2, GRIA2, HDACl, KCNJl 1, MMP9, PDE8A, PLKl, POLE, SRD5A2, TUBA8 or DPP4, wherein the pathogen is not HIV.
  • imatinib can be used to inhibit BCR.
  • Acetazolamide, benzothiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate or trichloromethiazide can be used to inhibit CA2.
  • Amobarbitol, atracurium, cisatracurium, D-tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enflurane or isoflurane can be used to inhibit CHRNA7.
  • Plerixafor can be used to inhibit CXCR4.
  • Ambrisentan, avosentan, bosentan, BSF 302146, clazosentan, PD 180988, SB 234551, sitaxsentan, TBC 3214, ZD4054 or atrasentan can be utilized to inhibit EDNRA.
  • AEE 788, ARRY-334543, BMS-599626, canertinib, cetuximab, erlotinib, gefitinib, HKI-272, lapatinib, PD 153035, vandetanib or XL647 can be used to inhibit EGFR.
  • Antithrombin, alfa, argatroban, bivalirudin, dabigatran etexilate, desirudil, enoxaparin, or lepirudin can be utilized to inhibit F2.
  • Talampanel or tezampanel can be utilized to inhibit
  • GRIA2 MGCD0103, PXDlOl, pyroxamide, romidepsin, tributyrin or vorinostat can be utilized to inhibit HDACl .
  • Acetohexamide, chlorpropamide, glimepiride, glipizide, glyburide, minoxidil, nateglinide, phentolamine, repaglinide, tolazamide or tolbutamide can be utilized to inhibit KCNJl 1.
  • Aminophylline, anagrelide, dipyridamole, dyphylline, milrinone, nitroglycerin, pentoxifylline, theophylline or tolbutamide can be utilized to inhibit PDE8A.
  • BI2536 can be utilized to inhibit PLKl .
  • Clofarabine, gemcitabine, nelarabine or trifluridine can be utilized to inhibit POLE.
  • Dutasteride or finasteride can be utilized to inhibit SRD5A2.
  • Docetaxel, ixabepilone, milataxel, NPI-2358, podophyllotoxin, TPI 287, AL 108, paclitaxel, TTI-237, XRP9881 (larotaxel), EC145, E7389, vinblastine, vincristine, vinflunine or vinorelbine can be utilized to inhibit TUBA8.
  • Saxagliptin, sitagliptin, SYR-322 or talabostat can be utilized to inhibit DPP4.
  • Minocycline can be utilized to inhibit MMP9.
  • Combinations of two or more of the compounds set forth can be utilized to inhibit infection. These combinations can be utilized to inhibit one or more of the gene products selected from the group consisting of BCR, CA2, CHRNA7, CXCR4, EDNRA, EGFR, F2, GRI A2, HDACl, KCNJl 1, PDE8A, PLKl, POLE, SRD5A2, TUBA8 and DPP4. Therefore, two or more compounds that inhibit expression or activity of the same gene product can be used, or two or more compounds that inhibit expression or activity of two or more different gene products can be used to inhibit infection.
  • Aminophylline, amrinone, cilostazol, dipyridamole, dyphylline, medorinone, nitroglycerin, pentoxifylline, theophylline or tolbutamide can be utilized to inhibit PDE3B.
  • a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 3, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
  • a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of ADRAlD, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 4, wherein the pathogen is not hepatitis C virus.
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or STAT (SOATl) or TBXA2R, wherein the pathogen is not hepatitis C virus.
  • Pactimibe can be utilized to inhibit STAT (SOATl).
  • S-18886 or torsemide can be utilized to inhibit TBXA2R.
  • Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, wherein the pathogen is not HIV. Further provided is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is not West Nile Virus. Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
  • Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, wherein the pathogen is not HCV.
  • one or more includes, two or more, three or more, four or more, five or more, six or more, seven or more, etc.
  • an infection can be a viral infection, bacterial infection, fungal infection or a parasitic infection, to name a few.
  • a decrease or inhibition of infection can occur in a cell, in vitro, ex vivo or in vivo.
  • the term "infection” encompasses all phases of pathogenic life cycles including, but not limited to, attachment to cellular receptors, entry, internalization, disassembly, replication, genomic integration of pathogenic sequences, transcription of viral RNA, translation of viral RNA, transcription of host cell mRNA, translation of host cell mRNA, proteolytic cleavage of pathogenic proteins or cellular proteins, assembly of particles, endocytosis, cell lysis, budding, and egress of the pathogen from the cells.
  • a decrease in infection can be a decrease in attachment to cellular receptors, a decrease in entry, a decrease in internalization, a decrease in disassembly, a decrease in replication, a decrease in genomic integration of pathogenic sequences, a decrease in translation of mRNA, a decrease in proteolytic cleavage of pathogenic proteins or cellular proteins, a decrease in assembly of particles, a decrease in endocytosis, a decrease in cell lysis, a decrease in budding, or a decrease in egress of the pathogen from the cells.
  • This decrease does not have to be complete as this can range from a slight decrease to complete ablation of the infection.
  • a decrease in infection can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of infection in a cell wherein expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4 has not been decreased.
  • expression can be inhibited, for example by inhibiting transcription of the gene, or inhibiting translation of its gene product.
  • the activity of a gene product for example, an mRNA, a polypeptide or a protein
  • Inhibition or a decrease in expression does not have to be complete as this can range from a slight decrease in expression to complete ablation of expression.
  • expression can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression of a gene or gene product set forth in Table 1 , 2, 3 or 4 has not been decreased or inhibited.
  • inhibition or decrease in the activity of a gene product does not have to be complete as this can range from a slight decrease to complete ablation of the activity of the gene product.
  • the activity of a gene product can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein activity of a gene or gene product set forth in Table 1, 2, 3 or 4 has not been decreased or inhibited.
  • activity of a gene product can be an activity that is involved in pathogenicity, for example, interacting directly or indirectly, with pathogen, e.g. viral protein or viral nucleic acids, or an activity that the gene product performs in a normal cell, i.e. in a non-infected cell.
  • an activity of the proteins and nucleic acids listed herein can be the ability to bind or interact with other proteins.
  • the present invention also provides a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and other cellular proteins, such as, for example, transcription factors, receptors, nuclear proteins, transporters, microtubules, membrane proteins, enzymes (for example, ATPases, phosphorylases, oxidoreductases, kinases, phosphatases, synthases, lyases, aromatases, helicases, hydrolases, proteases, transferases, nucleases, ligases, reductases and polymerases) and hormones, provided that such inhibition correlates with decreasing infection by the pathogen. Also provided is a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).
  • a viral, bacterial, parasitic or fungal protein i.e. a non-host protein
  • the cells of the present invention can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli.
  • the cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. Therefore, the cell can also be part of a population of cells.
  • the cell(s) can also be in a subject.
  • viral infections include but are not limited to, infections caused by RNA viruses (including negative stranded RNA viruses, positive stranded RNA viruses, double stranded RNA viruses and retroviruses), DNA viruses. All strains and types of RNA viruses and DNA viruses are contemplated herein.
  • RNA viruses include, but are not limited to picornaviruses, which include aphthoviruses (for example, foot and mouth disease virus O, A, C, Asia 1, SATl, SAT2 and SAT3), cardioviruses (for example, encephalomycarditis virus and Theiller's murine encephalomyelitis virus), enteroviruses (for example polioviruses 1 , 2 and 3, human enteroviruses A-D, bovine enteroviruses 1 and 2, human coxsackieviruses A1-A22 and A24, human coxsackieviruses B1-B5, human echoviruses 1-7, 9, 11-12, 24, 27, 29-33, human enteroviruses 68-71 , porcine enteroviruses 8-10 and simian enteroviruses 1-18), erboviruses (for example, equine rhinitis virus), hepatovirus (for example human hepatitis A virus and
  • RNA viruses include caliciviruses, which include noroviruses (for example, Norwalk virus), sapoviruses (for example, Sapporo virus), lagoviruses (for example, rabbit hemorrhagic disease virus and European brown hare syndrome) and vesiviruses (for example vesicular exanthema of swine virus and feline calicivirus).
  • caliciviruses include noroviruses (for example, Norwalk virus), sapoviruses (for example, Sapporo virus), lagoviruses (for example, rabbit hemorrhagic disease virus and European brown hare syndrome) and vesiviruses (for example vesicular exanthema of swine virus and feline calicivirus).
  • Other RNA viruses include astroviruses, which include mastorviruses and avastroviruses. Togaviruses are also RNA viruses.
  • Togaviruses include alphaviruses (for example, Sindbis virus, Chikungunya virus, Semliki Forest virus, Western equine encephalitis, Getah virus, Everglades virus, Venezuelan equine encephalitis virus and Aura virus) and rubella viruses.
  • alphaviruses for example, Sindbis virus, Chikungunya virus, Semliki Forest virus, Western equine encephalitis, Getah virus, Everglades virus, Venezuelan equine encephalitis virus and Aura virus
  • rubella viruses for example, Sindbis virus, Chikungunya virus, Semliki Forest virus, Western equine encephalitis, Getah virus, Everglades virus, Venezuelan equine encephalitis virus and Aura virus.
  • RNA viruses include the flaviviruses (for example, tick- borne encephalitis virus, Tyuleniy virus, Aroa virus, Dengue virus (types 1 to 4), Kedougou virus, Japanese encephalitis virus (JEV), West Nile virus (WNV), Kokobera virus, Ntaya virus, Spondweni virus, Yellow fever virus, Entebbe bat virus, Modoc virus, Rio Bravo virus, Cell fusing agent virus, pestivirus, GB virus A, GBV-A like viruses, GB virus C, Hepatitis G virus, hepacivirus (hepatitis C virus (HCV)) all six genotypes), bovine viral diarrhea virus, and GB virus B).
  • flaviviruses for example, tick- borne encephalitis virus, Tyuleniy virus, Aroa virus, Dengue virus (types 1 to 4), Kedougou virus, Japanese encephalitis virus (JEV), West Nile virus (WNV), Kokobera virus, N
  • RNA viruses are the coronaviruses which include, human respiratory coronaviruses such as SARS-CoV, HCoV-229E, HCoV-NL63 and HCoV-OC43. Coronaviruses also include bat SARS-like CoV, turkey coronavirus, chicken coronavirus, feline coronavirus and canine coronavirus. Additional RNA viruses include arteriviruses (for example, equine arterivirus, porcine reproductive and respiratory syndrome virus, lactate dehyrogenase elevating virus of mice and simian hemorraghic fever virus).
  • arteriviruses for example, equine arterivirus, porcine reproductive and respiratory syndrome virus, lactate dehyrogenase elevating virus of mice and simian hemorraghic fever virus.
  • RNA viruses include the rhabdoviruses, which include lyssaviruses (for example, rabies, Lagos bat virus, Mokola virus, Duvenhage virus and European bat lyssavirus), vesiculoviruses (for example, VSV-Indiana, VSV-New Jersey, VSV-Alagoas, Piry virus, Cocal virus, Maraba virus, Isfahan virus and Chandipura virus), and ephemeroviruses (for example, bovine ephemeral fever virus, Sydney River virus and Berrimah virus).
  • RNA viruses include the filoviruses. These include the Marburg and Ebola viruses (for example, EBOV-Z, EBOV-S, EBOV-IC and EBOV-R.
  • the paramyxoviruses are also RNA viruses.
  • these viruses are the rubula viruses (for example, mumps, parainfluenza virus 5, human parainfluenza virus type 2, Mapuera virus and porcine rubulavirus), avulaviruses (for example, Newcastle disease virus), respoviruses (for example, Sendai virus, human parainfluenza virus type 1 and type 3, bovine parainfluenza virus type 3), henipaviruses (for example, Hendra virus and Nipah virus), morbilloviruses (for example, measles, Cetacean morvilliirus, Canine distemper virus, Peste- des-petits-ruminants virus, Phocine distemper virus and Rinderpest virus), pneumoviruses (for example, human respiratory syncytial virus A2, Bl and S2, bovine respiratory syncytial virus and pneumonia virus of mice), metapneumoviruses (for example, human metapneumovirus and
  • RNA viruses include the orthomyxoviruses.
  • influenza viruses and strains e.g., influenza A (HlNl (including but not limited to A/WS/33 or A/California/04/2009 strains) H2N2, H3N2, H5N1 , H7N7, H1N2, H9N2, H7N2, H7N3 and H10N7), influenza A strain A/Victoria/3/75, influenza A strain A/Puerto Rico/8/34, influenza B strain Lee, influenza B (for example B strain Lee) and C viruses, as well as avian influenza (for example, strains H5N1, H5N2, H7N1, H7N7 and H9N2) thogotoviruses and isaviruses.
  • influenza A HlNl (including but not limited to A/WS/33 or A/California/04/2009 strains) H2N2, H3N2, H5N1 , H7N7, H1N2, H9N2,
  • Orthobunyaviruses for example, Akabane virus, California encephalitis, Cache Valley virus, Snowshoe hare virus,) nairoviruses (for example, Washington sheep virus, Crimean-Congo hemorrhagic fever virus Group and Hughes virus), phleboviruses (for example, Candiru, Punta Toro, Rift Valley Fever, Sandfly Fever, Naples, Toscana, Sicilian and Chagres), and hantaviruses (for example, Hantaan, Dobrava, Seoul, Puumala, Sin Nombre, Bayou, Black Creek Canal, Andes and Thottapalayam) are also RNA viruses.
  • phleboviruses for example, Candiru, Punta Toro, Rift Valley Fever, Sandfly Fever, Naples, Toscana, Sicilian and Chagres
  • hantaviruses for example, Hantaan, Dobrava, Seoul, Puumala, Sin Nombre,
  • Arenaviruses such as lymphocytic choriomeningitis virus, Lassa fever virus, Argentine hemorrhagic fever virus, Venezuelan hemorrhagic fever virus, SABV and WWAV are also RNA viruses.
  • Borna disease virus is also an RNA virus.
  • Hepatitis D (Delta) virus and hepatitis E are also RNA viruses.
  • Additional RNA viruses include reoviruses, rotaviruses, birnaviruses, chrysoviruses, cystoviruses, hypoviruses, partitiviruses and totoviruses.
  • Orbiviruses such as African horse sickness virus, Blue tongue virus, Changuinola virus, Chenuda virus, Chobar Gorge Corriparta virus, epizootic hemorraghic disease virus, equine encephalosis virus, Eubenangee virus, Ieri virus, Great Island virus, Lebombo virus, Orungo virus, Palyam virus, Peruvian Horse Sickness virus, St. Croix River virus, Umatilla virus, Wad Medani virus, Wallal virus, Warrego virus and Wongorr virus are also RNA viruses.
  • Retroviruses include alpharetroviruses (for example, Rous sarcoma virus and avian leukemia virus), betaretroviruses (for example, mouse mammary tumor virus, Mason-Pfizer monkey virus and Jaagsiekte sheep retrovirus), gammaretroviruses (for example, murine leukemia virus and feline leukemia virus, deltraretroviruses (for example, human T cell leukemia viruses (HTLV- 1 , HTL V-2), bovine leukemia virus, STLV- 1 and STLV-2), epsilonretriviruses (for example, Walleye dermal sarcoma virus and Walleye epidermal hyperplasia virus 1), reticuloendotheliosis virus (for example, chicken syncytial virus, lentiviruses (for example, human immunodeficiency virus (HIV) type 1, human immunodeficiency virus (HIV) type 2, simian immunodeficiency virus, equine infectious an
  • DNA viruses include polyomaviruses (for example, simian virus 40, simian agent 12, BK virus, JC virus, Merkel Cell polyoma virus, bovine polyoma virus and lymphotrophic papovavirus), papillomaviruses (for example, human papillomavirus, bovine papillomavirus, adenoviruses (for example, adenoviruses A-F, canine adenovirus type I, canined adeovirus type 2), circoviruses (for example, porcine circovirus and beak and feather disease virus (BFDV)), parvoviruses (for example, canine parvovirus), ery thro viruses (for example, adeno-associated virus types 1-8), betaparvoviruses, amdoviruses, densoviruses, iteraviruses, brevidensoviruses, pefudensoviruses, herpes viruses (for example, poly
  • viruses include, but are not limited to, the animal counterpart to any above listed human virus.
  • the provided genes can also decrease infection by newly discovered or emerging viruses. Such viruses are continuously updated on http://en.wikipedia.org/wiki/Virus and www.virology.net.
  • bacterial infections include, but are not limited to infections caused by the following bacteria: Listeria (sp.), Franscicella tularemis, Mycobacterium tuberculosis, Rickettsia (all types), Ehrlichia, Chlamydia.
  • Further examples of bacteria that can be targeted by the present methods include M. tuberculosis, M. bovis, M. bovis strain BCG, BCG substrains, M. avium, M. intracellular, M. africanum, M. kansasii, M. marinum, M. ulcerans, M.
  • avium subspecies paratuberculosis Nocardia asteroides, other Nocardia species, Legionella pneumophila, other Legionella species, Salmonella typhi, other Salmonella species, Shigella species, Yersinia pestis, Pasteurella haemolytica, Pasteurella multocida, other Pasteurella species, Actinobacillus pleuropneumoniae, Listeria monocytogenes, Listeria ivanovii, Brucella abortus, other Brucella species, Cowdria ruminantium, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydia psittaci, Coxiella burnetti, other Rickettsial species, Ehrlichia species, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Streptococcus agalactiae, Bacillus anth
  • Campylobacter species Neiserria meningitidis, Neiserria gonorrhea, Pseudomonas aeruginosa, other Pseudomonas species, Haemophilus influenzae, Haemophilus ducreyi, other Hemophilus species, Clostridium tetani, other Clostridium species, Yersinia enterolitica, and other Yersinia species.
  • parasitic infections include, but are not limited to infections caused by the following parasites: Cryptosporidium, Plasmodium (all species), American trypanosomes (T.
  • African trypanosomes Acanthamoeba, Entaoeba histolytica, Angiostrongylus, Anisakis, Ascaris, Babesia, Balantidium, Baylisascaris, lice, ticks, mites, fleas, Capillaria, Clonorchis, Chilomastix mesnili, Cyclspora, Diphyllobothrium, Dipylidium caninum, Fasciola, Giardia, Gnathostoma, Hetetophyes, Hymenolepsis, Isospora, Loa loa, Microsporidia, Naegleria, Toxocara, Onchocerca, Opisthorchis, Paragonimus, Baylisascaris, Strongyloides, Taenia, Trichomonas and Trichuris.
  • protozoan and fungal species contemplated within the present methods include, but are not limited to, Plasmodium falciparum, other Plasmodium species, Toxoplasma gondii, Pneumocystis carinii, Trypanosoma cruzi, other trypanosomal species, Leishmania donovani, other Leishmania species, Theileria annulata, other Theileria species, Eimeria tenella, other Eimeria species, Histoplasma capsulatum, Cryptococcus neoformans, Blastomyces dermatitidis, Coccidioides immitis, Paracoccidioides brasiliensis, Penicillium marnejfei, and Candida species.
  • the provided genes can also decrease infection by newly discovered or emerging bacteria, parasites or fungi, including multidrug resistant strains of same.
  • Respiratory viruses include, but are not limited to, picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses. More specifically, and not to be limiting, the respiratory virus can be an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus or a respiratory syncytial virus (RSV).
  • RSV respiratory syncytial virus
  • Gastrointestinal viruses include, but are not limited to, picornavi ruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses.
  • the gastrointestinal virus can be a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, an adenovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.
  • Also provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is a gastrointestinal virus that is not West Nile virus.
  • Gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses.
  • the gastrointestinal virus can be a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, or a Dengue fever virus.
  • a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is a gastrointestinal virus that is not West Nile virus or Dengue fever virus.
  • Gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses.
  • the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, or an enterovirus.
  • gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a West Nile virus or a Dengue fever virus.
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , wherein the pathogen is a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses,
  • the pathogen is a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis
  • Hepatitis A LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, a hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow F
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human
  • Papilloma Virus CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies or Chikungunya virus.
  • the present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chik
  • Also provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2 or 4, wherein the pathogen is a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.
  • a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus, Ebola virus or Marburg virus.
  • a method of decreasing the toxicity of a toxin in a cell comprising administering to the cell an effective amount of a composition that decreases expression or activity of one or more gene(s) or a gene product(s) set forth in Table 1, 2, 3 or 4.
  • Toxins can include, but are not limited to, a bacterial toxin, a neurotoxin, such as a botulinum neurotoxin, a mycotoxin, ricin, a Clostridium perfringens toxin, a Clostridium difficile toxin, a saxitoxin, a tetrodotoxin, abrin, a conotoxin, a Staphylococcal toxin, such as staphylococcal aureus, an E.
  • a neurotoxin such as a botulinum neurotoxin, a mycotoxin, ricin, a Clostridium perfringens toxin, a Clostridium difficile toxin, a saxitoxin, a tetrodotoxin, abrin, a conotoxin
  • Staphylococcal toxin such as staphylococcal aureus, an E.
  • the decrease in toxicity can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of toxicity in a cell wherein expression or activity of the same gene or gene product from Table 1, 2, 3 or 4 that has not been decreased, or compared to the level of toxicity in a cell not contacted with a compound that decreases expression or activity of the gene or gene product.
  • Toxicity can be measured, for example, via a cell viability, apopotosis assay, LDH release assay or cytotoxicity assay (See, for example, Kehl-Fie and St. Geme “Identification and characterization of an RTX toxin in the emerging pathogen Kingella kingae,” J. Bacteriol. 189(2):430-6 (2006) and Kirby “Anthrax Lethal Toxin Induces Human Endothelial cell Apoptosis,” Infection and Immunity 72: 430-439 (2004), both of which are incorporated herein in their entireties by this reference.)
  • expression and/or activity of a gene or gene product set forth in Table 1, 2, 3 or 4 can be decreased by contacting the cell with any composition that can decrease expression or activity.
  • the composition can be a chemical, a small or large molecule (organic or inorganic), a drug, a protein, a peptide, a cDNA, an antibody, an aptamer, a morpholino, a triple helix molecule, an siRNA, an LNA, a shRNA, an miRNA, an antisense RNA, a ribozyme or any other compound now known or identified in the future that decreases the expression and/or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4.
  • a decrease in expression or activity can occur by decreasing transcription of mRNA or decreasing translation of RNA.
  • a composition can also be a mixture or "cocktail" of two or more of the compositions described herein.
  • a decrease in expression and/or activity can also occur by inhibiting the interaction between any of the proteins set forth in Table 1 , 2, 3 or 4 and other cellular proteins, such as, for example, transcription factors, receptors, nuclear proteins, transporters, microtubules, membrane proteins, enzymes (for example, ATPases, phosphorylases, oxidoreductases, kinases, phosphatases, synthases, lyases, aromatases, helicases, hydrolases, proteases, transferases, nucleases, ligases, reductases and polymerases) and hormones.
  • a decrease in expression and/or activity can also occur by inhibiting or decreasing the interaction between any of the proteins of the present invention and a cellular nucleic acid or a viral nucleic acid.
  • a decrease can also occur by inhibiting or decreasing the interaction, either direct or indirect, between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).
  • a composition can also be single composition or a mixture, cocktail or combination of two or more compositions, for example, two or more compositions selected from the group consisting of chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, an aptamer, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an LNA, an antisense nucleic acid or a ribozyme.
  • the two or more compositions can be the same or different types of compositions.
  • the two or more compositions can decrease expression or activity of the same target or different targets, as one or more genes or gene products set forth in Table 1,2, 3 or 4 can be modulated to effect a decrease in infection. It is understood that two or more compositions comprises three or more, four or more, five or more etc.
  • two or more compositions can be any two or more compositions comprising an antisense and a small molecule; or two or more antisense molecules; or two or more small molecules; or two or more compositions comprising an siRNA and a small molecule, etc. It is understood that any combination of the types of compositions set forth herein can be utilized in the methods set forth herein.
  • compositions can be used alone or in combination with other therapeutic agents such as antiviral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti- inflammatory agents, anti-cancer agents, etc. All of the compounds described herein can be contacted with a cell in vitro, ex vivo or in vivo.
  • antiviral compounds include, but are not limited to, amantadine, rimantadine, zanamavir and oseltamavir (Tamiflu) for the treatment of flu and its associated symptoms.
  • Antiviral compounds useful in the treatment of HIV include Combivir® (lamivudine-zidovudine), maraviroc, Crixivan® (indinavir), Emtriva® (emtricitabine), Epivir® (lamivudine), Fortovase® (saquinavir-sg), Hivid® (zalcitabine), Invirase® (saquinavir-hg), Kaletra® (lopinavir-ritonavir), LexivaTM (fosamprenavir), Norvir® (ritonavir), Retrovir® (zidovudine) Sustiva® (efavirenz), Videx EC® (didanosine), Videx® (didanosine), Viracept® (nelf ⁇ navir)
  • antiviral compounds useful in the treatment of Ebola and other filoviruses include ribavirin and cyanovirin-N (CV-N).
  • CV-N cyanovirin-N
  • antibacterial agents include, but are not limited to, antibiotics (for example, penicillin and ampicillin), sulfa Drugs and folic acid Analogs, Beta-Lactams, aminoglycosides, tetracyclines, macrolides, lincosamides, streptogramins, fluoroquinolones, rifampin, mupirocin, cycloserine, aminocyclitol and oxazolidinones.
  • Antifungal agents include, but are not limited to, amphotericin, nystatin, terbinafine, itraconazole, fluconazole, ketoconazole, and griselfulvin.
  • Antiparasitic agents include, but are not limited to, antihelmintics, antinematodal agents, antiplatyhelmintic agents, antiprotozoal agents, amebicides, antimalarials, antitrichomonal agents, aoccidiostats and trypanocidal agents.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression and/or activity of a gene product set forth in Table 1,2 3, or 4 and a composition that decreases expression and/or activity of a different gene product(s) set forth in Table 1, 2, 3 or 4.
  • the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression and/or activity of EDNRA and a composition that decreases expression and/or activity of KCNJl 1. This is merely exemplary, as any two or more different gene product(s) set forth in Table 1, 2, 3 or 4 can be inhibited to decrease infection.
  • compositions can be a combination of two, three, four, five or six compositions, wherein each composition comprises a chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, a drug (for example, an FDA approved drug, a European Medicines Agency approved drug, a drug approved by the Japanese Pharmaceutical and Medical Device Agency) a protein, a cDNA, an aptamer, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an LNA, an miRNA, an antisense nucleic acid or a ribozyme.
  • a drug for example, an FDA approved drug, a European Medicines Agency approved drug, a drug approved by the Japanese Pharmaceutical and Medical Device Agency
  • a protein for example, an FDA approved drug, a European Medicines Agency approved drug, a drug approved by the Japanese Pharmaceutical and Medical Device Agency
  • a protein for example, an FDA approved drug, a European Medicines Agency approved drug, a drug approved
  • these combinations can comprise two or more compounds selected from the group consisting of imatinib, acetazolamide, benzthiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate, trichloromethiazide, amobarbital, atracurium, cisatracurium, D- tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enflurane, isoflurane, plerixafor, ambrisentan, avosentan, bosentan, BSF 302146, clazosentan, PD 180988, SB
  • Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens.
  • the two or more pathogens can be two or more, three or more, four or more, five or more, six or more; or seven or more viruses, bacteria, parasites, fungi or a combination thereof.
  • the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses.
  • Table 6 shows gene products set forth in Table 1, 2, 3 or 4 for which at least 50% inhibition of viral replication was observed via siRNA experiments as indicated by an X. It is understood that one of skill in the art can select a gene product from Table 6 and decrease its expression or activity to reduce infection by one or more viruses marked with an X, for that gene product.
  • One of skill in the art can readily select the gene product(s) in Table 6 that can be inhibited in order to effect inhibition of infection by two or more, three or more, four or more, five or more, six or more, seven or more viruses etc.
  • the viruses are not restricted to the viruses set forth in Table 6 as this data can be combined with data for a composition that decreases expression and/or activity of any gene product from Table 1, 2, 3 or 4 and inhibits any other virus, bacteria, fungi or parasite not listed in Table 6.
  • data for EDNRA can be combined with data showing that decreased expression and/or activity of EDNRA leads to inhibition of HCV infection, thus showing that activity or expression of EDNRA can be decreased to inhibit infection by RSV, pox, rhinovirus, Dengue fever virus, HIV and HCV.
  • This example is merely exemplary as data for any gene product set forth in Table 6 can be combined with data for any other virus, bacteria, fungi or parasite.
  • the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses, wherein at least two or more of the viruses are selected from the group consisting of influenza, RSV, HSV, cowpox, rhinovirus, HIV and Dengue.
  • Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1,2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses, wherein at least two or more of the viruses are selected from the group consisting of influenza, RSV, HSV, cowpox, rhinovirus, Dengue, HIV and HCV. TABLE 6
  • the present invention also provides antibodies that specifically bind to the gene products, proteins and fragments thereof set forth in Table 1, 2, 3 or 4.
  • the antibody of the present invention can be a polyclonal antibody or a monoclonal antibody.
  • the antibody of the invention selectively binds a polypeptide.
  • selectively binds or “specifically binds” is meant an antibody binding reaction which is determinative of the presence of the antigen (in the present case, a polypeptide set forth in Table 1 , 2,3 or 4, or antigenic fragment thereof among a heterogeneous population of proteins and other biologies).
  • the specified antibodies bind preferentially to a particular peptide and do not bind in a significant amount to other proteins in the sample.
  • selective binding includes binding at about or above 1.5 times assay background and the absence of significant binding is less than 1.5 times assay background.
  • This invention also contemplates antibodies that compete for binding to natural interactors or ligands to the proteins set forth in Table 1, 2, 3 or 4.
  • the present invention provides antibodies that disrupt interactions between the proteins set forth in Table 1, 2, 3 or 4 and their binding partners.
  • an antibody of the present invention can compete with a protein for a binding site (e.g. a receptor) on a cell or the antibody can compete with a protein for binding to another protein or biological molecule, such as a nucleic acid that is under the transcriptional control of a transcription factor set forth in Table 1 , 2, 3 or 4.
  • An antibody can also disrupt the interaction between a protein set forth in Table 1 , 2, 3 or 4 and a pathogen, or the product of a pathogen.
  • an antibody can disrupt the interaction between a protein set forth in Table 1 , 2, 3 or 4 and a viral protein, a bacterial protein, a parasitic protein, a fungal protein or a toxin.
  • the antibody optionally can have either an antagonistic or agonistic function as compared to the antigen.
  • Antibodies that antagonize pathogenic infection are utilized to decrease infection.
  • antibodies that antagonize toxicity can be utilized to decrease toxicity in a cell or subject.
  • the antibody binds a polypeptide in vitro, ex vivo or in vivo.
  • the antibody of the invention is labeled with a detectable moiety.
  • the detectable moiety can be selected from the group consisting of a fluorescent moiety, an enzyme-linked moiety, a biotin moiety and a radiolabeled moiety.
  • the antibody can be used in techniques or procedures such as diagnostics, screening, or imaging. Anti-idiotypic antibodies and affinity matured antibodies are also considered to be part of the invention.
  • antibody encompasses chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab')2, Fab', Fab and the like, including hybrid fragments.
  • fragments of the antibodies that retain the ability to bind their specific antigens are provided.
  • Such antibodies and fragments can be made by techniques known in the art and can be screened for specificity and activity according to the methods set forth in the Examples and in general methods for producing antibodies and screening antibodies for specificity and activity (See Harlow and Lane. Antibodies, A Laboratory Manual. Cold Spring Harbor Publications, New York, (1988)).
  • antibody conjugates of antibody fragments and antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference.
  • the antibodies are generated in other species and “humanized” for administration in humans.
  • the "humanized” antibody is a human version of the antibody produced by a germ line mutant animal.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • the present invention provides a humanized version of an antibody, comprising at least one, two, three, four, or up to all CDRs of a monoclonal antibody that specifically binds to a protein or fragment thereof set forth in Table 1, 2, 3 or 4.
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of or at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non- human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., Nature, 321 :522-525 (1986); Riechmann et al., Nature, 332:323- 327 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).
  • Fc immunoglobulin constant region
  • a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321 :522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534- 1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody.
  • humanized antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • Peptides that inhibit expression or activity of a gene or a gene product set forth in Table 1 , 2, 3 or 4 are also provided herein.
  • Peptide libraries can be screened utilizing the screening methods set forth herein to identify peptides that inhibit activity of any of the genes or gene products set forth in Table 1 , 2, 3 or 4. These peptides can be derived from a protein that binds to any of the genes or gene products set forth in Table 1, 2, 3 or 4.
  • peptides can be any peptide in a purified or non-purified form, such as peptides made of D-and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam et al., Nature 354:82-4, 1991), phosphopeptides (such as in the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang et al, Cell 72:767-78, 1993).
  • RNA interference technologies are known to those of skill in the art, for example, but not limited to, siRNA, antisense, locked nucleic acids (LNA), morpholinos, and shRNA.
  • LNA locked nucleic acids
  • shRNA shRNA
  • One of skill in the art can utilize any RNA interference technology, known or identified in the future to decrease expression of any gene product set forth in Table 1, 2, 3 or 4.
  • siRNAs Short interfering RNAs
  • small interfering RNAs are double- stranded RNAs that can induce sequence-specific post-transcriptional gene silencing, thereby decreasing gene expression
  • siRNas can be of various lengths as long as they maintain their function. In some examples, siRNA molecules are about 19-23 nucleotides in length, such as at least 21 nucleotides, for example at least 23 nucleotides.
  • siRNA triggers the specific degradation of homologous RNA molecules, such as mRNAs, within the region of sequence identity between both the siRNA and the target RNA.
  • RNA molecules such as mRNAs
  • WO 02/44321 discloses siRNAs capable of sequence-specific degradation of target mRNAs when base-paired with 3' overhanging ends. The direction of dsRNA processing determines whether a sense or an antisense target RNA can be cleaved by the produced siRNA endonuclease complex.
  • siRNAs can be used to modulate transcription or translation, for example, by decreasing expression of a gene set forth in Table 1, 2, 3 or 4. The effects of siRNAs have been demonstrated in cells from a variety of organisms, including Drosophila, C.
  • siRNAs can be designed to specifically target one or more of the genes set forth in Table 1 , 2, 3 or 4 for decreased gene expression.
  • siRNAs that inhibit or silence gene expression can be obtained from numerous commercial entities that synthesize siRNAs, for example, Ambion Inc.
  • siRNAs synthesized by Ambion Inc., Qiagen Inc. or Dharmacon Inc, can be readily obtained from these and other entities by providing a GenBank Accession No. for the mRNA of any gene set forth in Table 1, 2, 3 or 4.
  • siRNAs can be generated by utilizing Invitrogen's BLOCK-ITTM RNAi Designer https://rnaidesigner.invitrogen.com/rnaiexpress.
  • siRNA sequences can comprise a 3 'TT overhang and/or additional sequences that allow efficient cloning and expression of the siRNA sequences.
  • siRNA sequences can be cloned into vectors and utilized in vitro, ex vivo or in vivo to decrease gene expression.
  • One of skill in the art would know that it is routine to utilize publicly available algorithms for the design of siRNA to target mRNA sequences. These sequences can then be assayed for inhibition of gene expression in vitro, ex vivo or in vivo.
  • shRNA shRNA short hairpin RNA is a DNA molecule that can be cloned into expression vectors to express siRNA (typically 19-29 nt RNA duplex) for RNAi interference studies.
  • shRNA has the following structural features: a short nucleotide sequence ranging from about 19- 29 nucleotides derived from the target gene, followed by a short spacer of about 4-15 nucleotides (i.e. loop) and about a 19-29 nucleotide sequence that is the reverse complement of the initial target sequence.
  • a short nucleotide sequence ranging from about 19- 29 nucleotides derived from the target gene, followed by a short spacer of about 4-15 nucleotides (i.e. loop) and about a 19-29 nucleotide sequence that is the reverse complement of the initial target sequence.
  • the term "antisense” refers to a nucleic acid molecule capable of hybridizing to a portion of an RNA sequence (such as mRNA) by virtue of some sequence complementarity.
  • the antisense nucleic acids disclosed herein can be oligonucleotides that are double-stranded or single-stranded, RNA or DNA or a modification or derivative thereof, which can be directly administered to a cell (for example by administering the antisense molecule to the subject), or which can be produced intracellularly by transcription of exogenous, introduced sequences (for example by administering to the subject a vector that includes the antisense molecule under control of a promoter).
  • Antisense nucleic acids are polynucleotides, for example nucleic acid molecules that are at least 6 nucleotides in length, at least 10 nucleotides, at least 15 nucleotides, at least 20 nucleotides, at least 100 nucleotides, at least 200 nucleotides, such as 6 to 100 nucleotides.
  • antisense molecules can be much longer.
  • the nucleotide is modified at one or more base moiety, sugar moiety, or phosphate backbone (or combinations thereof), and can include other appending groups such as peptides, or agents facilitating transport across the cell membrane (Letsinger et al, Proc. Natl. Acad. Sci.
  • modified base moieties include, but are not limited to: 5-fluorouracil, 5- bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N ⁇ 6- sopentenyladenine, 1 -methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, methoxyarninomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'- methoxycarboxymethylurac
  • modified sugar moieties include, but are not limited to: arabinose, 2- fluoroarabinose, xylose, and hexose, or a modified component of the phosphate backbone, such as phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, or a formacetal or analog thereof.
  • an antisense molecule is an ⁇ -anomeric oligonucleotide.
  • An ⁇ - anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gautier et al., Nucl. Acids Res. 15:6625-41, 1987).
  • the oligonucleotide can be conjugated to another molecule, such as a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization- triggered cleavage agent.
  • Oligonucleotides can include a targeting moiety that enhances uptake of the molecule by host cells.
  • the targeting moiety can be a specific binding molecule, such as an antibody or fragment thereof that recognizes a molecule present on the surface of the host cell.
  • antisense molecules that recognize a nucleic acid set forth herein include a catalytic RNA or a ribozyme (for example see WO 90/11364; WO 95/06764; and Sarver et al, Science 247:1222-5, 1990).
  • Conjugates of antisense with a metal complex, such as terpyridylCu (II), capable of mediating mRNA hydrolysis, are described in Bashkin et al. ⁇ Appl. Biochem Biotechnol 54:43-56, 1995).
  • the antisense nucleotide is a 2'-O- methylribonucleotide (Inoue et al., Nucl.
  • Antisense molecules can be generated by utilizing the Antisense Design algorithm of Integrated DNA Technologies, Inc. (1710 Commercial Park, Coralville, IA 52241 USA; http://www.idtdna.com/Scitools/ Applications/ AntiSense/ Antisense. aspx.
  • any antisense sequence that is not the full length mRNA for any of the genes listed in Table 1, 2, 3 or 4 can be used as antisense sequences. It is known to those of skill in the art that once a mRNA sequence is routinely obtained for any of the genes set forth in Table 1, 2, 3 or 4, it is routine to walk along the mRNA sequence to generate antisense sequences that decrease expression of the gene. Therefore all antisense sequences for the genes set forth herein are disclosed. The methods of the present invention can utilize any antisense sequence that decreases the expression of a gene set forth in Table 1, 2, 3 or 4.
  • Morpholinos are synthetic antisense oligos that can block access of other molecules to small (about 25 base) regions of ribonucleic acid (RNA). Morpholinos are often used to determine gene function using reverse genetics methods by blocking access to mRNA. Morpholinos, usually about 25 bases in length, bind to complementary sequences of RNA by standard nucleic acid base-pairing. Morpholinos do not degrade their target RNA molecules. Instead, Morpholinos act by "steric hindrance", binding to a target sequence ' within an RNA and simply interfering with molecules which might otherwise interact with the RNA. Morpholinos have been used in mammals, ranging from mice to humans.
  • Morpholinos can interfere with progression of the ribosomal initiation complex from the 5' cap to the start codon. This prevents translation of the coding region of the targeted transcript (called “knocking down" gene expression). Morpholinos can also interfere with pre-mRNA processing steps, usually by preventing the splice-directing snRNP complexes from binding to their targets at the borders of introns on a strand of pre-RNA. Preventing Ul (at the donor site) or U2/U5 (at the polypyrimidine moiety & acceptor site) from binding can cause modified splicing, commonly leading to exclusions of exons from the mature mRNA.
  • Ul at the donor site
  • U2/U5 at the polypyrimidine moiety & acceptor site
  • splice targets results in intron inclusions, while activation of cryptic splice sites can lead to partial inclusions or exclusions. Targets of Ul 1/U12 snRNPs can also be blocked. Splice modification can be conveniently assayed by reverse-transcriptase polymerase chain reaction (RT-PCR) and is seen as a band shift after gel electrophoresis of RT-PCR products.
  • RT-PCR reverse-transcriptase polymerase chain reaction
  • Preferred small molecules are those small molecules that have IC 50 values of less than about ImM, less than about 500 micromolar, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar, less than about 1 micromolar, less than about 0.5 micromolar, less than about 0.25 micromolar, or less than about .10 micromolar.
  • the half maximal inhibitory concentration (IC 50 ) is a measure of the effectiveness of a compound in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular compound or other substance (inhibitor) is needed to inhibit a given biological process (or component of a process, i.e.
  • IC 50 inhibitory concentration
  • IC 50 represents the concentration of a drug that is required for 50% inhibition in vitro. It is comparable to an EC 50 for agonist drugs. EC 50 also represents the plasma concentration required for obtaining 50% of a maximum effect in vivo.
  • the present invention also provides the synthesis of small molecules that inhibit activity of a gene product set forth in Table 1, 2, 3 or 4.
  • the present invention describes gene products for which three-dimensional structures are well known and can be obtained from the RCSB Protein Databank http://www.rcsb.org/pdb/home/home.do or http://www.rcsb.org.
  • Crystal structures can also be generated.
  • one of skill in the art can obtain crystal structures for proteins, or domains of proteins, that are homologous to the proteins set forth in Table 1 , 2,3 or 4 from the RCSB Protein Databank or elsewhere in the scientific literature for use in homology modeling studies.
  • a filter can be applied to the results to yield one or more compounds with a binding affinity in a particular range, for example, and not to be limiting, from about 100 micromolar to about 100 nanomolar, from about 10 micromolar to about 10 nanomolar, from about 1 micromolar to about 1 nanomolar, or from about 0.5 micromolar to about 0.5 nanomolar.
  • Another filter can provide compounds with a certain binding affinity and size, for example, less than 1000 daltons, less than 500 daltons, less than 400 daltons, less than 300 daltons, less than 200 daltons, less than 100 daltons or less than 50 daltons or any size in between.
  • the ranges and properties can be modified depending on the protein being studied.
  • the compounds identified via this screening method can be further studied in silico, in vitro or in vivo.
  • the compounds can be modified in silico and rescreened in silico to determine the effects of chemical modifications on binding affinity or other properties being assessed in silico.
  • the compounds identified in silico can be synthesized for in vitro or in vivo analysis.
  • All of the screening leading up to in vivo testing can be done in silico or in combination with in vitro assays.
  • the initial compounds identified in silico and the resulting modified compounds can be screened in vitro, for example, in cellular assays to determine the effect of the compound on the cellular host protein as well as in viral assays, to determine antiviral activity.
  • IC 50 values can be obtained from the cellular assays, which may or may not be similar to the concentration necessary to effect 50% inhibition of viral infection in a viral assay.
  • a compound that has an IC 50 value of less than about ImM less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar , less than about 1 micromolar, less than about 0.5 micromolar or less than about 0.1 micromolar.
  • filters can be applied to the in silico screening process, for example, a filter that takes ADMET (adsorption, distribution, metabolism, excretion, toxicity) properties into consideration can be applied.
  • ADMET modeling can be used during compound optimization to define an acceptable property space that contains compounds likely to have the desired properties.
  • Libraries for virtual or in vitro screening are available for the skilled artisan, for example from ChemBridge Corporation (San Diego, CA), such as a GPCR library, a kinase targeted library (KINACore), or an ion channel library (Ion Channel Set), to name a few.
  • Compound libraries can also be obtained from the National Institutes of Health. For example, the NIH Clinical Collection of compounds that have been used in clinical trials can also be screened. Biofocus DPI (Essex, United Kingdom) also maintains and designs compound libraries that can be purchased for screening.
  • One of skill in the art can select a library based on the protein of interest.
  • a kinase library can be screened to identify a compound that binds to and/or modulates a kinase.
  • Other libraries that target enzyme families for example, ATPases, hydrolases, isomerases, polymerases, transferases, phosphatases, etc., can also be screened, depending on the type of enzyme.
  • Compound libraries can also be screened in order to identify a compound that disrupts or inhibits specific interactions between two proteins.
  • Compounds can be administered to cells comprising two or more proteins that interact with each other in order to identify compounds that disrupt this interaction and result in decreased interaction between of the two or more proteins.
  • Co-immunoprecipiation experiments can be utilized.
  • FRET analysis or surface plasmon resonance can be utilized, to identify compounds that disrupt the interaction between a two proteins.
  • Transcription factors set forth herein are available to identify inhibitors in screening assays that are known to the skilled person.
  • Gel shift assays can be employed to assess the activity of a transcription factor (see, for example, Wang et al. 2000 The Journal of Biological Chemistry, 275, 27013-27020).
  • One of skill in the art can also utilize the techniques set forth in Wang et al., as they apply to the transcription factors set forth herein to assess transcription factor activity via binding of the transcription factor to a regulatory region that drives expression of a reporter protein.
  • These cellular assays and other transcription factor assays standard in the art are applicable to any transcription factor described herein.
  • Compounds can be introduced into these assays to assess the effect of the compound on transcriptional activity.
  • a cellular assay can be utilized to measure reporter activity in the presence of a transcription factor, in the presence or absence of a compound, to determine the effect of the compound on transcription. If reporter activity is reduced, the compound is an inhibitor of transcription by that particular transcription factor. Utilizing these assays can also identify compounds that inhibit the activity of for example, any zinc finger protein set forth in this application. Interactions between membrane proteins and their ligands, can also be measured, for example, via plasmon- waveguide resonance (PWR) spectroscopy (see, for example, Hruby and Tollin, Curr. Opin. Pharmacol. 2007 October: 7(5): 507-514; and Salamon et al. Methods Enzymol.
  • PWR plasmon- waveguide resonance
  • Additional inhibitors include compositions comprising carbon and hydrogen, and optionally comprising one or more of -S, -N, -O, -Cl, -Br, or -Fl, appropriately bonded as a structure, with a size of less than about 1000 daltons, less than about 500 daltons, less than about 300 daltons, less than about 200 daltons, or less than about 100 daltons, that fits into a binding pocket or an active site of a gene product set forth herein.
  • inhibitors that have the properties described in Lipinsky's Rule of Five are included herein.
  • the present invention provides a method of decreasing infection by a pathogen, in a cell or a subject, said method comprising administering to the cell or subject an effective amount of one or more compounds selecting from the group of compounds having the structure of imatinib, acetazolamide, benzthiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate, trichloromethiazide, amobarbital, atracurium, cisatracurium, D-tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enfiura
  • salts are those salts derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids.
  • Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g. magnesium) and ammonium salts.
  • Preferred salts include hydrochlorides, hydrobromides, sulfates, mesylates, maleates, and fumarates.
  • references to a compound, according to the invention includes the compounds as well as its pharmaceutically acceptable salts.
  • the infection can be a viral, bacterial, fungal or parasitic infection.
  • the pathogen can be a respiratory virus.
  • Respiratory viruses include, but are not limited to, picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses.
  • the respiratory virus can be an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus or a respiratory syncytial virus (RSV) or any strain thereof.
  • the pathogen can also be a gastrointestinal virus.
  • Gastrointestinal viruses include, but are not limited to, picornaviruses, adenoviruses, filoviruses, flaviviruses, caliciviruses and reoviruses.
  • the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.
  • the pathogen can also be a hemorraghic fever virus. These include, but are not limited to, flaviviruses, bunyaviruses, arenaviruses, filoviruses and hantaviruses.
  • the hemorraghic fever virus can be an Ebola virus, a Marburg virus, a Dengue fever virus (types 1-4), a yellow fever virus, a Sin Nombre virus, a Junin virus, Crimean Congo hemorraghic fever virus, a Machupo virus, a Lassa virus, a Rift Valley fever virus, or a Kyasanur forest disease virus.
  • the pathogen can also be a pathogen selected from the group consisting of: a pox virus, a herpes virus, an RSV virus, HIV, an influenza virus, a hepatitis C virus, a hepatitis B virus, a hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • the compounds can also be utilized to decrease
  • Other methods of decreasing expression and/or activity include methods of interrupting or altering transcription of mRNA molecules by site-directed mutagenesis (including mutations caused by a transposon or an insertional vector).
  • Chemical mutagenesis can also be performed in which a cell is contacted with a chemical (for example ENU) that mutagenizes nucleic acids by introducing mutations into a gene set forth in Table 1 , 2, 3 or 4.
  • Transcription of mRNA molecules can also be decreased by modulating a transcription factor that regulates expression of any of the genes set forth in Table 1 , 2, 3 or 4. Radiation can also be utilized to effect mutagenesis.
  • the present invention provides_a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, in the subject, wherein the pathogen is not HIV.
  • said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, in the subject, wherein the pathogen is not West Nile Virus.
  • said method comprising administering to the subject an effective amount of a that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, in the subject, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
  • the present invention also provides a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, in the subject, wherein the pathogen is not hepatitis C.
  • Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by three or more respiratory viruses.
  • Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by five or more respiratory viruses.
  • These can be selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus and an adenovirus.
  • picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses are families of viruses, two or more, three or more, four or more, or five or more respiratory viruses can be from the same or from different families.
  • the composition can inhibit infection by two or more orthomyxoviruses; two or more picornaviruses; an orthomyxovirus, an adenovirus, and a picornavirus; an orthomyxovirus, a paramyxovirus and an adenovirus; an orthomyxovirus, two picornaviruses and a paramyxovirus; three orthomyxoviruses, a picornavirus and an adenovirus, etc. More particularly, the composition can inhibit infection by two or more, three or more or four or more respiratory viruses selected from the group consisting of an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus and an RSV virus.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more gastrointestinal viruses.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene produces) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by three or more gastrointestinal viruses.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more gastrointestinal viruses.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by five or more gastrointestinal viruses.
  • viruses can be selected from the group consisting of: a filovirus, an adenovirus, a picornavirus, a calcivirus, a flavivirus or a reovirus. Since filoviruses, picornaviruses, calciviruses, flaviviruses and reoviruses are families of viruses, the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses from the same or from different families.
  • the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses selected from the group consisting of a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus.
  • a reovirus selected from the group consisting of a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus.
  • a reovirus selected from the group consisting of a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by one or more pathogens selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, and inhibits infection by one or more pathogens selected from the group consisting of: a flavivirus, a filovirus, an adenovirus, a calicivirus or a reovirus.
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s)set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 W
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.
  • a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by three or more pathogens.
  • the three or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein.
  • the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an R
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more pathogens.
  • the three or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein.
  • the four or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an R
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by five or more pathogens.
  • the five or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein.
  • the five or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an R
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by six or more pathogens.
  • the six or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein.
  • the five or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an R
  • the present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits co-infection by HIV and one or more viruses, bacteria, parasites or fungi.
  • a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits co-infection by HIV and one or more viruses, bacteria, parasites or fungi.
  • decreasing co-infection of HIV and any of the viruses including for example any families, genus, species, or group of viruses.
  • co-infection of HIV and a respiratory virus is provided herein.
  • Respiratory viruses include picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, and adenoviruses.
  • the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV. Also provided is decreasing co-infection of HIV and a gastrointestinal virus.
  • Gastrointestinal viruses include picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.
  • a method of decreasing co-infection of HIV with a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus More particularly, decreasing co-infection of HIV and a hepatitis virus, such as Hepatitis A, Hepatitis B or Hepatitis C is provided. It is noted that in a method of decreasing co-infection of HIV and Hepatitis C, the gene or gene product is not ERCCl, THAPl 1, COASY, NC0A6 or MAP3K14.
  • HIV- infection of HIV and a herpes virus for example, HSV-I or HSV-2.
  • a herpes virus for example, HSV-I or HSV-2.
  • decreasing co- infection of HIV and tuberculosis is also provided.
  • decreasing co-infection of HIV and CMV is also provided.
  • the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of two or more respiratory viruses. Therefore, the present invention provides methods of treating or preventing an unspecified respiratory infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more respiratory viruses.
  • the present invention provides a method of decreasing an unspecified respiratory infection in a subject comprising: a) diagnosing a subject with an unspecified respiratory infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more respiratory viruses selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses.
  • the two or more respiratory viruses can be from the same family or from a different family of respiratory viruses.
  • the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV.
  • the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more respiratory viruses selected from the group consisting of a picornaviruses, an orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses.
  • the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of two or more gastrointestinal viruses. Therefore, the present invention provides methods of treating or preventing an unspecified gastrointestinal infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more gastrointestinal viruses.
  • the present invention provides a method of decreasing an unspecified gastrointestinal infection in a subject comprising: a) diagnosing a subject with an unspecified gastrointestinal infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more gastrointestinal viruses selected from the group consisting of a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus. It is noted that if the gene or gene product is from Table 3, the two gastrointestinal viruses are not West Nile virus and Dengue fever virus.
  • the two or more gastrointestinal viruses can be from the same family or from a different family of gastrointestinal viruses. More particularly, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.
  • the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more gastrointestinal viruses selected from the group consisting of a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus.
  • the present invention also provides a method of preventing or decreasing an unspecified pandemic or bioterror threat in a subject comprising: a) diagnosing a subject with an unspecified pandemic or bioterrorist inflicted infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more, three or more, four or more; or five or more viruses selected from the group consisting of a pox virus, an influenza virus, West Nile virus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encepha
  • the present invention also provides a method wherein the composition administered is not a composition that decreases expression or activity of a gene set forth in Table 3 for West Nile virus and Dengue Fever virus.
  • a method of managing secondary infections in a patient comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition can inhibit infection by HIV and one or more, two or more, three or more, four or more; or five or more secondary infections.
  • the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of three or more pathogens.
  • the present invention provides methods of treating or preventing an unspecified infection by administering a composition that decreases the activity or expression of a gene that is involved in the pathogenesis of three or more pathogens. Therefore, the present invention provides a method of decreasing infection in a subject comprising: a) diagnosing a subject with an unspecified infection and; b) administering a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition decreases infection by three or more pathogens.
  • the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, CaI ici viruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an R
  • the infection can be a viral infection, a parasitic infection, a bacterial infection or a fungal infection, to name a few.
  • an unspecified infection is an infection that presents symptoms associated with an infection, but is not identified as specific infection.
  • a physician, a nurse, a physician's assistant, a medic or any other health practitioner would know how to diagnose the symptoms of infection even though the actual pathogen may not be known.
  • the patient can present with one or more symptoms, including, but not limited to, a fever, fatigue, lesions, weight loss, inflammation, a rash, pain (for example, muscle ache, headache, ear ache, joint pain, etc.), urinary difficulties, respiratory symptoms (for example, coughing, bronchitis, lung failure, breathing difficulties, bronchiolitis, airway obstruction, wheezing, runny nose, sinusitis, congestion, etc.), gastrointestinal symptoms (for example, nausea, diarrhea, vomiting, dehydration, abdominal pain, intestinal cramps, rectal bleeding, etc.). This can occur in the event of a bioterrorist attack or a pandemic.
  • a fever, fatigue, lesions, weight loss, inflammation, a rash, pain for example, muscle ache, headache, ear ache, joint pain, etc.
  • urinary difficulties respiratory symptoms
  • respiratory symptoms for example, coughing, bronchitis, lung failure, breathing difficulties, bronchiolitis, airway obstruction, wheezing, runny nose,
  • compositions that inhibits infection by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 that is involved in the pathogenesis of several pathogens.
  • a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4 can be administered prophylactically to a subject to prevent an unspecified infection in a subject.
  • treat By “treat,” “treating,” or “treatment” is meant a method of reducing the effects of an existing infection.
  • Treatment can also refer to a method of reducing the disease or condition itself rather than just the symptoms.
  • the treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease or the symptoms of the disease.
  • Treatment can range from a positive change in a symptom or symptoms of viral infection to complete amelioration of the viral infection as detected by art-known techniques.
  • a disclosed method is considered to be a treatment if there is about a 10% reduction in one or more symptoms of the disease in a subject with the disease when compared to native levels in the same subject or control subjects.
  • the reduction can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
  • a decrease in infection can be a decrease of hours, a day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, eleven days, twelve days, thirteen days, fourteen days, fifteen days or any time in between that it takes to see improvement in the symptoms, viral load or any other parameter utilized to measure improvement in a subject. For example, if it normally takes 7 days to see improvement in a subject not taking the composition, and after administration of the composition, improvement is seen at 6 days, the composition is effective in decreasing infection. This example is not meant to be limiting as one of skill in the art would know that the time for improvement will vary depending on the infection.
  • prevent is meant a method of precluding, delaying, averting, obviating, forestalling, stopping, or hindering the onset, incidence, severity, or recurrence of infection.
  • the disclosed method is considered to be a prevention if there is about a 10% reduction in onset, incidence, severity, or recurrence of infection, or symptoms of infection (e.g., inflammation, fever, lesions, weight loss, etc.) in a subject exposed to an infection when compared to control subjects exposed to an infection that did not receive a composition for decreasing infection.
  • the reduction in onset, incidence, severity, or recurrence of infection can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to control subjects. For example, and not to be limiting, if about 10% of the subjects in a population do not become infected as compared to subjects that did not receive preventive treatment, this is considered prevention.
  • Also provided is a method of decreasing infection in a subject comprising: a) administering a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4, in a subject with an unspecified infection; b) diagnosing the type of infection in the subject and; c) administering a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 for the diagnosed infection. Also provided is a method wherein the diagnosed infection is HIV and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 1.
  • the diagnosed infection is West Nile Virus and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 2. Further provided is a method wherein the diagnosed infection is West Nile Virus or Dengue fever virus and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 3. Further provided is a method wherein the diagnosed infection is Hepatitis C and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 4.
  • a method of treating viral infection comprising: a) diagnosing a subject with a viral infection; and b) removing a drug from the subject that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4, if the viral infection is not a viral infection that is inhibited by a composition that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4.
  • a practitioner can prescribe or administer a composition that decreases the expression or activity of the gene or gene product.
  • the practitioner who can be the same practitioner or a different practitioner, can diagnose the type of infection in a subject. This diagnosis can be a differential diagnosis where the practitioner distinguishes between infections by comparing signs or symptoms and eliminates certain types of infection before arriving at the diagnosis for a specific infection, or a diagnosis based on a test that is specific for a particular infection.
  • the practitioner can prescribe or administer a composition that decreases the expression or activity of that gene or gene product. This can be the same composition administered prior to diagnosis of the specific infection or a different composition that decreases expression or activity.
  • a method of preventing infection in a subject comprising administering to a subject susceptible to an unspecified infection a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4.
  • the composition can be administered in response to a lethal outbreak of an infection.
  • the infection can be a pandemic or a bioterrorist created infection.
  • a composition can be administered prophylactically to a subject to prevent an unspecified infection in a subject.
  • the threat can also come in the form of a toxin.
  • compositions that inhibits infection by decreasing the expression or activity of any one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 that is involved in the pathogenesis of two or more, three or more, four or more; or five or more pathogens.
  • Such prophylactic use can decrease the number of people in a population that are infected, thus preventing further spread of a pandemic or decreasing the effects of a bioterrorist attack.
  • the composition can comprise one or more of, a chemical, a compound, a small molecule, an inorganic molecule, an aptamer, an organic molecule, a drug, a protein, a cDNA, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an LNA, an antisense nucleic acid or a ribozyme that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4.
  • the composition can be administered before or after infection.
  • the decrease in infection in a subject need not be complete as this decrease can be a 10% , 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any other percentage decrease in between as long as a decrease occurs.
  • This decrease can be correlated with amelioration of symptoms associated with infection.
  • These compositions can be administered to a subject alone or in combination with other therapeutic agents described herein, such as anti-viral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti-inflammatory agents, anti-cancer agents, etc. Examples of viral infections, bacterial infections, fungal infections parasitic infections are set forth above.
  • the compounds set forth herein or identified by the screening methods set forth herein can be administered to a subject to decrease infection by any pathogen or infectious agent set forth herein.
  • any of the compounds set forth herein or identified by the screening methods of the present invention can also be administered to a subject to decrease infection by any pathogen, now known or later discovered in which a gene or gene product set forth in Table 1 , 2, 3 or 4 is involved.
  • Various delivery systems for administering the therapies disclosed herein are known, and include encapsulation in liposomes, microparticles, microcapsules, expression by recombinant cells, receptor-mediated endocytosis (Wu and Wu, J. Biol. Chem. 1987, 262:4429-32), and construction of therapeutic nucleic acids as part of a retroviral or other vector.
  • Methods of introduction include, but are not limited to, mucosal, topical, intradermal, intrathecal, intratracheal, via nebulizer, via inhalation, intramuscular, otic delivery (ear), eye delivery (for example, eye drops), intraperitoneal, vaginal, rectal, intravenous, subcutaneous, intranasal, and oral routes.
  • the compounds can be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (for example, oral mucosa, rectal, vaginal and intestinal mucosa, etc.) and can be administered together with other biologically active agents. Administration can be systemic or local.
  • Pharmaceutical compositions can be delivered locally to the area in need of treatment, for example by topical application or local injection.
  • compositions include a therapeutically effective amount of a RNA, DNA, antisense molecule, ribozyme, an LNA, siRNA, shRNA molecule, miRNA molecule, aptamer, drug, protein, small molecule, peptide inorganic molecule, organic molecule, antibody or other therapeutic agent, alone or with a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions or methods of treatment can be administered in combination with (such as before, during, or following) other therapeutic treatments, such as other antiviral agents, antibacterial agents, antifungal agents and antiparasitic agents.
  • each method can optionally comprise the step of diagnosing a subject with an infection or diagnosing a subject in need of prophylaxis or prevention of infection.
  • compositions and formulations suitable for pharmaceutical delivery of the therapeutic agents herein disclosed are conventional.
  • Remington 's Pharmaceutical Sciences, by Martin, Mack Publishing Co., Easton, PA, 15th Edition (1975) describes compositions and formulations suitable for pharmaceutical delivery of the therapeutic agents herein disclosed.
  • the nature of the carrier will depend on the mode of administration being employed.
  • parenteral formulations usually include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, sesame oil, glycerol, ethanol, combinations thereof, or the like, as a vehicle.
  • the carrier and composition can be sterile, and the formulation suits the mode of administration.
  • compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • non-toxic auxiliary substances such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • the composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder.
  • conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, sodium saccharine, cellulose, magnesium carbonate, or magnesium stearate.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Embodiments of the disclosure including medicaments can be prepared with conventional pharmaceutically acceptable carriers, adjuvants and counterions as would be known to those of skill in the art.
  • the amount of therapeutic agent effective in decreasing or inhibiting infection can depend on the nature of the pathogen and its associated disorder or condition, and can be determined by standard clinical techniques. Therefore, these amounts will vary depending on the type of virus, bacteria, fungus, parasite or other pathogen.
  • the dosage can be anywhere from 0.01 mg/kg to 100 mg/kg. Multiple dosages can also be administered depending on the type of pathogen, and the subject's condition.
  • in vitro assays can be employed to identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the disclosure also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Instructions for use of the composition can also be included.
  • Instructions for the use of a composition to decrease infection or toxicity can also be included.
  • instructions for decreasing infection by an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, a Hepatitis C virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus can be provided.
  • nucleic acid in an example in which a nucleic acid is employed to reduce infection, such as an antisense or siRNA molecule, the nucleic acid can be delivered intracellularly (for example by expression from a nucleic acid vector or by receptor-mediated mechanisms), or by an appropriate nucleic acid expression vector which is administered so that it becomes intracellular, for example by use of a retroviral vector (see U.S. Patent No.
  • siRNA carriers also include, polyethylene glycol (PEG), PEG-liposomes, branched carriers composed of histidine and lysine (HK polymers), chitosan-thiamine pyrophosphate carriers, surfactants (for example, Survanta and Infasurf), nanochitosan carriers, and D5W solution.
  • PEG polyethylene glycol
  • PEG-liposomes branched carriers composed of histidine and lysine
  • HK polymers branched carriers composed of histidine and lysine
  • chitosan-thiamine pyrophosphate carriers for example, Survanta and Infasurf
  • nanochitosan carriers for example, D5W solution.
  • D5W solution D5W solution.
  • the present disclosure includes all forms of nucleic acid delivery, including synthetic oligos, naked DNA, plasmid and viral delivery, integrated into the genome or not.
  • vector delivery can be via a viral system, such as a retroviral vector system which can package a recombinant retroviral genome (see e.g., Pastan et al., Proc. Natl. Acad. Sci. U.S.A. 85:4486, 1988; Miller et al., MoI. Cell Biol. 6:2895, 1986).
  • the recombinant retrovirus can then be used to infect and thereby deliver to the infected cells a nucleic acid, for example an antisense molecule or siRNA.
  • the exact method of introducing the altered nucleic acid into mammalian cells is, of course, not limited to the use of retroviral vectors. Other techniques are widely available for this procedure including the use of adenoviral vectors (Mitani et al., Hum. Gene Ther. 5:941-948, 1994), adeno-associated viral (AAV) vectors
  • Physical transduction techniques can also be used, such as liposome delivery and receptor-mediated and other endocytosis mechanisms (see, for example, Schwartzenberger et al., Blood 87:472-478, 1996) to name a few examples.
  • This invention can be used in conjunction with any of these or other commonly used gene transfer methods.
  • the present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, 2, 3 or 4, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a fungus or a parasite.
  • a pathogen such as a virus, a bacterium, a fungus or a parasite.
  • exemplary transgenic non-human mammals include, but are not limited to, ferrets, fish, guinea piags, chinchilla, mice, monkeys, rabbits, rats, chickens, cows, and pigs. Such knock-out animals are useful for reducing the transmission of viruses from animals to humans and for further validating a target.
  • one or both alleles of a gene set forth in Table 1, 2, 3 or 4 can be functionally deleted.
  • decreased susceptibility is meant that the animal is less susceptible to infection or experiences decreased infection by a pathogen as compared to an animal that does not have one or both alleles of a a gene set forth in Table 1, 2, 3 or 4 functionally deleted.
  • the animal does not have to be completely resistant to the pathogen.
  • the animal can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any percentage in between less susceptible to infection by a pathogen as compared to an animal that does not have a functional deletion of a gene set forth in Table 1, 2, 3 or 4.
  • decreasing infection or decreasing susceptibility to infection includes decreasing entry, replication, pathogenesis, insertion, lysis, or other steps in the replication strategy of a virus or other pathogen into a cell or subject, or combinations thereof. Therefore, the present invention provides a non- human transgenic mammal comprising a functional deletion of a gene set forth in Table 1 , 2, 3 or 4, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a parasite or a fungus.
  • a functional deletion is a mutation, partial or complete deletion, insertion, or other variation made to a gene sequence that inhibits production of the gene product or renders a gene product that is not completely functional or non-functional.
  • Functional deletions can be made by insertional mutagenesis (for example via insertion of a transposon or insertional vector), by site directed mutagenesis, via chemical mutagenesis, via radiation or any other method now known or developed in the future that results in a transgenic animal with a functional deletion of a gene set forth in Table 1, 2, 3 or 4. More particularly, the present invention provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not HIV.
  • non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 2, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not West Nile Virus.
  • non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 3, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
  • the present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 4, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not hepatitis C.
  • a nucleic acid sequence such as siRNA, antisense, LNA, a morpholino or another agent that interferes with a gene set forth in Table 1 , 2, 3 or 4 mRNA expression can be delivered.
  • the expression of the sequence used to knock-out or functionally delete the desired gene can be regulated by an appropriate promoter sequence.
  • constitutive promoters can be used to ensure that the functionally deleted gene is not expressed by the animal.
  • an inducible promoter can be used to control when the transgenic animal does or does not express the gene of interest.
  • Exemplary inducible promoters include tissue- specific promoters and promoters responsive or unresponsive to a particular stimulus (such as light, oxygen, chemical concentration, such as a tetracycline inducible promoter).
  • the transgenic animals of the present invention that comprise a functionally deleted a gene set forth in Table 1 , 2, 3 or 4 can be examined during exposure to various pathogens. Comparison data can provide insight into the life cycles of pathogens.
  • knock-out animals or functionally deleted such as birds or pigs
  • that are otherwise susceptible to an infection for example influenza
  • these transgenic animals can be bred to establish flocks or herds that are less susceptible to infection.
  • Transgenic animals including methods of making and using transgenic animals, are described in various patents and publications, such as WO 01/43540; WO 02/1981 1; U.S. Pub. Nos: 2001-0044937 and 2002-00661 17; and U.S. Pat. Nos: 5,859,308; 6,281,408; and 6,376,743; and the references cited therein.
  • the transgenic animals of this invention also include conditional gene knockdown animals produced, for example, by utilizing the SIRIUS-Cre system that combines siRNA for specific gene-knockdown, Cre-loxP for tissue-specific expression and tetracycline-on for inducible expression. These animals can be generated by mating two parental lines that contain a specific siRNA of interest gene and tissue-specific recombinase under tetracycline control. See Chang et al. "Using siRNA Technique to Generate Transgenic Animals with Spatiotemporal and Conditional Gene Knockdown.” American Journal of Pathology 165: 1535-1541 (2004) which is hereby incorporated in its entirety by this reference regarding production of conditional gene knockdown animals.
  • the present invention also provides cells including an altered or disrupted gene set forth in Table 1, 2, 3 or 4 that are resistant to infection by a pathogen. These cells can be in vitro, ex vivo or in vivo cells and can have one or both alleles altered. These cells can also be obtained from the transgenic animals of the present invention. Such cells therefore include cells having decreased susceptibility to a virus or any of the other pathogens described herein, including bacteria, parasites and fungi.
  • genes set forth herein are involved in viral infection, also provided herein are methods of overexpressing any of the genes set forth in Table 1 in host cells. Overexpression of these genes can provide cells that increase the amount of virus produced by the cell, thus allowing more efficient production of viruses. Also provided is the overexpression of the genes set forth herein in avian eggs, for example, in chicken eggs.
  • Methods of screening agents such as a chemical, a compound, a small or large molecule, an organic molecule, an inorganic molecule, a peptide, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an antisense nucleic acid or a ribozyme set forth using the transgenic animals described herein are also provided.
  • the nucleic acid or amino acid sequence of a subject can be isolated, sequenced, and compared to the wildtype sequence of a gene set forth in Table 1 , 2, 3 or 4. The greater the similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more susceptible that person is to infection, while a decrease in similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more resistant that subject can be to infection.
  • Such screens can be performed for any gene set forth in Table 1, 2, 3 or 4 for any species.
  • Assessing the genetic characteristics of a population can provide information about the susceptibility or resistance of that population to viral infection. For example, polymorphic analysis of alleles in a particular human population, such as the population of a particular city or geographic area, can indicate how susceptible that population is to infection. A higher percentage of alleles substantially similar to a wild-type gene set forth in Table 1, 2, 3 or 4 can indicate that the population is more susceptible to infection, while a large number of polymorphic alleles that are substantially different than a wild-type gene sequence can indicate that a population is more resistant to infection. Such information can be used, for example, in making public health decisions about vaccinating susceptible populations.
  • the present invention also provides a method of screening a cell for a variant form of a gene set forth in Table 1, 2, 3 or 4.
  • a variant can be a gene with a functional deletion, mutation or alteration in the gene such that the amount or activity of the gene product is altered.
  • These cells containing a variant form of a gene can be contacted with a pathogen to determine if cells comprising a naturally occurring variant of a gene set forth in Table 1, 2, 3 or 4 differs in their resistance to infection.
  • cells from an animal for example, a chicken, can be screened for a variant form of a gene set forth in Table 1, 2, 3 or 4.
  • a naturally occurring variant is found and chickens possessing a variant form of the gene in their genome are less susceptible to infection, these chickens can be selectively bred to establish flocks that are resistant to infection. By utilizing these methods, flocks of chickens that are resistant to avian flu or other pathogens can be established. Similarly, other animals can be screened for a variant form of a gene set forth in Table 1, 2, 3 or 4. If a naturally occurring variant is found and animals possessing a variant form of the gene in their genome are less susceptible to infection, these animals can be selectively bred to establish populations that are resistant to infection.
  • These animals include, but are not limited to, cats, dogs, fish, livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, mouse,monkey, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, flocks of chickens, geese, turkeys, ducks, pheasants, pigeons, doves etc.). Therefore, the present application provides populations of animals that comprise a naturally occurring variant of a gene set forth in Table 1, 2, 3 or 4 that results in decreased susceptibility to viral infection, thus providing populations of animals that are less susceptible to viral infection.
  • livestock for example, cattle, horses, pigs, sheep, goats, etc.
  • laboratory animals for example, mouse,monkey, rabbit, rat, gerbil, guinea pig, etc.
  • avian species for example, flocks of chickens, geese, turkeys, ducks, pheasants, pigeons
  • the present invention provides a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection of a cell by a pathogen comprising: a) contacting a compound with a gene product set forth in Table 1, 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating the binding with a decrease in infection by the pathogen.
  • the present invention provides a method of identifying an agent that decreases infection of a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4; and b) detecting the level and/or activity of the gene product produced by the cellular gene, a decrease or elimination of the gene product and/or gene product activity indicating an agent with antipathogenic activity.
  • Also provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; b) contacting the cell with a pathogen, wherein the pathogen is not HIV; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
  • a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 2; b) contacting the cell with a pathogen, wherein the pathogen is not West Nile Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
  • a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 3; b) contacting the cell with a pathogen, wherein the pathogen is not West Nile Virus or Dengue Fever Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
  • the present invention also provides a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 4; b) contacting the cell with a pathogen, wherein the pathogen is not hepatitis C virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
  • This method can further comprise measuring the level of expression and/or activity of the gene product.
  • the level of infection can be determined by determining the level of replication of the pathogen.
  • the present invention also provides a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection by three or more pathogens comprising: a) contacting a compound with a gene product set forth in Table 1, 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens.
  • This method can further comprise optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens.
  • This method can be cell based or an in vivo assay.
  • the three or more pathogens can be any three or more pathogens set forth herein.
  • the three or more pathogens can be respiratory pathogens selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses or adenoviruses.
  • the three or more pathogens can be gastrointestinal pathogens selected from filoviruses, flaviviruses, calciviruses and reoviruses.
  • the three or more pathogens can also be a combination of respiratory and gastrointestinal viruses.
  • the three or more pathogens can be selected from the group consisting of : an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an
  • the cell population used in the method can be the same cell population for each pathogen or can be different cell populations.
  • the agent would be administered to a different cell population for each pathogen assayed.
  • the pathogens are viruses
  • a cell population is contacted with the agent and a first virus
  • another cell population is contacted with the agent and second virus
  • a third cell population is contacted with the agent and a third virus etc. in order to determine whether the agent inhibits infection by three or more viruses. Since the cell type will vary depending on whether or not a given virus can infect the cell, one of skill in the art would know how to pair the cell type with the virus in order to perform the assay.
  • This method can further comprise measuring the level of expression and/or activity of the gene product set forth in Table 1, 2, 3 or 4.
  • This method can further comprise associating the level of infection with the level of expression and/or activity a gene product set forth in Table 1, 2, 3 or 4.
  • the level of infection can be measured, for example, by measuring viral replication.
  • the agent has previously been identified as an agent that decreases or inhibits the level and/or activity of a gene product set forth in Table 1, 2, 3 or 4
  • this can indicate a decrease in infection.
  • Previous identification of the agent can occur via experimentation, literature, chemical databases, catalogues, or any other means that allow one of skill in the art to associate the agent as an inhibitor of the gene or gene product.
  • a decrease in infection as compared to infection in a cell that was not contacted with the agent known to decrease or inhibit the level and/or activity of the gene product can be sufficient to identify the agent as an agent that decreases or inhibits infection.
  • the methods described above can be utilized to identify any agent with an activity that decreases infection, prevents infection or promotes cellular survival after infection with a pathogen(s). Therefore, the cell can be contacted with a pathogen before, or after being contacted with the agent. The cell can also be contacted concurrently with the pathogen and the agent.
  • the agents identified utilizing these methods can be used to inhibit infection in cells either in vitro, ex vivo or in vivo.
  • any cell that can be infected with a pathogen can be utilized.
  • the cell can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli.
  • the cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture.
  • the cell can also be in a nonhuman subject thus providing in vivo screening of agents that decrease infection by a pathogen.
  • Cells susceptible to infection are well known and can be selected based on the pathogen of interest.
  • test agents or compounds used in the methods described herein can be, but are not limited to, chemicals, small molecules, inorganic molecules, organic molecules, drugs, proteins, cDNAs, aptamers, large molecules, antibodies, morpholinos, triple helix molecule, peptides, siRNAs, shRNAs, miRNAs, LNAs, antisense RNAs, ribozymes or any other compound.
  • the compound can be random or from a library optimized to bind to a gene or gene product set forth in Table 1, 2, 3 or 4.
  • Drug libraries optimized for the proteins in the class of proteins provided herein can also be screened or tested for binding or activity.
  • Compositions identified with the disclosed approaches can be used as lead compositions to identify other compositions having even greater antipathogenic activity.
  • chemical analogs of identified chemical entities, or variants, fragments or fusions of peptide agents can be tested for their ability to decrease infection using the disclosed assays.
  • Such analogs can be purchased or synthesized via routine methods known to those of skill in the art.
  • Candidate agents can also be tested for safety in animals and then used for clinical trials in animals or humans.
  • the level of infection can be assessed by measuring an antigen or other product associated with a particular infection.
  • the level of viral infection can be measured by real-time quantitative reverse transcription-poly merase chain reaction (RT-PCR) assay (See for example, Payungporn et al. "Single step multiplex real-time RT-PCR for H5N1 influenza A virus detection.” J Virol Methods. Sep 22, 2005; Landolt et Ia.
  • RT-PCR real-time quantitative reverse transcription-poly merase chain reaction
  • composition is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between.
  • the level of the gene product can be measured by any standard means, such as by detection with an antibody specific for the protein.
  • the nucleic acids set forth herein and fragments thereof can be utilized as primers to amplify nucleic acid sequences, such as a gene transcript of a gene set forth in Table 1, 2, 3 or 4 by standard amplification techniques.
  • expression of a gene transcript can be quantified by real time PCR using RNA isolated from cells.
  • PCR techniques are familiar to those skilled in the art. For a review of PCR technology, see White (1997) and the publication entitled “PCR Methods and Applications” (1991, Cold Spring Harbor Laboratory Press), which is incorporated herein by reference in its entirety for amplification methods.
  • PCR primers on either side of the nucleic acid sequences to be amplified are added to a suitably prepared nucleic acid sample along with dNTPs and a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase.
  • a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase.
  • the nucleic acid in the sample is denatured and the PCR primers are specifically hybridized to complementary nucleic acid sequences in the sample.
  • the hybridized primers are extended. Thereafter, another cycle of denaturation, hybridization, and extension is initiated. The cycles are repeated multiple times to produce an amplified fragment containing the nucleic acid sequence between the primer sites.
  • PCR has further been described in several patents including U.S. Pat. Nos.
  • a detectable label may be included in an amplification reaction.
  • Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein (6-FAM), 2',7'-dimethoxy-4',5'-dichloro-6- carboxyfluorescein (JOE), 6-carboxy-X-rhodamine (ROX), 6-carboxy-2',4',7',4,7- hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N',N'-tetramethyl-6- carboxyrhodamine (TAMRA), radioactive labels, e.g., 32 P, 35 S, 3 H; etc.
  • FITC fluorescein isothiocyanate
  • rhodamine Texas Red
  • the label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label.
  • the label may be conjugated to one or both of the primers.
  • the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.
  • the sample nucleic acid e.g. amplified fragment
  • the nucleic acid can be sequenced by dideoxy or other methods. Hybridization with the sequence can also be used to determine its presence, by Southern blots, dot blots, etc.
  • the level of gene product can be compared to the level of the gene product in a control cell not contacted with the compound.
  • the level of gene product can be compared to the level of the gene product in the same cell prior to addition of the compound.
  • Activity or function can be measured by any standard means, such as by enzymatic assays that measure the conversion of a substrate to a product or binding assays that measure the binding of a gene product set forth in Table 1, 2, 3 or 4 to another protein, for example.
  • the regulatory region of a gene set forth in Table 1 , 2, 3 or 4 can be functionally linked to a reporter gene and compounds can be screened for inhibition of reporter gene expression.
  • Such regulatory regions can be isolated from genomic sequences and identified by any characteristics observed that are characteristic for regulatory regions of the species and by their relation to the start codon for the coding region of the gene.
  • a reporter gene encodes a reporter protein.
  • a reporter protein is any protein that can be specifically detected when expressed. Reporter proteins are useful for detecting or quantitating expression from expression sequences. Many reporter proteins are known to one of skill in the art. These include, but are not limited to, ⁇ -galactosidase, luciferase, and alkaline phosphatase that produce specific detectable products.
  • Fluorescent reporter proteins can also be used, such as green fluorescent protein (GFP), cyan fluorescent protein (CFP), red fluorescent protein (RFP) and yellow fluorescent protein (YFP). Viral infection can also be measured via cell-based assays. Briefly, cells (20,000 to
  • the antiviral agent can be applied to the cells before, during, or after infection with the pathogen.
  • the amount of virus and agent administered can be determined by skilled practitioners. In some examples, several different doses of the potential therapeutic agent can be administered, to identify optimal dose ranges. Following transfection, assays are conducted to determine the resistance of the cells to infection by various agents.
  • the presence of a viral antigen can be determined by using antibody specific for the viral protein then detecting the antibody.
  • the antibody that specifically binds to the viral protein is labeled, for example with a detectable marker such as a fluorophore.
  • the antibody is detected by using a secondary antibody containing a label. The presence of bound antibody is then detected, for example using microscopy, flow cytometry and ELISA. Similar methods can be used to monitor bacterial, protozoal, or fungal infection (except that the antibody would recognize a bacterial, protozoal, or fungal protein, respectively).
  • the ability of the cells to survive viral infection is determined, for example, by performing a cell viability assay, such as trypan blue exclusion. Plaque assays can be utilized as well.
  • the amount of protein in a cell can be determined by methods standard in the art for quantitating proteins in a cell, such as Western blotting, ELISA, ELISPOT, immunoprecipitation, immunofluorescence (e.g., FACS), immunohistochemistry, immunocytochemistry, etc., as well as any other method now known or later developed for quantitating protein in or produced by a cell.
  • methods standard in the art for quantitating proteins in a cell such as Western blotting, ELISA, ELISPOT, immunoprecipitation, immunofluorescence (e.g., FACS), immunohistochemistry, immunocytochemistry, etc., as well as any other method now known or later developed for quantitating protein in or produced by a cell.
  • the amount of a nucleic acid in a cell can be determined by methods standard in the art for quantitating nucleic acid in a cell, such as in situ hybridization, quantitative PCR, RT-PCR, Taqman assay, Northern blotting, ELISPOT, dot blotting, etc., as well as any other method now known or later developed for quantitating the amount of a nucleic acid in a cell.
  • an antiviral agent to prevent or decrease infection by a virus, for example, any of the viruses listed above, can be assessed in an animal model.
  • animal models for viral infection are known in the art. For example, mouse HIV models are disclosed in Sutton et al. (Res. Initiat Treat. Action, 8:22-4, 2003) and Pincus et al. (AIDS Res. Hum. Retroviruses 19:901-8, 2003); guinea pig models for Ebola infection are disclosed in Parren et al. (J. Virol. 76:6408-12, 2002) and Xu et al. (Nat. Med.
  • chimpanzee models for HCV are disclosed (Lanford et al., ILAR J. 2001 ;42(2):117-26); cynomolgus monkey (Macaca fascicularis) models for influenza infection are disclosed in Kuiken et al. (Vet. Pathol. 40:304- 10, 2003); mouse models for RSV are also disclosed (Sudo et al. Antivir Chem Chemother. 1999 May; 10(3): 135-9) mouse models for herpes are disclosed in Wu et al. (Cell Host Microbe 22:5(l):84-94. 2009); mouse models for rhinovorus are disclosed (Bartlett et al. Nat Med.
  • Vaccine 26(52): 6874-82, 2008 Other animal models for influenza infection are also available. These include, but are not limited to, a cotton rat model disclosed by Ottolini et al. (J. Gen. Virol, 86(Pt 10): 2823-30, 2005), as well as ferret and mouse models disclosed by Maines et al. (J. Virol. 79(18): 11788-11800, 2005).
  • One of skill in the art would know how to select an animal model for assessing the in vivo activity of an agent for its ability to decrease infection by viruses, bacteria, fungi and parasites.
  • LD50 is an index of toxicity (lethal dose 50%), the amount of the substance that kills 50% of the test population of experimental animals when administered as a single dose.
  • ED 50 is the dose of a drug that is pharmacologically effective for 50% of the population exposed to the drug or a 50% response in a biological system that is exposed to the drug.
  • Animal models can also be used to assess antibacterial, antifungal and antiparasitic agents.
  • Animals of any species including, but not limited to, birds, ferrets, cats, mice, rats, rabbits, fish (for example, zebrafish or koi) guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, can be used to generate an animal model of viral infection, bacterial infection, fungal infection or parasitic infection if needed.
  • the appropriate animal is inoculated with the desired virus, in the presence or absence of the antiviral agent.
  • the amount of virus and agent administered can be determined by skilled practitioners.
  • the potential therapeutic agent for example, an antiviral agent
  • the therapeutic agent can be administered before, during, or after infection with the virus.
  • animals are observed for the development of the appropriate viral infection and symptoms associated therewith.
  • a decrease in the development of the appropriate viral infection, or symptoms associated therewith, in the presence of the agent provides evidence that the agent is a therapeutic agent that can be used to decrease or even inhibit viral infection in a subject.
  • a virus can be tested which is lethal to the animal and survival is assessed.
  • the weight of the animal or viral titer in the animal can be measured. Similar models and approaches can be used for bacterial, fungal and parasitic infections.
  • the level of infection can be associated with the level of gene expression and/or activity, such that a decrease or elimination of infection associated with a decrease or elimination of gene expression and/or activity indicates that the agent is effective against the pathogen.
  • the level of infection can be measured in a cell after administration of siRNA that is known to inhibit a gene product set forth in Table 1, 2, 3 or 4. If there is a decrease in infection then the siRNA is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between.
  • the level of expression and/or activity of can be measured utilizing the methods set forth above and associated with the level of infection.
  • a decrease in expressionand/or activity By correlating a decrease in expressionand/or activity with a decrease in infection, one of skill in the art can confirm that a decrease in infection is effected by a decrease in expression and/or activity of a gene or gene product set forth in Table 1, 2, 3 or 4.
  • the level of infection can be measured in a cell, utilizing the methods set forth above and known in the art, after administration of a chemical, small molecule, drug, protein, cDNA, antibody, aptamer, shRNA, miRNA, morpholino, antisense RNA, LNA, ribozyme or any other compound. If there is a decrease in infection, then the chemical, small molecule, drug, protein, cDNA, antibody, shRNA, miRNA, morpholino, antisense RNA, ribozyme or any other compound is an effective antpathogenic agent.
  • the genes and nucleic acids of the invention can also be used in polynucleotide arrays.
  • Polynucleotide arrays provide a high throughput technique that can assay a large number of polynucleotide sequences in a single sample.
  • This technology can be used, for example, to identify samples with reduced expression of as compared to a control sample.
  • This technology can also be utilized to determine the effects of reduced expression of a gene set forth in Table 1, 2, 3 or 4 on other genes.
  • single-stranded polynucleotide probes can be spotted onto a substrate in a two-dimensional matrix or array.
  • Each single-stranded polynucleotide probe can comprise at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or 30 or more contiguous nucleotides selected from nucleotide sequences set forth under GenBank Accession Nos. herein and other nucleic acid sequences that would be selected by one of skill in the art depending on what genes, in addition to one or more of the genes set forth in Table 1 , 2, 3 or 4 are being analyzed.
  • the array can also be a microarray that includes probes to different polymorphic alleles of these genes.
  • a polymorphism exists when two or more versions of a nucleic acid sequence exist within a population of subjects.
  • a polymorphic nucleic acid can be one where the most common allele has a frequency of 99% or less.
  • Different alleles can be identified according to differences in nucleic acid sequences, and genetic variations occurring in more than 1% of a population (which is the commonly accepted frequency for defining polymorphism) are useful polymorphisms for certain applications.
  • the allelic frequency (the proportion of all allele nucleic acids within a population that are of a specified type) can be determined by directly counting or estimating the number and type of alleles within a population. Polymorphisms and methods of determining allelic frequencies are discussed in Hartl, D.L. and Clark, A.G., Principles of Population Genetics, Third Edition (Sinauer Associates, Inc., Sunderland Massachusetts, 1997), particularly in
  • microarrays can be utilized to detect polymorphic alleles in samples from subjects. Such alleles may indicate that a subject is more susceptible to infection or less susceptible to infection.
  • microarrays can be utilized to detect polymorphic versions of genes set forth in Table 1 , 2, 3 or 4 that result in decreased gene expression and/or decreased activity of the gene product to identify subjects that are less susceptible to viral infection.
  • the existence of an allele associated with decreased expression in a healthy individual can be used to determine which genes are likely to have the least side effects if the gene product is inhibited or bound or may be selected for in commercial animals and bred into the population.
  • the substrate can be any substrate to which polynucleotide probes can be attached, including but not limited to glass, nitrocellulose, silicon, and nylon.
  • Polynucleotide probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions. Techniques for constructing arrays and methods of using these arrays are described in EP No. 0 799 897; PCT No. WO 97/29212; PCT No. WO 97/27317; EP No. 0 785 280; PCT No. WO 97/02357; U.S. Pat. Nos. 5,593,839; 5,578,832; EP No. 0 728 520; U.S. Pat. No.
  • the present invention also provides a method of identifying an agent that can decrease infection by three or more pathogens comprising: a) administering the agent to three or more cell populations containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4; b) contacting the three or more cell populations with a pathogen selected wherein each population is contacted with a different pathogen; and c) determining the level of infection, a decrease or elimination of infection by three or more pathogens indicating that the agent is an agent that decreases infection by three or more pathogens.
  • the three or more pathogens can be three or more respiratory viruses selected from the one or more families from group consisting of: picornaviruses, an orthomyxoviruses, a paramyxoviruses, a coronaviruses, or an adenoviruses.
  • the three or more pathogens can be three or more gastrointestinal viruses selected from one or more families from the group consisting of: flaviviruses, filoviruses, calciviruses or reoviruses.
  • the three or more pathogens can be three or more viruses selected from gastrointestinal viruses and respiratory viruses.
  • the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, West Nile virus, Chikungunya virus or a Dengue fever virus.
  • an HIV virus a pox virus
  • a herpes virus an influenza
  • the three or more pathogens can also be selected from the group consisting of: HIV, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.
  • the three or more pathogens can be selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus
  • Also provided is a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection by three or more pathogens comprising: a) contacting a compound with a gene product set forth in Table 1 , 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens.
  • This method can further comprise optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens.
  • a set forth above, as utilized herein "three or more” includes four or more, five or more, six or more, seven or more, etc. This assay can be cell based or in vivo based.
  • the present invention provides_a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; c) contacting the cell with an infectious pathogen, wherein the pathogen is not HIV; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
  • Also provided is a method of making a compound that decreases infection of a cell by a pathogen comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 2; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile Virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
  • a method of making a compound that decreases infection of a cell by a pathogen comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 3; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile virus or Dengue fever virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
  • the present invention also provides_a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 4; c) contacting the cell with an infectious pathogen, wherein the pathogen is not hepatitis C; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
  • the association can be made by measuring the level of expression and/or activity of the gene product.
  • the association can also be made by one of skill in the art if there is a priori knowledge, for example, via experimentation, literature, chemical databases or catalogues that the agent decreases expression and or activity of a gene or gene product.
  • the level of infection can be determined by determining the level of replication of the pathogen.
  • a method of making a compound that decreases infection in a cell by a pathogen comprising: a) optimizing a compound to bind a gene product set forth in Table 1, 2, 3 or 4; b) administering the compound to a cell containing a cellular gene encoding the gene product; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating the making of a compound that decreases infection in a cell by a pathogen.
  • This method can further synthesizing therapeutic quantities of the compound.
  • the method can further comprise packaging of the therapeutic quantities.
  • the present invention also provides a method of synthesizing a compound that binds to a gene product set forth in Table 1 , 2, 3 or 4 and decreases infection by a pathogen comprising: a) contacting a library of compounds with a gene product set forth in Table 1, 2, 3 or 4; b) associating binding with a decrease in infection; and c) synthesizing derivatives of the compounds from the library that bind to the gene product.
  • the present invention also provides a business method to reduce the cost of drug discovery of drugs that can reduce infection by a pathogen comprising: screening, outside of the United States, for drugs that reduce infection by binding to or reducing the function of a gene product set forth in Table 1, 2, 3 or 4; and b) importing drugs that reduce infection into the United States. Also provided is a method of making drugs comprising directing the synthesis of drugs that reduce infection by binding to or reducing the function of a gene or gene product set forth in Table 1, 2, 3 or 4. EXAMPLES siRNA studies
  • any of the genes set forth in Table 1, 2, 3 or 4 is further analyzed by contacting cells comprising a gene set forth in Table 1, 2, 3 or 4 with siRNA that specifically targets the gene product of the gene, and any pathogen set forth herein to identify the gene as a gene involved in pathogenic infection (for example, and not to be limiting, an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Ban- Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE,
  • siRNA Transfections can be performed as follows: Pools of 4 duplexed siRNA molecules targeting a gene of interest are reconstituted to a final working concentration of 5OuM as directed by the manufacturer (Qiagen). Twenty-four hours prior to transfection, cells are plated in 6-well dishes at 3x10 5 cells per well, such that at the time of transfection, the cells are approximately 30% confluent.
  • transfection reagent Lipofectamine2000 (Invitrogen) or Oligofectamine (Invitrogen) are diluted in Opti-MEM as directed by the manufacturer. Following a 5 minute incubation at room temperature, the diluted siRNA is added to the transfection reagent mixture, and incubated for an additional 20 minutes prior to adding to independent wells of the 6-well dishes. Transfections are incubated at 37°C for 48 hours without changing the medium.
  • Virus Infections Following 48-hour transfection, medium is aspirated from 6-well plates. Viruses are diluted in the appropriate medium and 500ul of either virus-free medium or virus dilution is added to each well, and adsorption is allowed to occur at the appropriate temperature for 1 hour. Following adsorption, inoculum is aspirated off the cells, cells are washed once with IX phosphate buffered saline, and 2ml growth medium is added to the cells. The infected cells are incubated for 72 hours at the appropriate temperature prior to harvesting samples for viral titration.
  • Viral Genomic Extractions Seventy-hours after inoculating cells, medium is harvested from 6-well dishes and centrifuged for 2 minutes at 10,000 rpm to remove any cellular debris. 200ul of clarified medium is added to 25ul Proteinase K, to which 200ul PureLink96 Viral RNA/DNA lysis buffer (Invitrogen) is added according to the manufacturer. Samples were processed and viral genomic RNA or DNA is extracted using an epMotion 5075 robotics station (Eppendorf) and the PureLink96 Viral RNA/DNA kit (Invitrogen).
  • RNA and Quantitative Real-Time PCR Reactions 3ul of extracted viral RNA is converted to cDNA using M-MLV reverse transcriptase (Invitrogen) and AmpliTaq Gold PCR buffer (Applied Biosystems). MgCl 2 , dNTPs and RNAseOUT (Invitrogen) are added to achieve a final concentration of 5mM, ImM and 2U/ul, respectively. Random hexamers (Applied Biosystems).
  • Quantitative real-time PCR reactions are set up in lOul volumes using IuI of template cDNA or extracted viral DNA using virus-specific TaqMan probes (Applied Biosystems) and RealMasterMix (Eppendorf). 2-step reactions are allowed to proceed through 40 to 50 cycles on an ep RealPlex thermocycler (Eppendorf). Quantitative standards for real-time PCR are constructed by cloning purified amplicons into pCR2-TOPO (Invitrogen) and sequenced as necessary. The amount of viral replication in the cells contacted with siRNA to the gene of interest is calculated and compared to the amount of viral replication in control cells that did not receive siRNA targeting the gene of interest.
  • Compounds are assessed as follows. Duplicate experiments are performed as follows. Prior to compound exposure cells are seeded into 12-well tissue culture plates and allowed to adhere to the vessel surface. The compounds are serially diluted and lOO ⁇ l is added to the growth medium in triplicate wells to obtain the indicated final concentration of compound or solvent. Cells are incubated in the presence of the test compound for about 24 hours at 37°C under 5% CO 2 .
  • the pre-treatment medium is then aspirated off the cells, and the specified virus, diluted in serum- and compound- free growth medium, is added to the cells. Virus is allowed to adsorb to the cells at 37°C under 5% CO 2 . Following adsorption, the viral inoculum is aspirated off the cells and replaced with growth medium supplemented with 2% FBS, 1% L-glutamine, 1% penicillin/streptomycin and the respective concentration of test compound. Cells are incubated under the same conditions for an additional 72 hours. As stated above, incubation period can vary depending on the pathogen.
  • RNA or DNA is extracted using the PureLink 96 Viral RNA/DNA extraction kit (Invitrogen) as directed by the manufacturer.
  • Viral nucleic acid is subjected to reverse transcription to generate cDNA, which is subsequently used as the template for quantitative real-time TaqMan PCR to titrate viral replication using a Mastercycler ep realplex 2 (Eppendorf).
  • the experiment described above can be performed to determine the effect of any of the compounds set forth herein on any pathogenic infection, for example, and not to be limiting, a pox virus, BVDV, HIV, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, CaI ici viruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE 5 WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus
  • any of the experiments set forth herein can optionally comprise the step of assessing toxicity of any of the compounds set forth herein via any of the toxicity measurement methods described herein, or via any of the toxicity measurement methods known to one of skill in the art, such as, for example, the CytoTox-Glo assay (see Niles, A. et al. (2007) Anal. Biochem. 366, 197-206) or the Cell-Titer-Glo assay from Promega.

Abstract

The present invention relates to cellular proteins that are involved in infection or are otherwise associated with the life cycle of one or more pathogens.

Description

MAMMALIAN GENES INVOLVED IN INFECTION
This application claims the benefit of U.S. Application No. 61/211,178, filed on March 27, 2009, which is hereby incorporated in its entirety by this reference.
FIELD OF THE INVENTION
The present invention relates to nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of one or more pathogens, such as a virus, a bacteria, a fungus or a parasite. The invention also relates to modulators of nucleic acid sequences and cellular proteins encoded by these sequences that are involved in infection or are otherwise associated with the life cycle of a pathogen.
BACKGROUND
Infectious diseases affect the health of people and animals around the world, causing serious illness and death. Black Plague devastated the human population in Europe during the middle ages. Pandemic flu killed millions of people in the 20lh century and is a threat to reemerge.
Some of the most feared, widespread, and devastating human diseases, are caused by viruses that interfere with normal cellular processes. These include influenza, poliomyelitis, smallpox, Ebola, yellow fever, measles and AIDS, to name a few. Viruses are also responsible for many cases of human disease including encephalitis, meningitis, pneumonia, hepatitis and cervical cancer, warts and the common cold. Furthermore, viruses causing respiratory infections, and diarrhea in young children lead to millions of deaths each year in less-developed countries. Also, a number of newly emerging human diseases such as SARS are caused by viruses. In addition, the threat of a bioterrorist-designed pathogen is ever present.
While vaccines have been effective to prevent certain viral infections, relatively few vaccines are available or wholly effective, have inherent risks and tend to be specific for particular conditions. Vaccines are of limited value against rapidly mutating viruses and cannot anticipate emerging viruses or new bioterrorist-designed viruses. Currently there is no good answer to these threats. Traditional treatments for viral infection include pharmaceuticals aimed at specific virus derived proteins, such as HIV protease or reverse transcriptase, or the administration of recombinant (cloned) immune modulators (host derived), such as the interferons. However, the vast majority of viruses lack an effective drug. Those drugs that exist have several limitations and drawbacks that including limited effectiveness, toxicity, and high rates of viral mutations which render antiviral pharmaceuticals ineffective. Thus, an urgent need exists for alternative treatments for viruses and other infectious diseases, and methods of identifying new drugs to combat these threats. SUMMARY OF THE INVENTION
The present invention provides genes and gene products set forth in Tables 1, 2, 3 and 4 that are involved in infection by one or more pathogens such as a virus, a parasite, a bacteria or a fungus, or are otherwise associated with the life cycle of a pathogen. Also provided are methods of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more of these genes or gene products set forth in Tables 1, 2, 3 and 4. Also provided are methods of decreasing infection by a pathogen in a subject by administering an agent that decreases the expression and/or activity of of the genes or gene products set forth in Tables 1, 2, 3 and 4. Further provided are methods of identifying an agent that decreases infection by a pathogen.
DETAILED DESCRIPTION OF THE INVENTION
The present invention may be understood more readily by reference to the following detailed description of preferred embodiments of the invention and the Examples included therein.
Before the present compounds, compositions and/or methods are disclosed and described, it is to be understood that this invention is not limited to specific nucleic acids, specific polypeptides, or to particular methods, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise. The term "or" refers to a single element of stated alternative elements or a combination of two or more elements, unless the context clearly indicates otherwise. As used herein, "comprises" means "includes." Thus, "comprising A or B," means "including A, B, or A and B," without excluding additional elements.
Ranges may be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, the phrase "optionally obtained prior to treatment" means obtained before treatment, after treatment, or not at all. As used throughout, by "subject" is meant an individual. Preferably, the subject is a mammal such as a primate, and, more preferably, a human. Non-human primates include marmosets, monkeys, chimpanzees, gorillas, orangutans, and gibbons, to name a few. The term "subject" includes domesticated animals, such as cats, dogs, etc., livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, ferret, chinchilla, mouse, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, chickens, turkeys, ducks, pheasants, pigeons, doves, parrots, cockatoos, geese, etc.). The subjects of the present invention can also include, but are not limited to fish (for example, zebrafish, koi, goldfish, tilapia, salmon and trout), amphibians and reptiles.
In the present application, the genes listed in Table 1 , 2, 3 or 4 are host genes involved in viral infection. As utilized throughout, "gene product" is the RNA or protein resulting from the expression of a gene listed in Table 1, 2, 3 or 4.
The nucleic acids of these genes and their encoded proteins can be involved in all phases of the viral life cycle including, but not limited to, viral attachment to cellular receptors, viral infection, viral entry, internalization, disassembly of the virus, viral replication, genomic integration of viral sequences, transcription of viral RNA, translation of viral mRNA, transcription of cellular proteins, translation of cellular proteins, trafficking, proteolytic cleavage of viral proteins or cellular proteins, assembly of viral particles, budding, cell lysis and egress of virus from the cells.
Although the genes set forth herein were identified as cellular genes involved in viral infection, as discussed throughout, the present invention is not limited to viral infection.
Therefore, any of these nucleic acid sequences and the proteins encoded by these sequences can be involved in infection by any infectious pathogen such as a bacteria, a fungus or a parasite which includes involvement in any phase of the infectious pathogen's life cycle.
As utilized herein, when referring to any of the genes in Table 1, 2, 3 or 4, for example, and not to be limiting, ABTBl , this includes any ABTBl gene, ABTBl gene product, for example, an ABTBl nucleic acid (DNA or RNA) or ABTBl protein, from any organism that retains at least one activity of ABTBl and can function as an ABTBl nucleic acid or protein utilized by a pathogen. For example, the nucleic acid or protein sequence can be from or in a cell in a human, a non-human primate, a mouse, a rat, a cat, a dog, a chimpanzee, a horse, a cow, a pig, a sheep, a guinea pig, a rabbit, a zebrafish, a chicken, to name a few. As used herein, a gene is a nucleic acid sequence that encodes a polypeptide under the control of a regulatory sequence, such as a promoter or operator. The coding sequence of the gene is the portion transcribed and translated into a polypeptide (in vivo, in vitro or in situ) when placed under the control of an appropriate regulatory sequence. The boundaries of the coding sequence can be determined by a start codon at the 5' (amino) terminus and a stop codon at the 3' (carboxyl) terminus. If the coding sequence is intended to be expressed in a eukaryotic cell, a polyadenylation signal and transcription termination sequence can be included 3' to the coding sequence.
Transcriptional and translational control sequences include, but are not limited to, DNA regulatory sequences such as promoters, enhancers, and terminators that provide for the expression of the coding sequence, such as expression in a host cell. A polyadenylation signal is an exemplary eukaryotic control sequence. A promoter is a regulatory region capable of binding RNA polymerase and initiating transcription of a downstream (3' direction) coding sequence. Additionally, a gene can include a signal sequence at the beginning of the coding sequence of a protein to be secreted or expressed on the surface of a cell. This sequence can encode a signal peptide, N-terminal to the mature polypeptide, which directs the host cell to translocate the polypeptide.
Tables 1 , 2, 3 and 4, provide the Entrez Gene numbers for the human genes set forth herein. The information provided under the Entrez Gene numbers listed in Tables 1 , 2, 3 and 4 is hereby incorporated entirely by this reference. One of skill in the art can readily obtain this information from the National Center for Biotechnology Information at the National Library of Medicine (http://www.ncbi. nlm.nih.gov/entrez/query. fcgi?db=gene). By accessing Entrez Gene, one of skill in the art can readily obtain information about every gene listed in Table 1, 2, 3 or 4, such as the genomic location of the gene, a summary of the properties of the protein encoded by the gene, expression patterns, function, information on homologs of the gene as well as numerous reference sequences, such as the genomic, mRNA and protein sequences for each gene. Therefore, one of skill in the art can readily obtain sequences, such as genomic, mRNA and protein sequences by accessing information available under the Entrez Gene number provided for each gene. Thus, all of the information readily obtained from the Entrez Gene Nos. set forth herein is also hereby incorporated by reference in its entirety. These examples are not meant to be limiting as one of skill in the art would know that sequences for the genes and gene products listed in Table 1 , 2, 3 or 4 from human or from other species are available at GenBank (Benson et al. Nucleic Acids Res. 2004 January 1; 32(Database issue); D23-D26), the EMBL Database (Stoesser et al., (2000) Nucleic Acids Res., 28, 19-23) or other sequence databases. Table 5 (column 2) provides one or more aliases for genes set forth herein. Therefore, it is clear that when referring to a gene, this also includes known alias(es) and any aliases attributed to the genes listed herein, in the future. The proteins encoded by the genes, if available, are also listed in column 3 of Table 5. In addition to the function of being involved in pathogenic infection as provided herein, a function of the proteins is also provided, if available, in column 4 of Table 5. The chromosomal location of the gene in the human genome (column 5) is also set forth. Thus, the present invention identifies a genomic loci of genes associated with viral infection. By identifying the gene and its location in the genome, the invention provides both the gene and its product(s) as targets for therapies such as antiviral, antibacterial, antifungal and antiparasitic therapies, to name a few. Also provided in Table 5 are the GenBank Accession Nos. for the coding sequences
(human mRNA sequences) (column 6) and the GenBank Accession Nos. for the human protein sequences (column 7), for genes and gene products set forth in Tables 1, 2, 3 or 4. It is understood that in any coding sequence, a T can be replaced with a U to obtain an RNA sequence for each gene. The nucleic acid sequences and protein sequences provided under the GenBank Accession numbers mentioned herein are hereby incorporated in their entireties by this reference. One of skill in the art would know that the nucleotide sequences provided under the GenBank Accession numbers set forth herein can be readily obtained from the National Center for Biotechnology Information at the National Library of Medicine (http://www.ncbi. nlm.nih.gov/entrez/query.fcgi?db=nucleotide). Similarly, the protein sequences set forth herein can be readily obtained from the National Center for Biotechnology Information at the National Library of Medicine
(http://www.ncbi.nlm. nih.gov/entrez/query.fcgi?db=protein). The nucleic acid sequences and protein sequences provided under the GenBank Accession numbers mentioned herein are hereby incorporated in their entireties by this reference. Compounds that can be utilized to inhibit genes or gene products set forth herein are also set forth in Table 5.
TABLE 1 maBBgHMHi ■H HJ
339010 A26B1
53947 A4GALT
404744 AAAl
22848 AAKl
10157 AASS
28 ABO
80325 ABTBl
32 ACACB
36 ACADSB
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000007_0002
Figure imgf000008_0001
Figure imgf000008_0002
Figure imgf000008_0003
Figure imgf000009_0001
Figure imgf000009_0002
Figure imgf000009_0003
Figure imgf000010_0001
Figure imgf000010_0002
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000012_0001
Figure imgf000012_0002
Figure imgf000012_0003
Figure imgf000013_0001
Figure imgf000013_0002
Figure imgf000013_0003
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
TABLE 2
Figure imgf000015_0004
Figure imgf000015_0005
Figure imgf000016_0001
Figure imgf000016_0002
TABLE 3
Figure imgf000017_0001
Figure imgf000017_0002
TABLE 4
Figure imgf000017_0003
Figure imgf000017_0004
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000019_0002
Table 5
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
As used herein, the term "nucleic acid" refers to single or multiple stranded molecules which may be DNA or RNA, or any combination thereof, including modifications to those nucleic acids. The nucleic acid may represent a coding strand or its complement, or any combination thereof. Nucleic acids may be identical in sequence to the sequences which are naturally occurring for any of the moieties discussed herein or may include alternative codons which encode the same amino acid as that which is found in the naturally occurring sequence. These nucleic acids can also be modified from their typical structure. Such modifications include, but are not limited to, methylated nucleic acids, the substitution of a non-bridging oxygen on the phosphate residue with either a sulfur (yielding phosphorothioate deoxynucleotides), selenium (yielding phosphorselenoate deoxynucleotides), or methyl groups (yielding methylphosphonate deoxynucleotides), a reduction in the AT content of AT rich regions, or replacement of non-preferred codon usage of the expression system to preferred codon usage of the expression system. The nucleic acid can be directly cloned into an appropriate vector, or if desired, can be modified to facilitate the subsequent cloning steps. Such modification steps are routine, an example of which is the addition of oligonucleotide linkers which contain restriction sites to the termini of the nucleic acid. General methods are set forth in in Sambrook et al. (2001) Molecular Cloning - A Laboratory Manual (3rd ed.) Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, (Sambrook).
Once the nucleic acid sequence is obtained, the sequence encoding the specific amino acids can be modified or changed at any particular amino acid position by techniques well known in the art. For example, PCR primers can be designed which span the amino acid position or positions and which can substitute any amino acid for another amino acid. Alternatively, one skilled in the art can introduce specific mutations at any point in a particular nucleic acid sequence through techniques for point mutagenesis. General methods are set forth in Smith, M. "In vitro mutagenesis" Ann. Rev. Gen., 19:423-462 (1985) and Zoller, M.J. "New molecular biology methods for protein engineering" Curr. Opin. Struct. Biol., 1 :605-610 (1991), which are incorporated herein in their entirety for the methods. These techniques can be used to alter the coding sequence without altering the amino acid sequence that is encoded.
The sequences contemplated herein include full-length wild-type (or native) sequences, as well as allelic variants, variants, fragments, homologs or fusion sequences that retain the ability to function as the cellular nucleic acid or protein involved in viral infection. In certain examples, a protein or nucleic acid sequence has at least 50% sequence identity, for example at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 98% sequence identity to a native sequences of the genes set forth in any of tables 1-4. In other examples, a nucleic acid sequence involved in viral infection has a sequence that hybridizes to a sequence of a gene set forth in Table 1, 2 3 or 4 and retains the activity of the sequence of the gene set forth in Table 1, 2 3, or 4. For example, and not to be limiting, a nucleic acid that hybridizes to an ABTBl nucleic acid sequence and encodes a protein that retains ABTBl activity is contemplated by the present invention. Such sequences include the genomic sequence for the genes set forth in Table 1, 2, 3 or 4. The examples set forth above for ABTBl are merely illustrative and should not be limited to ABTBl as the analysis set forth in this example applies to every nucleic acid and protein for the genes listed in Table 1 , 2, 3 or 4.
Unless otherwise specified, any reference to a nucleic acid molecule throughout this application includes the reverse complement of the nucleic acid. For example, any siRNA sequence set forth herein also includes the reverse complement of that sequence. Except where single-strandedness is required by the text herein (for example, a ssRNA molecule), any nucleic acid written to depict only a single strand encompasses both strands of a corresponding double- stranded nucleic acid. For example, depiction of a plus-strand of a dsDN A also encompasses the complementary minus-strand of that dsDNA. Additionally, any reference to the nucleic acid molecule that encodes a specific protein, or a fragment thereof, encompasses both the sense strand and its reverse complement. Fragments of the nucleic acids set forth in Table 1, 2, 3 or 4 are also contemplated. These fragments can be utilized as primers and probes to amplify, inhibit or detect any of the nucleic acids or genes set forth in Table 1 , 2, 3 or 4.
Hybridization conditions resulting in particular degrees of stringency will vary depending upon the nature of the hybridization method and the composition and length of the hybridizing nucleic acid sequences. Generally, the temperature of hybridization and the ionic strength (such as the Na+ concentration) of the hybridization buffer will determine the stringency of hybridization. Calculations regarding hybridization conditions for attaining particular degrees of stringency are discussed in Sambrook et al., (1989) Molecular Cloning, second edition, Cold Spring Harbor Laboratory, Plainview, NY (chapters 9 and 11). The following is an exemplary set of hybridization conditions and is not limiting:
Very High Stringency (detects sequences that share 90% identity) Hybridization: 5x SSC at 65°C for 16 hours Wash twice: 2x SSC at room temperature (RT) for 15 minutes each
Wash twice: 0.5x SSC at 65°C for 20 minutes each
High Stringency (detects sequences that share 80% identity or greater) Hybridization: 5x-6x SSC at 65°C-70°C for 16-20 hours Wash twice: 2x SSC at RT for 5-20 minutes each Wash twice: Ix SSC at 55°C-70°C for 30 minutes each
Low Stringency (detects sequences that share greater than 50% identity) Hybridization: 6x SSC at RT to 55°C for 16-20 hours Wash at least twice: 2x-3x SSC at RT to 55°C for 20-30 minutes each.
Also provided is a vector, comprising a nucleic acid set forth herein. The vector can direct the in vivo or in vitro synthesis of any of the proteins or polypeptides described herein. The vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid. These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene (See generally, Sambrook et al). The vector, for example, can be a plasmid. The vectors can contain genes conferring hygromycin resistance, ampicillin resistance, gentamicin resistance, neomycin resistance or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification.
There are numerous other E. coli (Escherichia coli) expression vectors, known to one of ordinary skill in the art, which are useful for the expression of the nucleic acid insert. Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species. In these prokaryotic hosts one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication). In addition, any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (Trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. Additionally, yeast expression can be used. The invention provides a nucleic acid encoding a polypeptide of the present invention, wherein the nucleic acid can be expressed by a yeast cell. More specifically, the nucleic acid can be expressed by Pichia past or is or S. cerevisiae.
Mammalian cells also permit the expression of proteins in an environment that favors important post-translational modifications such as folding and cysteine pairing, addition of complex carbohydrate structures, and secretion of active protein. Vectors useful for the expression of active proteins are known in the art and can contain genes conferring hygromycin resistance, genticin or G418 resistance, or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification. A number of suitable host cell lines capable of secreting intact human proteins have been developed in the art, and include the CHO cell lines, HeLa cells, COS-7 cells, myeloma cell lines, Jurkat cells, etc. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, etc.
The expression vectors described herein can also include nucleic acids of the present invention under the control of an inducible promoter such as the tetracycline inducible promoter or a glucocorticoid inducible promoter. The nucleic acids of the present invention can also be under the control of a tissue-specific promoter to promote expression of the nucleic acid in specific cells, tissues or organs. Any regulatable promoter, such as a metal lothionein promoter, a heat-shock promoter, and other regulatable promoters, of which many examples are well known in the art are also contemplated. Furthermore, a Cre-loxP inducible system can also be used, as well as a FIp recombinase inducible promoter system, both of which are known in the art. Insect cells also permit the expression of mammalian proteins. Recombinant proteins produced in insect cells with baculovirus vectors undergo post-translational modifications similar to that of wild-type proteins. The invention also provides for the vectors containing the contemplated nucleic acids in a host suitable for expressing the nucleic acids. The host cell can be a prokaryotic cell, including, for example, a bacterial cell. More particularly, the bacterial cell can be an E. coli cell. Alternatively, the cell can be a eukaryotic cell, including, for example, a Chinese hamster ovary (CHO) cell, a COS-7 cell, a HELA cell, an avian cell, a myeloma cell, a Pichia cell, or an insect cell. A number of other suitable host cell lines have been developed and include myeloma cell lines, fibroblast cell lines, a cell line suitable for infection by a pathogen, and a variety of tumor cell lines such as melanoma cell lines. The vectors containing the nucleic acid segments of interest can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transformation is commonly utilized for prokaryotic cells, whereas calcium phosphate, DEAE dextran, Lipofectamine, or lipofectin mediated transfection, electroporation or any method now known or identified in the future can be used for other eukaryotic cellular hosts. Polypeptides
The present invention provides isolated polypeptides comprising the polypeptide or protein sequences for the genes set forth in Table 1, 2, 3 or 4. The present invention also provides fragments of these polypeptides. These fragments can be of sufficient length to serve as antigenic peptides for the generation of antibodies. The present invention also contemplates functional fragments that possess at least one activity of a gene or gene product listed in Table 1, 2, 3 or 4, for example, involved in viral infection.
By "isolated polypeptide" or "purified polypeptide" is meant a polypeptide that is substantially free from the materials with which the polypeptide is normally associated in nature or in culture. The polypeptides of the invention can be obtained, for example, by extraction from a natural source if available (for example, a mammalian cell), by expression of a recombinant nucleic acid encoding the polypeptide (for example, in a cell or in a cell-free translation system), or by chemically synthesizing the polypeptide. In addition, a polypeptide can be obtained by cleaving full length polypeptides. When the polypeptide is a fragment of a larger naturally occurring polypeptide, the isolated polypeptide is shorter than and excludes the full-length, naturally-occurring polypeptide of which it is a fragment.
Also provided by the present invention is a polypeptide comprising an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the native polypeptide sequence for any gene set forth in Table 1, 2, 3 or 4.
It is understood that as discussed herein the use of the terms "homology" and "identity" mean the same thing as similarity. Thus, for example, if the use of the word homology is used to refer to two non-natural sequences, it is understood that this is not necessarily indicating an evolutionary relationship between these two sequences, but rather is looking at the similarity or relatedness between their nucleic acid sequences. Many of the methods for determining homology between two evolutionarily related molecules are routinely applied to any two or more nucleic acids or proteins for the purpose of measuring sequence similarity regardless of whether they are evolutionarily related.
In general, it is understood that one way to define any known variants and derivatives or those that might arise, of the disclosed nucleic acids and polypeptides herein, is through defining the variants and derivatives in terms of homology to specific known sequences. In general, variants of nucleic acids and polypeptides herein disclosed typically have at least, about 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent homology to the stated sequence or the native sequence. Those of skill in the art readily understand how to determine the homology of two polypeptides or nucleic acids. Methods of Decreasing Infection
The present invention provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 1, wherein the pathogen is not HIV. For example, and not to be limiting, the present invention provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of, BCR, CA2, CHRNA7, CXCR4, EDNRA, EGFR, F2, GRIA2, HDACl, KCNJl 1, MMP9, PDE8A, PLKl, POLE, SRD5A2, TUBA8 or DPP4, wherein the pathogen is not HIV. For example, imatinib can be used to inhibit BCR. Acetazolamide, benzothiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate or trichloromethiazide can be used to inhibit CA2. Amobarbitol, atracurium, cisatracurium, D-tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enflurane or isoflurane can be used to inhibit CHRNA7. Plerixafor can be used to inhibit CXCR4. Ambrisentan, avosentan, bosentan, BSF 302146, clazosentan, PD 180988, SB 234551, sitaxsentan, TBC 3214, ZD4054 or atrasentan can be utilized to inhibit EDNRA. AEE 788, ARRY-334543, BMS-599626, canertinib, cetuximab, erlotinib, gefitinib, HKI-272, lapatinib, PD 153035, vandetanib or XL647 can be used to inhibit EGFR. Antithrombin, alfa, argatroban, bivalirudin, dabigatran etexilate, desirudil, enoxaparin, or lepirudin can be utilized to inhibit F2. Talampanel or tezampanel can be utilized to inhibit
GRIA2. MGCD0103, PXDlOl, pyroxamide, romidepsin, tributyrin or vorinostat can be utilized to inhibit HDACl . Acetohexamide, chlorpropamide, glimepiride, glipizide, glyburide, minoxidil, nateglinide, phentolamine, repaglinide, tolazamide or tolbutamide can be utilized to inhibit KCNJl 1. Aminophylline, anagrelide, dipyridamole, dyphylline, milrinone, nitroglycerin, pentoxifylline, theophylline or tolbutamide can be utilized to inhibit PDE8A. BI2536 can be utilized to inhibit PLKl . Clofarabine, gemcitabine, nelarabine or trifluridine can be utilized to inhibit POLE. Dutasteride or finasteride can be utilized to inhibit SRD5A2. Docetaxel, ixabepilone, milataxel, NPI-2358, podophyllotoxin, TPI 287, AL 108, paclitaxel, TTI-237, XRP9881 (larotaxel), EC145, E7389, vinblastine, vincristine, vinflunine or vinorelbine can be utilized to inhibit TUBA8. Saxagliptin, sitagliptin, SYR-322 or talabostat can be utilized to inhibit DPP4. Minocycline can be utilized to inhibit MMP9. Combinations of two or more of the compounds set forth can be utilized to inhibit infection. These combinations can be utilized to inhibit one or more of the gene products selected from the group consisting of BCR, CA2, CHRNA7, CXCR4, EDNRA, EGFR, F2, GRI A2, HDACl, KCNJl 1, PDE8A, PLKl, POLE, SRD5A2, TUBA8 and DPP4. Therefore, two or more compounds that inhibit expression or activity of the same gene product can be used, or two or more compounds that inhibit expression or activity of two or more different gene products can be used to inhibit infection.
Also provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 2, wherein the pathogen is not West Nile Virus. Further provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of CACNAl A, PDE3B or TUBB4, wherein the pathogen is not West Nile Virus. Clevidipine butyrate can be utilized to inhibit CACNAlA. Aminophylline, amrinone, cilostazol, dipyridamole, dyphylline, medorinone, nitroglycerin, pentoxifylline, theophylline or tolbutamide can be utilized to inhibit PDE3B. ABT-751, BMS-275183, docetaxel, E7389, milataxel, MST-997, NPI-2358, podophyllotoxin, TPI 287, AL 108, ixabepilone, paclitaxel, TTI-237, XRP9881, epothilone B, EC 145, vinblastine, vincristine, vinflunine, minocycline or vinorelbinecan be utilized to inhibit TUBB4.
Further provided is a method of inhibiting infection in a cell by a pathogen, comprising decreasing expression or activity of a gene or gene product set forth in Table 3, wherein the pathogen is not West Nile Virus or Dengue Fever Virus. Further provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of ADRAlD, wherein the pathogen is not West Nile Virus or Dengue Fever Virus. Carvedilol, dapiprazole, ergotamine, guanadrel, Labetalol, phenoxybenzamine, prazosin, terazosine, alfuzosin, aripiprazole, asenapine, dihydroergotamine, DL 017, epinastine, guanethidine, iloperidone, nefazodone, olanzapine, paliperidone, phentolamine, quetiapine, quinidine, risperidone, tolazoline, UK-294315, ziprasidone, dobutamine, apraclonidine, arbutamine, clonidine, tamsulosin or venlafaxine can be utilized to inhibit ADRAlD.
The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of a gene or gene product set forth in Table 4, wherein the pathogen is not hepatitis C virus. The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or STAT (SOATl) or TBXA2R, wherein the pathogen is not hepatitis C virus. Pactimibe can be utilized to inhibit STAT (SOATl). S-18886 or torsemide can be utilized to inhibit TBXA2R. Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, wherein the pathogen is not HIV. Further provided is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is not West Nile Virus. Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is not West Nile Virus or Dengue Fever Virus. Also provided by the present invention is a method of decreasing infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, wherein the pathogen is not HCV. As utilized herein, one or more includes, two or more, three or more, four or more, five or more, six or more, seven or more, etc.
As stated above, an infection can be a viral infection, bacterial infection, fungal infection or a parasitic infection, to name a few. A decrease or inhibition of infection can occur in a cell, in vitro, ex vivo or in vivo. As utilized throughout, the term "infection" encompasses all phases of pathogenic life cycles including, but not limited to, attachment to cellular receptors, entry, internalization, disassembly, replication, genomic integration of pathogenic sequences, transcription of viral RNA, translation of viral RNA, transcription of host cell mRNA, translation of host cell mRNA, proteolytic cleavage of pathogenic proteins or cellular proteins, assembly of particles, endocytosis, cell lysis, budding, and egress of the pathogen from the cells. Therefore, a decrease in infection can be a decrease in attachment to cellular receptors, a decrease in entry, a decrease in internalization, a decrease in disassembly, a decrease in replication, a decrease in genomic integration of pathogenic sequences, a decrease in translation of mRNA, a decrease in proteolytic cleavage of pathogenic proteins or cellular proteins, a decrease in assembly of particles, a decrease in endocytosis, a decrease in cell lysis, a decrease in budding, or a decrease in egress of the pathogen from the cells. This decrease does not have to be complete as this can range from a slight decrease to complete ablation of the infection. A decrease in infection can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of infection in a cell wherein expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4 has not been decreased. In the methods set forth herein, expression can be inhibited, for example by inhibiting transcription of the gene, or inhibiting translation of its gene product. Similarly, the activity of a gene product (for example, an mRNA, a polypeptide or a protein) can be inhibited, either directly or indirectly. Inhibition or a decrease in expression does not have to be complete as this can range from a slight decrease in expression to complete ablation of expression. For example, expression can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression of a gene or gene product set forth in Table 1 , 2, 3 or 4 has not been decreased or inhibited. Similarly, inhibition or decrease in the activity of a gene product does not have to be complete as this can range from a slight decrease to complete ablation of the activity of the gene product. For example, the activity of a gene product can be inhibited by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein activity of a gene or gene product set forth in Table 1, 2, 3 or 4 has not been decreased or inhibited. As utilized herein, "activity of a gene product" can be an activity that is involved in pathogenicity, for example, interacting directly or indirectly, with pathogen, e.g. viral protein or viral nucleic acids, or an activity that the gene product performs in a normal cell, i.e. in a non-infected cell. Depending on the gene product, one of skill in the art would know how to assay for an activity that is involved in pathogenicity, an activity that is involved in normal cellular function, or both. As set forth above, an activity of the proteins and nucleic acids listed herein can be the ability to bind or interact with other proteins. Therefore, the present invention also provides a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and other cellular proteins, such as, for example, transcription factors, receptors, nuclear proteins, transporters, microtubules, membrane proteins, enzymes (for example, ATPases, phosphorylases, oxidoreductases, kinases, phosphatases, synthases, lyases, aromatases, helicases, hydrolases, proteases, transferases, nucleases, ligases, reductases and polymerases) and hormones, provided that such inhibition correlates with decreasing infection by the pathogen. Also provided is a method of decreasing infection by inhibiting or decreasing the interaction between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).
The cells of the present invention can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli. The cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. Therefore, the cell can also be part of a population of cells. The cell(s) can also be in a subject.
Examples of viral infections include but are not limited to, infections caused by RNA viruses (including negative stranded RNA viruses, positive stranded RNA viruses, double stranded RNA viruses and retroviruses), DNA viruses. All strains and types of RNA viruses and DNA viruses are contemplated herein.
Examples of RNA viruses include, but are not limited to picornaviruses, which include aphthoviruses (for example, foot and mouth disease virus O, A, C, Asia 1, SATl, SAT2 and SAT3), cardioviruses (for example, encephalomycarditis virus and Theiller's murine encephalomyelitis virus), enteroviruses (for example polioviruses 1 , 2 and 3, human enteroviruses A-D, bovine enteroviruses 1 and 2, human coxsackieviruses A1-A22 and A24, human coxsackieviruses B1-B5, human echoviruses 1-7, 9, 11-12, 24, 27, 29-33, human enteroviruses 68-71 , porcine enteroviruses 8-10 and simian enteroviruses 1-18), erboviruses (for example, equine rhinitis virus), hepatovirus (for example human hepatitis A virus and simian hepatitis A virus), kobuviruses (for example, bovine kobuvirus and Aichi virus), parechoviruses (for example, human parechovirus 1 and human parechovirus 2), rhinovirus (for example, rhinovirus A, rhinovirus B, HRV16 (VR-11757), HRV14 (VR-284), or HRV1 A (VR-1559), human rhinovirus 1-100 and bovine rhinoviruses 1-3) and teschoviruses (for example, porcine teschovirus).
Additional examples of RNA viruses include caliciviruses, which include noroviruses (for example, Norwalk virus), sapoviruses (for example, Sapporo virus), lagoviruses (for example, rabbit hemorrhagic disease virus and European brown hare syndrome) and vesiviruses (for example vesicular exanthema of swine virus and feline calicivirus). Other RNA viruses include astroviruses, which include mamastorviruses and avastroviruses. Togaviruses are also RNA viruses. Togaviruses include alphaviruses (for example, Sindbis virus, Chikungunya virus, Semliki Forest virus, Western equine encephalitis, Getah virus, Everglades virus, Venezuelan equine encephalitis virus and Aura virus) and rubella viruses. Additional examples of RNA viruses include the the flaviviruses (for example, tick- borne encephalitis virus, Tyuleniy virus, Aroa virus, Dengue virus (types 1 to 4), Kedougou virus, Japanese encephalitis virus (JEV), West Nile virus (WNV), Kokobera virus, Ntaya virus, Spondweni virus, Yellow fever virus, Entebbe bat virus, Modoc virus, Rio Bravo virus, Cell fusing agent virus, pestivirus, GB virus A, GBV-A like viruses, GB virus C, Hepatitis G virus, hepacivirus (hepatitis C virus (HCV)) all six genotypes), bovine viral diarrhea virus, and GB virus B).
Other examples of RNA viruses are the coronaviruses which include, human respiratory coronaviruses such as SARS-CoV, HCoV-229E, HCoV-NL63 and HCoV-OC43. Coronaviruses also include bat SARS-like CoV, turkey coronavirus, chicken coronavirus, feline coronavirus and canine coronavirus. Additional RNA viruses include arteriviruses (for example, equine arterivirus, porcine reproductive and respiratory syndrome virus, lactate dehyrogenase elevating virus of mice and simian hemorraghic fever virus). Other RNA viruses include the rhabdoviruses, which include lyssaviruses (for example, rabies, Lagos bat virus, Mokola virus, Duvenhage virus and European bat lyssavirus), vesiculoviruses (for example, VSV-Indiana, VSV-New Jersey, VSV-Alagoas, Piry virus, Cocal virus, Maraba virus, Isfahan virus and Chandipura virus), and ephemeroviruses (for example, bovine ephemeral fever virus, Adelaide River virus and Berrimah virus). Additional example of RNA viruses include the filoviruses. These include the Marburg and Ebola viruses (for example, EBOV-Z, EBOV-S, EBOV-IC and EBOV-R.
The paramyxoviruses are also RNA viruses. Examples of these viruses are the rubula viruses (for example, mumps, parainfluenza virus 5, human parainfluenza virus type 2, Mapuera virus and porcine rubulavirus), avulaviruses (for example, Newcastle disease virus), respoviruses (for example, Sendai virus, human parainfluenza virus type 1 and type 3, bovine parainfluenza virus type 3), henipaviruses (for example, Hendra virus and Nipah virus), morbilloviruses (for example, measles, Cetacean morvilliirus, Canine distemper virus, Peste- des-petits-ruminants virus, Phocine distemper virus and Rinderpest virus), pneumoviruses (for example, human respiratory syncytial virus A2, Bl and S2, bovine respiratory syncytial virus and pneumonia virus of mice), metapneumoviruses (for example, human metapneumovirus and avian metapneumovirus). Additional paramyxoviruses include Fer-de-Lance virus, Tupaia paramyxovirus, Menangle virus, Tioman virus, Beilong virus, J virus, Mossman virus, Salem virus and Nariva virus.
Additional RNA viruses include the orthomyxoviruses. These viruses include influenza viruses and strains (e.g., influenza A (HlNl (including but not limited to A/WS/33 or A/California/04/2009 strains) H2N2, H3N2, H5N1 , H7N7, H1N2, H9N2, H7N2, H7N3 and H10N7), influenza A strain A/Victoria/3/75, influenza A strain A/Puerto Rico/8/34, influenza B strain Lee, influenza B (for example B strain Lee) and C viruses, as well as avian influenza (for example, strains H5N1, H5N2, H7N1, H7N7 and H9N2) thogotoviruses and isaviruses. Orthobunyaviruses (for example, Akabane virus, California encephalitis, Cache Valley virus, Snowshoe hare virus,) nairoviruses (for example, Nairobi sheep virus, Crimean-Congo hemorrhagic fever virus Group and Hughes virus), phleboviruses (for example, Candiru, Punta Toro, Rift Valley Fever, Sandfly Fever, Naples, Toscana, Sicilian and Chagres), and hantaviruses (for example, Hantaan, Dobrava, Seoul, Puumala, Sin Nombre, Bayou, Black Creek Canal, Andes and Thottapalayam) are also RNA viruses. Arenaviruses such as lymphocytic choriomeningitis virus, Lassa fever virus, Argentine hemorrhagic fever virus, Bolivian hemorrhagic fever virus, Venezuelan hemorrhagic fever virus, SABV and WWAV are also RNA viruses. Borna disease virus is also an RNA virus. Hepatitis D (Delta) virus and hepatitis E are also RNA viruses. Additional RNA viruses include reoviruses, rotaviruses, birnaviruses, chrysoviruses, cystoviruses, hypoviruses, partitiviruses and totoviruses. Orbiviruses such as African horse sickness virus, Blue tongue virus, Changuinola virus, Chenuda virus, Chobar Gorge Corriparta virus, epizootic hemorraghic disease virus, equine encephalosis virus, Eubenangee virus, Ieri virus, Great Island virus, Lebombo virus, Orungo virus, Palyam virus, Peruvian Horse Sickness virus, St. Croix River virus, Umatilla virus, Wad Medani virus, Wallal virus, Warrego virus and Wongorr virus are also RNA viruses. Retroviruses include alpharetroviruses (for example, Rous sarcoma virus and avian leukemia virus), betaretroviruses (for example, mouse mammary tumor virus, Mason-Pfizer monkey virus and Jaagsiekte sheep retrovirus), gammaretroviruses (for example, murine leukemia virus and feline leukemia virus, deltraretroviruses (for example, human T cell leukemia viruses (HTLV- 1 , HTL V-2), bovine leukemia virus, STLV- 1 and STLV-2), epsilonretriviruses (for example, Walleye dermal sarcoma virus and Walleye epidermal hyperplasia virus 1), reticuloendotheliosis virus (for example, chicken syncytial virus, lentiviruses (for example, human immunodeficiency virus (HIV) type 1, human immunodeficiency virus (HIV) type 2, simian immunodeficiency virus, equine infectious anemia virus, feline immunodeficiency virus, caprine arthritis encephalitis virus and Visna maedi virus) and spumaviruses (for example, human foamy virus and feline syncytia-forming virus).
Examples of DNA viruses include polyomaviruses (for example, simian virus 40, simian agent 12, BK virus, JC virus, Merkel Cell polyoma virus, bovine polyoma virus and lymphotrophic papovavirus), papillomaviruses (for example, human papillomavirus, bovine papillomavirus, adenoviruses (for example, adenoviruses A-F, canine adenovirus type I, canined adeovirus type 2), circoviruses (for example, porcine circovirus and beak and feather disease virus (BFDV)), parvoviruses (for example, canine parvovirus), ery thro viruses (for example, adeno-associated virus types 1-8), betaparvoviruses, amdoviruses, densoviruses, iteraviruses, brevidensoviruses, pefudensoviruses, herpes viruses (for example, herpes simplex virus 1 , herpes simplex virus 2, varicella- zoster virus, Epstein-Barr virus, cytomegalovirus, Kaposi's sarcoma associated herpes virus, human herpes virus-6 variant A, human herpes virus-6 variant B and cercophithecine herpes virus 1 (B virus)), poxviruses (for example, smallpox (variola), cowpox, monkeypox, vaccinia, Uasin Gishu, camelpox, psuedocowpox, pigeonpox, horsepox, fowlpox, turkeypox and swinepox), and hepadnaviruses (for example, hepatitis B and hepatitis B-like viruses). Chimeric viruses comprising portions of more than one viral genome are also contemplated herein.
For animals, in addition to the animal viruses listed above, viruses include, but are not limited to, the animal counterpart to any above listed human virus. The provided genes can also decrease infection by newly discovered or emerging viruses. Such viruses are continuously updated on http://en.wikipedia.org/wiki/Virus and www.virology.net.
Examples of bacterial infections include, but are not limited to infections caused by the following bacteria: Listeria (sp.), Franscicella tularemis, Mycobacterium tuberculosis, Rickettsia (all types), Ehrlichia, Chlamydia. Further examples of bacteria that can be targeted by the present methods include M. tuberculosis, M. bovis, M. bovis strain BCG, BCG substrains, M. avium, M. intracellular, M. africanum, M. kansasii, M. marinum, M. ulcerans, M. avium subspecies paratuberculosis, Nocardia asteroides, other Nocardia species, Legionella pneumophila, other Legionella species, Salmonella typhi, other Salmonella species, Shigella species, Yersinia pestis, Pasteurella haemolytica, Pasteurella multocida, other Pasteurella species, Actinobacillus pleuropneumoniae, Listeria monocytogenes, Listeria ivanovii, Brucella abortus, other Brucella species, Cowdria ruminantium, Chlamydia pneumoniae, Chlamydia trachomatis, Chlamydia psittaci, Coxiella burnetti, other Rickettsial species, Ehrlichia species, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, Streptococcus agalactiae, Bacillus anthracis, Escherichia coli, Vibrio cholerae, Kingella kingae,
Campylobacter species, Neiserria meningitidis, Neiserria gonorrhea, Pseudomonas aeruginosa, other Pseudomonas species, Haemophilus influenzae, Haemophilus ducreyi, other Hemophilus species, Clostridium tetani, other Clostridium species, Yersinia enterolitica, and other Yersinia species. Examples of parasitic infections include, but are not limited to infections caused by the following parasites: Cryptosporidium, Plasmodium (all species), American trypanosomes (T. cruzi), African trypanosomes, Acanthamoeba, Entaoeba histolytica, Angiostrongylus, Anisakis, Ascaris, Babesia, Balantidium, Baylisascaris, lice, ticks, mites, fleas, Capillaria, Clonorchis, Chilomastix mesnili, Cyclspora, Diphyllobothrium, Dipylidium caninum, Fasciola, Giardia, Gnathostoma, Hetetophyes, Hymenolepsis, Isospora, Loa loa, Microsporidia, Naegleria, Toxocara, Onchocerca, Opisthorchis, Paragonimus, Baylisascaris, Strongyloides, Taenia, Trichomonas and Trichuris.
Furthermore, examples of protozoan and fungal species contemplated within the present methods include, but are not limited to, Plasmodium falciparum, other Plasmodium species, Toxoplasma gondii, Pneumocystis carinii, Trypanosoma cruzi, other trypanosomal species, Leishmania donovani, other Leishmania species, Theileria annulata, other Theileria species, Eimeria tenella, other Eimeria species, Histoplasma capsulatum, Cryptococcus neoformans, Blastomyces dermatitidis, Coccidioides immitis, Paracoccidioides brasiliensis, Penicillium marnejfei, and Candida species. The provided genes can also decrease infection by newly discovered or emerging bacteria, parasites or fungi, including multidrug resistant strains of same.
Further provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the pathogen is a respiratory virus. Respiratory viruses include, but are not limited to, picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses. More specifically, and not to be limiting, the respiratory virus can be an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus or a respiratory syncytial virus (RSV).
Also provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one ore more gene(s) or gene product(s) set forth in Table 1 , wherein the pathogen is a gastrointestinal virus. Gastrointestinal viruses include, but are not limited to, picornavi ruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, an adenovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus.
Also provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is a gastrointestinal virus that is not West Nile virus. Gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses. More specifically, and not to be limiting, with respect to the genes listed in Table 2, the gastrointestinal virus can be a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, or a Dengue fever virus.
Further provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is a gastrointestinal virus that is not West Nile virus or Dengue fever virus. Gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, caliciviruses, adenoviruses and reoviruses. More specifically, and not to be limiting, with respect to the genes listed in Table 3, the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, or an enterovirus.
Also provided by the present invention is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, wherein the pathogen is a gastrointestinal virus. With respect to the genes listed in Table 4, gastrointestinal viruses include, but are not limited to, picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a West Nile virus or a Dengue fever virus.
The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , wherein the pathogen is a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses,
Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, a hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human
Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies or Chikungunya virus.
The present invention also provides a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus. Also provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2 or 4, wherein the pathogen is a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.
Further provided is a method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, wherein the pathogen is a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus, Ebola virus or Marburg virus. Further provided is a method of decreasing the toxicity of a toxin in a cell comprising administering to the cell an effective amount of a composition that decreases expression or activity of one or more gene(s) or a gene product(s) set forth in Table 1, 2, 3 or 4.
Toxins can include, but are not limited to, a bacterial toxin, a neurotoxin, such as a botulinum neurotoxin, a mycotoxin, ricin, a Clostridium perfringens toxin, a Clostridium difficile toxin, a saxitoxin, a tetrodotoxin, abrin, a conotoxin, a Staphylococcal toxin, such as staphylococcal aureus, an E. coli toxin, a streptococcal toxin, a shigatoxin, a T-2 toxin, an anthrax toxin, chimeric forms of the toxins listed herein, and the like. The decrease in toxicity can be at least about 10%, 20%, 30%, 40%, 50%, 60, 70%, 80%, 90%, 95%, 100% or any other percentage decrease in between these percentages as compared to the level of toxicity in a cell wherein expression or activity of the same gene or gene product from Table 1, 2, 3 or 4 that has not been decreased, or compared to the level of toxicity in a cell not contacted with a compound that decreases expression or activity of the gene or gene product.
Toxicity can be measured, for example, via a cell viability, apopotosis assay, LDH release assay or cytotoxicity assay (See, for example, Kehl-Fie and St. Geme "Identification and characterization of an RTX toxin in the emerging pathogen Kingella kingae," J. Bacteriol. 189(2):430-6 (2006) and Kirby "Anthrax Lethal Toxin Induces Human Endothelial cell Apoptosis," Infection and Immunity 72: 430-439 (2004), both of which are incorporated herein in their entireties by this reference.)
In the methods of the present invention, expression and/or activity of a gene or gene product set forth in Table 1, 2, 3 or 4, can be decreased by contacting the cell with any composition that can decrease expression or activity. For example, the composition can be a chemical, a small or large molecule (organic or inorganic), a drug, a protein, a peptide, a cDNA, an antibody, an aptamer, a morpholino, a triple helix molecule, an siRNA, an LNA, a shRNA, an miRNA, an antisense RNA, a ribozyme or any other compound now known or identified in the future that decreases the expression and/or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4. A decrease in expression or activity can occur by decreasing transcription of mRNA or decreasing translation of RNA. A composition can also be a mixture or "cocktail" of two or more of the compositions described herein. A decrease in expression and/or activity can also occur by inhibiting the interaction between any of the proteins set forth in Table 1 , 2, 3 or 4 and other cellular proteins, such as, for example, transcription factors, receptors, nuclear proteins, transporters, microtubules, membrane proteins, enzymes (for example, ATPases, phosphorylases, oxidoreductases, kinases, phosphatases, synthases, lyases, aromatases, helicases, hydrolases, proteases, transferases, nucleases, ligases, reductases and polymerases) and hormones. A decrease in expression and/or activity can also occur by inhibiting or decreasing the interaction between any of the proteins of the present invention and a cellular nucleic acid or a viral nucleic acid. A decrease can also occur by inhibiting or decreasing the interaction, either direct or indirect, between any of the proteins of the present invention and a viral, bacterial, parasitic or fungal protein (i.e. a non-host protein).
A composition can also be single composition or a mixture, cocktail or combination of two or more compositions, for example, two or more compositions selected from the group consisting of chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, an aptamer, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an LNA, an antisense nucleic acid or a ribozyme. The two or more compositions can be the same or different types of compositions. The two or more compositions can decrease expression or activity of the same target or different targets, as one or more genes or gene products set forth in Table 1,2, 3 or 4 can be modulated to effect a decrease in infection. It is understood that two or more compositions comprises three or more, four or more, five or more etc. For example, and not to be limiting two or more compositions can be any two or more compositions comprising an antisense and a small molecule; or two or more antisense molecules; or two or more small molecules; or two or more compositions comprising an siRNA and a small molecule, etc. It is understood that any combination of the types of compositions set forth herein can be utilized in the methods set forth herein.
These compositions can be used alone or in combination with other therapeutic agents such as antiviral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti- inflammatory agents, anti-cancer agents, etc. All of the compounds described herein can be contacted with a cell in vitro, ex vivo or in vivo.
Examples of antiviral compounds include, but are not limited to, amantadine, rimantadine, zanamavir and oseltamavir (Tamiflu) for the treatment of flu and its associated symptoms. Antiviral compounds useful in the treatment of HIV include Combivir® (lamivudine-zidovudine), maraviroc, Crixivan® (indinavir), Emtriva® (emtricitabine), Epivir® (lamivudine), Fortovase® (saquinavir-sg), Hivid® (zalcitabine), Invirase® (saquinavir-hg), Kaletra® (lopinavir-ritonavir), LexivaTM (fosamprenavir), Norvir® (ritonavir), Retrovir® (zidovudine) Sustiva® (efavirenz), Videx EC® (didanosine), Videx® (didanosine), Viracept® (nelfϊnavir) Viramune® (nevirapine), Zerit® (stavudine), Ziagen® (abacavir), Fuzeon® (enfuvirtide) Rescriptor® (delavirdine), Reyataz® (atazanavir), Trizivir® (abacavir-lamivudine- zidovudine) Viread® (tenofovir disoproxil fumarate) and Agenerase® (amprenavir). Other antiviral compounds useful in the treatment of Ebola and other filoviruses include ribavirin and cyanovirin-N (CV-N). For the treatment of herpes virus, Zovirax®(acyclovir) is available. Antibacterial agents include, but are not limited to, antibiotics (for example, penicillin and ampicillin), sulfa Drugs and folic acid Analogs, Beta-Lactams, aminoglycosides, tetracyclines, macrolides, lincosamides, streptogramins, fluoroquinolones, rifampin, mupirocin, cycloserine, aminocyclitol and oxazolidinones.
Antifungal agents include, but are not limited to, amphotericin, nystatin, terbinafine, itraconazole, fluconazole, ketoconazole, and griselfulvin. Antiparasitic agents include, but are not limited to, antihelmintics, antinematodal agents, antiplatyhelmintic agents, antiprotozoal agents, amebicides, antimalarials, antitrichomonal agents, aoccidiostats and trypanocidal agents.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression and/or activity of a gene product set forth in Table 1,2 3, or 4 and a composition that decreases expression and/or activity of a different gene product(s) set forth in Table 1, 2, 3 or 4. For example, and not to be limiting, the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression and/or activity of EDNRA and a composition that decreases expression and/or activity of KCNJl 1. This is merely exemplary, as any two or more different gene product(s) set forth in Table 1, 2, 3 or 4 can be inhibited to decrease infection. It is understood that these methods are not limited to administration of two compositions as the combination can be a combination of two, three, four, five or six compositions, wherein each composition comprises a chemical, a compound, a small molecule, an inorganic molecule, an organic molecule, a drug (for example, an FDA approved drug, a European Medicines Agency approved drug, a drug approved by the Japanese Pharmaceutical and Medical Device Agency) a protein, a cDNA, an aptamer, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an LNA, an miRNA, an antisense nucleic acid or a ribozyme. As set forth herein, these combinations can comprise two or more compounds selected from the group consisting of imatinib, acetazolamide, benzthiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate, trichloromethiazide, amobarbital, atracurium, cisatracurium, D- tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enflurane, isoflurane, plerixafor, ambrisentan, avosentan, bosentan, BSF 302146, clazosentan, PD 180988, SB
234551, sitaxsentan, TBC 3214, ZD4054, atrasentan, AEE 788, ARRY-334543, BMS-599626, canertinib, cetuximab, erlotinib, gefitinib, HKI-272, lapatinib, PD 153035, vandetanib, XL647, antithrombin alfa, argatroban, bivalirudin, dabigatran etexilate, desirudil, enoxaparin, lepirudin, lonafarnib, talampanel, tezampanel, MGCD0103, PXDlOl, pyroxamide, romidepsin, tributyrin, vorinostat, acetohexamide, chlorpropamide, glimepiride, glipizide, glyburide, minoxidil, nateglinide, phentolamine, repaglinide, tolazamide, tolbutamide, aminophylline, anagrelide, dipyridamole, dyphylline, milrinone, nitroglycerin, pentoxifylline, theophylline, tolbutamide, BI 2536, clofarabine, gemcitabine, nelarabine, trifluridine, dutasteride, finasteride, docetaxel, ixabepilone, milataxel, NPI-2358, podophyllotoxin, TPI 287, AL 108, paclitaxel, TTI-237, XRP9881 (larotaxel), EC145, E7389, vinblastine, vincristine, vinflunine, vinorelbine, enzastaurin, saxagliptin, sitagliptin, SYR-322, talabostat, mycophenolic acid, thioguanine, VX- 944, mycophenolic acid, ribavirin, thioguanine, VX-944, pactimibe, S-18886, torsemide, carvedilol, dapiprazole, ergotamine, guanadrel, Labetalol, phenoxybenzamine, prazosin, terazosine, alfuzosin, aripiprazole, asenapine, dihydroergotamine, DL 017, epinastine, guanethidine, iloperidone, nefazodone, olanzapine, paliperidone, phentolamine, quetiapine, quinidine, risperidone, tolazoline, UK-294315, ziprasidone, dobutamine, apraclonidine, arbutamine, clonidine, tamsulosin, venlafaxine, clevidipine butyrate, aminophylline, amrinone, cilostazol, dipyridamole, dyphylline, medorinone, nitroglycerin, pentoxifylline, theophylline, tolbutamide, ABT-751, BMS-275183, docetaxel, E7389, milataxel, MST-997, NPI-2358, podophyllotoxin, TPI 287, AL 108, ixabepilone, paclitaxel, TTI-237, XRP9881, epothilone B, EC 145, vinblastine, vincristine, vinflunine, minocycline and vinorelbine, or a pharmaceutically acceptable salt thereof. These combinations can further comprise an antiviral compound, an antiparasitic compound or an antifungal compound as described herein.
Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens. The two or more pathogens can be two or more, three or more, four or more, five or more, six or more; or seven or more viruses, bacteria, parasites, fungi or a combination thereof. In particular, the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses. Table 6 shows gene products set forth in Table 1, 2, 3 or 4 for which at least 50% inhibition of viral replication was observed via siRNA experiments as indicated by an X. It is understood that one of skill in the art can select a gene product from Table 6 and decrease its expression or activity to reduce infection by one or more viruses marked with an X, for that gene product. One of skill in the art can readily select the gene product(s) in Table 6 that can be inhibited in order to effect inhibition of infection by two or more, three or more, four or more, five or more, six or more, seven or more viruses etc.
The viruses are not restricted to the viruses set forth in Table 6 as this data can be combined with data for a composition that decreases expression and/or activity of any gene product from Table 1, 2, 3 or 4 and inhibits any other virus, bacteria, fungi or parasite not listed in Table 6. For example, data for EDNRA can be combined with data showing that decreased expression and/or activity of EDNRA leads to inhibition of HCV infection, thus showing that activity or expression of EDNRA can be decreased to inhibit infection by RSV, pox, rhinovirus, Dengue fever virus, HIV and HCV. This example is merely exemplary as data for any gene product set forth in Table 6 can be combined with data for any other virus, bacteria, fungi or parasite.
Therefore, the present invention provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses, wherein at least two or more of the viruses are selected from the group consisting of influenza, RSV, HSV, cowpox, rhinovirus, HIV and Dengue. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of a gene or a gene product set forth in Table 1,2, 3 or 4, wherein the composition inhibits infection by two or more, three or more, four or more, five or more, six or more; or seven or more viruses, wherein at least two or more of the viruses are selected from the group consisting of influenza, RSV, HSV, cowpox, rhinovirus, Dengue, HIV and HCV. TABLE 6
Figure imgf000046_0001
VO
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
ON
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000051_0002
Antibodies
The present invention also provides antibodies that specifically bind to the gene products, proteins and fragments thereof set forth in Table 1, 2, 3 or 4. The antibody of the present invention can be a polyclonal antibody or a monoclonal antibody. The antibody of the invention selectively binds a polypeptide. By "selectively binds" or "specifically binds" is meant an antibody binding reaction which is determinative of the presence of the antigen (in the present case, a polypeptide set forth in Table 1 , 2,3 or 4, or antigenic fragment thereof among a heterogeneous population of proteins and other biologies). Thus, under designated immunoassay conditions, the specified antibodies bind preferentially to a particular peptide and do not bind in a significant amount to other proteins in the sample. Preferably, selective binding includes binding at about or above 1.5 times assay background and the absence of significant binding is less than 1.5 times assay background.
This invention also contemplates antibodies that compete for binding to natural interactors or ligands to the proteins set forth in Table 1, 2, 3 or 4. In other words, the present invention provides antibodies that disrupt interactions between the proteins set forth in Table 1, 2, 3 or 4 and their binding partners. For example, an antibody of the present invention can compete with a protein for a binding site (e.g. a receptor) on a cell or the antibody can compete with a protein for binding to another protein or biological molecule, such as a nucleic acid that is under the transcriptional control of a transcription factor set forth in Table 1 , 2, 3 or 4. An antibody can also disrupt the interaction between a protein set forth in Table 1 , 2, 3 or 4 and a pathogen, or the product of a pathogen. For example, an antibody can disrupt the interaction between a protein set forth in Table 1 , 2, 3 or 4 and a viral protein, a bacterial protein, a parasitic protein, a fungal protein or a toxin. The antibody optionally can have either an antagonistic or agonistic function as compared to the antigen. Antibodies that antagonize pathogenic infection are utilized to decrease infection. Similarly antibodies that antagonize toxicity can be utilized to decrease toxicity in a cell or subject.
Preferably, the antibody binds a polypeptide in vitro, ex vivo or in vivo. Optionally, the antibody of the invention is labeled with a detectable moiety. For example, the detectable moiety can be selected from the group consisting of a fluorescent moiety, an enzyme-linked moiety, a biotin moiety and a radiolabeled moiety. The antibody can be used in techniques or procedures such as diagnostics, screening, or imaging. Anti-idiotypic antibodies and affinity matured antibodies are also considered to be part of the invention.
As used herein, the term "antibody" encompasses chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab')2, Fab', Fab and the like, including hybrid fragments. Thus, fragments of the antibodies that retain the ability to bind their specific antigens are provided. Such antibodies and fragments can be made by techniques known in the art and can be screened for specificity and activity according to the methods set forth in the Examples and in general methods for producing antibodies and screening antibodies for specificity and activity (See Harlow and Lane. Antibodies, A Laboratory Manual. Cold Spring Harbor Publications, New York, (1988)).
Also included within the meaning of "antibody" are conjugates of antibody fragments and antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference. Optionally, the antibodies are generated in other species and "humanized" for administration in humans. In one embodiment of the invention, the "humanized" antibody is a human version of the antibody produced by a germ line mutant animal. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2, or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In one embodiment, the present invention provides a humanized version of an antibody, comprising at least one, two, three, four, or up to all CDRs of a monoclonal antibody that specifically binds to a protein or fragment thereof set forth in Table 1, 2, 3 or 4. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of or at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non- human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., Nature, 321 :522-525 (1986); Riechmann et al., Nature, 332:323- 327 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)).
Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321 :522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534- 1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
Peptides
Peptides that inhibit expression or activity of a gene or a gene product set forth in Table 1 , 2, 3 or 4 are also provided herein. Peptide libraries can be screened utilizing the screening methods set forth herein to identify peptides that inhibit activity of any of the genes or gene products set forth in Table 1 , 2, 3 or 4. These peptides can be derived from a protein that binds to any of the genes or gene products set forth in Table 1, 2, 3 or 4. These peptides can be any peptide in a purified or non-purified form, such as peptides made of D-and/or L-configuration amino acids (in, for example, the form of random peptide libraries; see Lam et al., Nature 354:82-4, 1991), phosphopeptides (such as in the form of random or partially degenerate, directed phosphopeptide libraries; see, for example, Songyang et al, Cell 72:767-78, 1993).
RNA interference
Numerous RNA interference technologies are known to those of skill in the art, for example, but not limited to, siRNA, antisense, locked nucleic acids (LNA), morpholinos, and shRNA. One of skill in the art can utilize any RNA interference technology, known or identified in the future to decrease expression of any gene product set forth in Table 1, 2, 3 or 4. siRNAs
Short interfering RNAs (siRNAs), also known as small interfering RNAs, are double- stranded RNAs that can induce sequence-specific post-transcriptional gene silencing, thereby decreasing gene expression (See, for example, U.S. Patent Nos. 6,506,559, 7,056,704, 7,078, 196, 6, 107,094, 5,898,221 , 6,573,099, and European Patent No. 1.144,623, all of which are hereby incorporated in their entireties by this reference). siRNas can be of various lengths as long as they maintain their function. In some examples, siRNA molecules are about 19-23 nucleotides in length, such as at least 21 nucleotides, for example at least 23 nucleotides. In one example, siRNA triggers the specific degradation of homologous RNA molecules, such as mRNAs, within the region of sequence identity between both the siRNA and the target RNA. For example, WO 02/44321 discloses siRNAs capable of sequence-specific degradation of target mRNAs when base-paired with 3' overhanging ends. The direction of dsRNA processing determines whether a sense or an antisense target RNA can be cleaved by the produced siRNA endonuclease complex. Thus, siRNAs can be used to modulate transcription or translation, for example, by decreasing expression of a gene set forth in Table 1, 2, 3 or 4. The effects of siRNAs have been demonstrated in cells from a variety of organisms, including Drosophila, C. elegans, insects, frogs, plants, fungi, mice and humans (for example, WO 02/44321 ; Gitlin et at., Nature 418:430-4, 2002; Caplen et ai, Proc. Natl. Acad. ScL 98:9742-9747, 2001 ; and Elbashir et al., Nature 41 1 :494-8, 2001). Utilizing sequence analysis tools, one of skill in the art can design siRNAs to specifically target one or more of the genes set forth in Table 1 , 2, 3 or 4 for decreased gene expression. siRNAs that inhibit or silence gene expression can be obtained from numerous commercial entities that synthesize siRNAs, for example, Ambion Inc. (2130 Woodward Austin, TX 78744- 1832, USA), Qiagen Inc. (27220 Turnberry Lane, Valencia, CA USA) and Dharmacon Inc. (650 Crescent Drive, #100 Lafayette, CO 80026, USA). The siRNAs synthesized by Ambion Inc., Qiagen Inc. or Dharmacon Inc, can be readily obtained from these and other entities by providing a GenBank Accession No. for the mRNA of any gene set forth in Table 1, 2, 3 or 4. In addition, siRNAs can be generated by utilizing Invitrogen's BLOCK-IT™ RNAi Designer https://rnaidesigner.invitrogen.com/rnaiexpress. siRNA sequences can comprise a 3 'TT overhang and/or additional sequences that allow efficient cloning and expression of the siRNA sequences. siRNA sequences can be cloned into vectors and utilized in vitro, ex vivo or in vivo to decrease gene expression. One of skill in the art would know that it is routine to utilize publicly available algorithms for the design of siRNA to target mRNA sequences. These sequences can then be assayed for inhibition of gene expression in vitro, ex vivo or in vivo. shRNA shRNA (short hairpin RNA) is a DNA molecule that can be cloned into expression vectors to express siRNA (typically 19-29 nt RNA duplex) for RNAi interference studies. shRNA has the following structural features: a short nucleotide sequence ranging from about 19- 29 nucleotides derived from the target gene, followed by a short spacer of about 4-15 nucleotides (i.e. loop) and about a 19-29 nucleotide sequence that is the reverse complement of the initial target sequence. Antisense Nucleic Acids
Generally, the term "antisense" refers to a nucleic acid molecule capable of hybridizing to a portion of an RNA sequence (such as mRNA) by virtue of some sequence complementarity. The antisense nucleic acids disclosed herein can be oligonucleotides that are double-stranded or single-stranded, RNA or DNA or a modification or derivative thereof, which can be directly administered to a cell (for example by administering the antisense molecule to the subject), or which can be produced intracellularly by transcription of exogenous, introduced sequences (for example by administering to the subject a vector that includes the antisense molecule under control of a promoter).
Antisense nucleic acids are polynucleotides, for example nucleic acid molecules that are at least 6 nucleotides in length, at least 10 nucleotides, at least 15 nucleotides, at least 20 nucleotides, at least 100 nucleotides, at least 200 nucleotides, such as 6 to 100 nucleotides. However, antisense molecules can be much longer. In particular examples, the nucleotide is modified at one or more base moiety, sugar moiety, or phosphate backbone (or combinations thereof), and can include other appending groups such as peptides, or agents facilitating transport across the cell membrane (Letsinger et al, Proc. Natl. Acad. Sci. USA 1989, 86:6553- 6; Lemaitre et al., Proc. Natl. Acad. ScL USA 1987, 84:648-52; WO 88/09810) or blood-brain barrier (WO 89/10134), hybridization triggered cleavage agents (Krol et al , BioTechniques 1988, 6:958-76) or intercalating agents (Zon, Pharm. Res. 5:539-49, 1988). Additional modifications include those set forth in U.S. Patent Nos. 7,176,296; 7,329,648; 7,262,489, 7,115,579; and 7,105,495.
Examples of modified base moieties include, but are not limited to: 5-fluorouracil, 5- bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N~6- sopentenyladenine, 1 -methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, methoxyarninomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'- methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5- oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4- thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-S-oxyacetic acid, 5- methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, and 2,6-diaminopurine.
Examples of modified sugar moieties include, but are not limited to: arabinose, 2- fluoroarabinose, xylose, and hexose, or a modified component of the phosphate backbone, such as phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, or a formacetal or analog thereof.
In a particular example, an antisense molecule is an α-anomeric oligonucleotide. An α- anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gautier et al., Nucl. Acids Res. 15:6625-41, 1987). The oligonucleotide can be conjugated to another molecule, such as a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization- triggered cleavage agent. Oligonucleotides can include a targeting moiety that enhances uptake of the molecule by host cells. The targeting moiety can be a specific binding molecule, such as an antibody or fragment thereof that recognizes a molecule present on the surface of the host cell.
In a specific example, antisense molecules that recognize a nucleic acid set forth herein, include a catalytic RNA or a ribozyme (for example see WO 90/11364; WO 95/06764; and Sarver et al, Science 247:1222-5, 1990). Conjugates of antisense with a metal complex, such as terpyridylCu (II), capable of mediating mRNA hydrolysis, are described in Bashkin et al. {Appl. Biochem Biotechnol 54:43-56, 1995). In one example, the antisense nucleotide is a 2'-O- methylribonucleotide (Inoue et al., Nucl. Acids Res. 15:6131-48, 1987), or a chimeric RNA- DNA analogue (Inoue et al, FEBS Lett. 215:327-30, 1987). Antisense molecules can be generated by utilizing the Antisense Design algorithm of Integrated DNA Technologies, Inc. (1710 Commercial Park, Coralville, IA 52241 USA; http://www.idtdna.com/Scitools/ Applications/ AntiSense/ Antisense. aspx.
Any antisense sequence that is not the full length mRNA for any of the genes listed in Table 1, 2, 3 or 4 can be used as antisense sequences. It is known to those of skill in the art that once a mRNA sequence is routinely obtained for any of the genes set forth in Table 1, 2, 3 or 4, it is routine to walk along the mRNA sequence to generate antisense sequences that decrease expression of the gene. Therefore all antisense sequences for the genes set forth herein are disclosed. The methods of the present invention can utilize any antisense sequence that decreases the expression of a gene set forth in Table 1, 2, 3 or 4.
Morpholinos
Morpholinos are synthetic antisense oligos that can block access of other molecules to small (about 25 base) regions of ribonucleic acid (RNA). Morpholinos are often used to determine gene function using reverse genetics methods by blocking access to mRNA. Morpholinos, usually about 25 bases in length, bind to complementary sequences of RNA by standard nucleic acid base-pairing. Morpholinos do not degrade their target RNA molecules. Instead, Morpholinos act by "steric hindrance", binding to a target sequence 'within an RNA and simply interfering with molecules which might otherwise interact with the RNA. Morpholinos have been used in mammals, ranging from mice to humans. Bound to the 5 '-untranslated region of messenger RNA (mRNA), Morpholinos can interfere with progression of the ribosomal initiation complex from the 5' cap to the start codon. This prevents translation of the coding region of the targeted transcript (called "knocking down" gene expression). Morpholinos can also interfere with pre-mRNA processing steps, usually by preventing the splice-directing snRNP complexes from binding to their targets at the borders of introns on a strand of pre-RNA. Preventing Ul (at the donor site) or U2/U5 (at the polypyrimidine moiety & acceptor site) from binding can cause modified splicing, commonly leading to exclusions of exons from the mature mRNA. Targeting some splice targets results in intron inclusions, while activation of cryptic splice sites can lead to partial inclusions or exclusions. Targets of Ul 1/U12 snRNPs can also be blocked. Splice modification can be conveniently assayed by reverse-transcriptase polymerase chain reaction (RT-PCR) and is seen as a band shift after gel electrophoresis of RT-PCR products. Methods of designing, making and utilizing morpholinos are disclosed in U.S. Patent No. 6,867,349 which is incorporated herein by reference in its entirety.
Small Molecules
Any small molecule that inhibits activity of a gene or a gene product set forth in Table
1 ,2, 3 or 4 can be utilized in the methods of the present invention to decrease infection. These molecules are available in the scientific literature, in the StarLite/CHEMBL database available from the European Bioinformatics Institute, in DrugBank (Wishart et al. Nucleic Acids Res. 2006 Jan 1 ;34 (Database issue):D668-72), package inserts, brochures, chemical suppliers (for example, Sigma, Tocris, Aurora Fine Chemicals, to name a few), or by any other means, such that one of skill in the art makes the association between a gene product of Table 1, 2 3, or 4 and inhibition of this gene product by a molecule. Preferred small molecules are those small molecules that have IC50 values of less than about ImM, less than about 500 micromolar, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar, less than about 1 micromolar, less than about 0.5 micromolar, less than about 0.25 micromolar, or less than about .10 micromolar. The half maximal inhibitory concentration (IC50) is a measure of the effectiveness of a compound in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular compound or other substance (inhibitor) is needed to inhibit a given biological process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. In other words, it is the half maximal (50%) inhibitory concentration (IC) of a substance (50% IC, or IC50). It is commonly used as a measure of antagonist drug potency in pharmacological research. Sometimes, it is also converted to the pIC50 scale (-log IC50), in which higher values indicate exponentially greater potency. According to the FDA, IC50 represents the concentration of a drug that is required for 50% inhibition in vitro. It is comparable to an EC50 for agonist drugs. EC50 also represents the plasma concentration required for obtaining 50% of a maximum effect in vivo. The present invention also provides the synthesis of small molecules that inhibit activity of a gene product set forth in Table 1, 2, 3 or 4. The present invention describes gene products for which three-dimensional structures are well known and can be obtained from the RCSB Protein Databank http://www.rcsb.org/pdb/home/home.do or http://www.rcsb.org.
Crystal structures can also be generated. Alternatively, one of skill in the art can obtain crystal structures for proteins, or domains of proteins, that are homologous to the proteins set forth in Table 1 , 2,3 or 4 from the RCSB Protein Databank or elsewhere in the scientific literature for use in homology modeling studies.
Routine high throughput in silico or in vitro screening of compound libraries for the identification of small molecules is also provided by the present invention. Compound libraries are commercially available. With an available crystal structure, it is routine for one of skill in the art to screen a library in silico and identify compounds with desirable properties, for example, binding affinity.
Numerous computer programs are available and suitable for rational drug design and the processes of computer modeling, model building, and computationally identifying, selecting and evaluating potential compounds. These include, for example, SYBYL (available from TRIPOS, St. Louis Mo.), DOCK (available from University of California, San Francisco), GRID (available form Oxford University, UK), MCSS (available from Molecular Simulations Inc., Burlington, Mass.), AUTODOCK (available from Oxford Molecular Group), FLEX X (available from TRIPOS, St. Louis Mo.), CAVEAT (available from University of California, Berkeley), HOOK (available from Molecular Simulations Inc., Burlington, Mass.), and 3-D database systems such as MACCS-3D (available from MDL Information Systems, San Leandro, Calif.), UNITY (available from TRIPOS, St. Louis Mo.), and CATALYST (available from Molecular Simulations Inc., Burlington, Mass.). Compounds can also be computationally modified using such software packages as LUDI (available from Biosym TechMA), and LEAPFROG (TRIPOS Associates, St. Louis, Mo.). These computer-modeling techniques can be performed on any suitable hardware including for example, workstations available from Silicon Graphics, Sun Microsystems, and the like. These techniques, methods, hardware and software packages are representative and are not intended to be comprehensive listing. Other modeling techniques known in the art can also be employed in accordance with this invention. A filter can be applied to the results to yield one or more compounds with a binding affinity in a particular range, for example, and not to be limiting, from about 100 micromolar to about 100 nanomolar, from about 10 micromolar to about 10 nanomolar, from about 1 micromolar to about 1 nanomolar, or from about 0.5 micromolar to about 0.5 nanomolar. Another filter can provide compounds with a certain binding affinity and size, for example, less than 1000 daltons, less than 500 daltons, less than 400 daltons, less than 300 daltons, less than 200 daltons, less than 100 daltons or less than 50 daltons or any size in between. The ranges and properties can be modified depending on the protein being studied. The compounds identified via this screening method can be further studied in silico, in vitro or in vivo. For example, the compounds can be modified in silico and rescreened in silico to determine the effects of chemical modifications on binding affinity or other properties being assessed in silico. The compounds identified in silico can be synthesized for in vitro or in vivo analysis.
All of the screening leading up to in vivo testing can be done in silico or in combination with in vitro assays. The initial compounds identified in silico and the resulting modified compounds can be screened in vitro, for example, in cellular assays to determine the effect of the compound on the cellular host protein as well as in viral assays, to determine antiviral activity. IC50 values can be obtained from the cellular assays, which may or may not be similar to the concentration necessary to effect 50% inhibition of viral infection in a viral assay. However, although not required, it is desirable to have a compound that has an IC50 value of less than about ImM, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar , less than about 1 micromolar, less than about 0.5 micromolar or less than about 0.1 micromolar. Similarly, although not required, it is desirable to have a compound that effects 50% inhibition of viral infection at a concentration of less than about ImM, less than about 100 micromolar, less than about 75 micromolar, less than about 50 micromolar, less than about 25 micromolar, less than about 10 micromolar, less than about 5 micromolar, less than about 1 micromolar, less than about 0.5 micromolar, less than about 0.1 micromolar or any concentration in between.
Further modifications of the compounds can be done after in vitro screening, either in silico or via chemical synthesis, for further evaluation, prior to additional in vitro screening or in vivo studies. It is understood that this process can be iterative, involving a combination of in silico and wet chemistry techniques, but routine in drug development.
Other filters can be applied to the in silico screening process, for example, a filter that takes ADMET (adsorption, distribution, metabolism, excretion, toxicity) properties into consideration can be applied. ADMET modeling can be used during compound optimization to define an acceptable property space that contains compounds likely to have the desired properties. These filters can be applied sequentially or simultaneously depending.
Libraries for virtual or in vitro screening are available for the skilled artisan, for example from ChemBridge Corporation (San Diego, CA), such as a GPCR library, a kinase targeted library (KINACore), or an ion channel library (Ion Channel Set), to name a few. Compound libraries can also be obtained from the National Institutes of Health. For example, the NIH Clinical Collection of compounds that have been used in clinical trials can also be screened. Biofocus DPI (Essex, United Kingdom) also maintains and designs compound libraries that can be purchased for screening. One of skill in the art can select a library based on the protein of interest. For example, a kinase library can be screened to identify a compound that binds to and/or modulates a kinase. Other libraries that target enzyme families, for example, ATPases, hydrolases, isomerases, polymerases, transferases, phosphatases, etc., can also be screened, depending on the type of enzyme.
Compound libraries can also be screened in order to identify a compound that disrupts or inhibits specific interactions between two proteins. Compounds can be administered to cells comprising two or more proteins that interact with each other in order to identify compounds that disrupt this interaction and result in decreased interaction between of the two or more proteins. Co-immunoprecipiation experiments can be utilized. Similarly, FRET analysis or surface plasmon resonance can be utilized, to identify compounds that disrupt the interaction between a two proteins.
Transcription factors set forth herein are available to identify inhibitors in screening assays that are known to the skilled person. Gel shift assays can be employed to assess the activity of a transcription factor (see, for example, Wang et al. 2000 The Journal of Biological Chemistry, 275, 27013-27020). One of skill in the art can also utilize the techniques set forth in Wang et al., as they apply to the transcription factors set forth herein to assess transcription factor activity via binding of the transcription factor to a regulatory region that drives expression of a reporter protein. These cellular assays and other transcription factor assays standard in the art are applicable to any transcription factor described herein. Compounds can be introduced into these assays to assess the effect of the compound on transcriptional activity. For example, a cellular assay can be utilized to measure reporter activity in the presence of a transcription factor, in the presence or absence of a compound, to determine the effect of the compound on transcription. If reporter activity is reduced, the compound is an inhibitor of transcription by that particular transcription factor. Utilizing these assays can also identify compounds that inhibit the activity of for example, any zinc finger protein set forth in this application. Interactions between membrane proteins and their ligands, can also be measured, for example, via plasmon- waveguide resonance (PWR) spectroscopy (see, for example, Hruby and Tollin, Curr. Opin. Pharmacol. 2007 October: 7(5): 507-514; and Salamon et al. Methods Enzymol. 2009; 461 : 123-46) or plasmon surface resonance (Alves et al. Curr Protein Pept ScL 2005 August; 6(4): 293-312.) These techniques can be utilized to identify compounds that alter the interaction between a membrane protein and a ligand by quantitating changes in binding affinity, protein aggregation and other parameters detectable by these methods.
Additional inhibitors include compositions comprising carbon and hydrogen, and optionally comprising one or more of -S, -N, -O, -Cl, -Br, or -Fl, appropriately bonded as a structure, with a size of less than about 1000 daltons, less than about 500 daltons, less than about 300 daltons, less than about 200 daltons, or less than about 100 daltons, that fits into a binding pocket or an active site of a gene product set forth herein. In particular, inhibitors that have the properties described in Lipinsky's Rule of Five are included herein. Lipinski's rule of five states that a drug/inhibitor has a weight under 500 Daltons, a limited lipophilicity or octanol-water partition coefficient (expressed by Log P < 5, with P = [drug]org./[drug]aq.), a maximum of 5 H-bond donors (expressed as the sum of OHs and NHs), and a maximum of 10 H-bond acceptors (expressed as the sum of oxygen and nitrogen atoms). Inhibitors that violate no more than one of the above-listed five rules are also included herein.
The following compounds are provided as inhibitors of pathogenic infection and/or toxicity. More specifically, the present invention provides a method of decreasing infection by a pathogen, in a cell or a subject, said method comprising administering to the cell or subject an effective amount of one or more compounds selecting from the group of compounds having the structure of imatinib, acetazolamide, benzthiazide, brinzolamide, chlorothiazide, chlorthalidone, dorzolamide, dorzolamide/timolol, hydrochlorothiazide, methazolamide, sulfacetamide, topiramate, trichloromethiazide, amobarbital, atracurium, cisatracurium, D-tubocurarine, doxacurium, mecamylamine, metocurine, mivacurium, pancuronium, pipecuronium, rapacuronium, rocuronium, succinylcholine, vecuronium, enfiurane, isoflurane, plerixafor, ambrisentan, avosentan, bosentan, BSF 302146, clazosentan, PD 180988, SB 234551, sitaxsentan, TBC 3214, ZD4054, atrasentan, AEE 788, ARRY-334543, BMS-599626, canertinib, cetuximab, erlotinib, gefitinib, HKI-272, lapatinib, PD 153035, vandetanib, XL647, antithrombin alfa, argatroban, bivalirudin, dabigatran etexilate, desirudil, enoxaparin, lepirudin, lonafarnib, talampanel, tezampanel, MGCD0103, PXDlOl, pyroxamide, romidepsin, tributyrin, vorinostat, acetohexamide, chlorpropamide, glimepiride, glipizide, glyburide, minoxidil, nateglinide, phentolamine, repaglinide, tolazamide, tolbutamide, aminophylline, anagrelide, dipyridamole, dyphylline, milrinone, nitroglycerin, 2-chloro-N-(3,5-dichlorophenyl)-4- (trifluoromethyl)pyrimidine-5-carboxamide, pentoxifylline, theophylline, tolbutamide, BI 2536, clofarabine, gemcitabine, nelarabine, trifluridine, dutasteride, finasteride, docetaxel, ixabepilone, milataxel, NPI-2358, podophyllotoxin, TPI 287, AL 108, paclitaxel, TTI-237, XRP9881 (larotaxel), EC 145, E7389, vinblastine, vincristine, vinflunine, vinorelbine, enzastaurin, saxagliptin, sitagliptin, SYR-322, talabostat, mycophenolic acid, thioguanine, VX-944, mycophenolic acid, ribavirin, thioguanine, VX-944, pactimibe, S- 18886, torsemide, carvedilol, dapiprazole, ergotamine, guanadrel, Labetalol, phenoxybenzamine, prazosin, terazosine, alfuzosin, aripiprazole, asenapine, dihydroergotamine, DL 017, epinastine, guanethidine, iloperidone, nefazodone, olanzapine, paliperidone, phentolamine, quetiapine, quinidine, risperidone, tolazoline, UK-294315, ziprasidone, dobutamine, apraclonidine, arbutamine, clonidine, tamsulosin, venlafaxine, clevidipine butyrate, aminophylline, amrinone, cilostazol, dipyridamole, dyphylline, medorinone, nitroglycerin, pentoxifylline, theophylline, tolbutamide, ABT-751, BMS-275183, docetaxel, E7389, milataxel, MST-997, NPI-2358, podophyllotoxin, TPI 287, AL 108, ixabepilone, paclitaxel, TTI-237, XRP9881, epothilone B, EC145, vinblastine, vincristine, vinflunine, minocycline, vinorelbine and (5-(6-amino-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methyl dihydrogen phosphate, or a pharmaceutically acceptable salt thereof.
As utilized herein, "pharmaceutically acceptable salts" are those salts derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, formic, benzoic, malonic, naphthalene-2-sulfonic and benzenesulfonic acids. Salts derived from appropriate bases include alkali metal (e.g. sodium), alkaline earth metal (e.g. magnesium) and ammonium salts. Preferred salts include hydrochlorides, hydrobromides, sulfates, mesylates, maleates, and fumarates. Throughout this application, references to a compound, according to the invention, includes the compounds as well as its pharmaceutically acceptable salts.
As utilized herein, one or more includes, two or more, three or more, four or more, five or more, six or more, etc. Therefore, combinations of the compounds set forth herein are provided by the present invention. The available structures for these compounds are set forth below. As set forth throughout the application, the infection can be a viral, bacterial, fungal or parasitic infection. The pathogen can be a respiratory virus. Respiratory viruses include, but are not limited to, picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses. More specifically, and not to be limiting, the respiratory virus can be an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus or a respiratory syncytial virus (RSV) or any strain thereof. The pathogen can also be a gastrointestinal virus. Gastrointestinal viruses include, but are not limited to, picornaviruses, adenoviruses, filoviruses, flaviviruses, caliciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus. The pathogen can also be a hemorraghic fever virus. These include, but are not limited to, flaviviruses, bunyaviruses, arenaviruses, filoviruses and hantaviruses. More specifically and not to be limiting, the hemorraghic fever virus can be an Ebola virus, a Marburg virus, a Dengue fever virus (types 1-4), a yellow fever virus, a Sin Nombre virus, a Junin virus, Crimean Congo hemorraghic fever virus, a Machupo virus, a Lassa virus, a Rift Valley fever virus, or a Kyasanur forest disease virus. The pathogen can also be a pathogen selected from the group consisting of: a pox virus, a herpes virus, an RSV virus, HIV, an influenza virus, a hepatitis C virus, a hepatitis B virus, a hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. As mentioned above, the compounds can also be utilized to decrease toxicity of a toxin set forth herein.
TUBB4 and TUBA8A vinorelbine larotaxel paclitaxel
Figure imgf000065_0001
vinflunine
Figure imgf000065_0002
ixabepilone
Figure imgf000065_0003
vinblastine vincristine
Figure imgf000065_0004
Figure imgf000065_0005
Figure imgf000066_0001
Figure imgf000066_0002
O
Figure imgf000066_0003
PDE3B nitroglycerin Dvpyridamole Aminophylline tolbutamide
Figure imgf000067_0001
theophylline
Figure imgf000067_0002
Figure imgf000067_0003
CACNAlC clonidine Tolazoline
Figure imgf000068_0001
risperidone
Figure imgf000068_0002
venlafaxine
Figure imgf000068_0003
ziprasidone quinidine
Figure imgf000068_0004
Figure imgf000068_0006
Figure imgf000068_0007
tamsulosin apraclodinine UK-294315
Figure imgf000068_0005
Figure imgf000068_0008
epinastine aripiprazole phentolamine nefazodone
Figure imgf000069_0001
alfuzosin
Figure imgf000069_0003
terazosine
Figure imgf000069_0002
asenapine guanethidine
Figure imgf000069_0005
Figure imgf000069_0004
Figure imgf000069_0006
H
Figure imgf000070_0001
PLKl BI2536
Figure imgf000071_0001
finasteride OH clofarabine
Figure imgf000071_0002
Figure imgf000071_0003
PDE8A Tolazamide Nateglinide Glipizide anagrelide
.•C«I
Figure imgf000072_0001
acetohexamide romidepsin MGCDO 103 FNTA
Figure imgf000073_0001
CI
Cl
Figure imgf000074_0001
ZD4054 clazosentan avosentan CHRNA7 isoflurane
sita
Figure imgf000075_0002
xsentan ambrisentan
Figure imgf000075_0001
vecuronium
Figure imgf000075_0004
Figure imgf000075_0003
succinylcholine pipecuronium d-tubocurarine
Figure imgf000076_0001
Cisatracurium CA2 methazolamide timolol
Trichlormethiazide
Figure imgf000077_0001
hydrochlorothiazide
Figure imgf000077_0002
chlorthalidone
topiratnate atracurium
Figure imgf000077_0003
Figure imgf000077_0004
dorzolamide chlorothiazide
Figure imgf000077_0005
sulfacetamide amobarbital
Figure imgf000077_0006
Figure imgf000077_0007
Figure imgf000077_0008
Brinzolamide
BCR
Imatinib
Figure imgf000078_0001
benzthiazide
Figure imgf000078_0003
Figure imgf000078_0002
acetazolamide
Figure imgf000078_0004
Other methods of decreasing expression and/or activity include methods of interrupting or altering transcription of mRNA molecules by site-directed mutagenesis (including mutations caused by a transposon or an insertional vector). Chemical mutagenesis can also be performed in which a cell is contacted with a chemical (for example ENU) that mutagenizes nucleic acids by introducing mutations into a gene set forth in Table 1 , 2, 3 or 4. Transcription of mRNA molecules can also be decreased by modulating a transcription factor that regulates expression of any of the genes set forth in Table 1 , 2, 3 or 4. Radiation can also be utilized to effect mutagenesis.
Pharmaceutical Compositions and Modes of Administration
The present invention provides_a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, in the subject, wherein the pathogen is not HIV.
Also provided is a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table
2, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 2, in the subject, wherein the pathogen is not West Nile Virus.
Further provided is a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table
3, said method comprising administering to the subject an effective amount of a that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 3, in the subject, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
The present invention also provides a method of decreasing infection by a pathogen in a subject by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, said method comprising administering to the subject an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 4, in the subject, wherein the pathogen is not hepatitis C.
Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by three or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more respiratory viruses. Also provided is a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by five or more respiratory viruses. These can be selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus and an adenovirus. Since picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses and adenoviruses are families of viruses, two or more, three or more, four or more, or five or more respiratory viruses can be from the same or from different families. For example, and not to be limiting, the composition can inhibit infection by two or more orthomyxoviruses; two or more picornaviruses; an orthomyxovirus, an adenovirus, and a picornavirus; an orthomyxovirus, a paramyxovirus and an adenovirus; an orthomyxovirus, two picornaviruses and a paramyxovirus; three orthomyxoviruses, a picornavirus and an adenovirus, etc. More particularly, the composition can inhibit infection by two or more, three or more or four or more respiratory viruses selected from the group consisting of an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus and an RSV virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene produces) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by three or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more gastrointestinal viruses. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by five or more gastrointestinal viruses. These viruses can be selected from the group consisting of: a filovirus, an adenovirus, a picornavirus, a calcivirus, a flavivirus or a reovirus. Since filoviruses, picornaviruses, calciviruses, flaviviruses and reoviruses are families of viruses, the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses from the same or from different families. More particularly, the composition can inhibit infection by two or more, three or more, four or more, or five or more gastrointestinal viruses selected from the group consisting of a reovirus, a Norwalk virus, an adenovirus, an Ebola virus, a Marburg virus, a Dengue fever virus, a West Nile virus, a yellow fever virus, a rotavirus and an enterovirus. It is noted that in the methods of the present invention, if the composition decreases expression or activity of a gene or gene product set forth in Table 3, the combination of viruses inhibited by the composition is not West Nile Virus and Dengue fever virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by one or more pathogens selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, and inhibits infection by one or more pathogens selected from the group consisting of: a flavivirus, a filovirus, an adenovirus, a calicivirus or a reovirus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s)set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more pathogens selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by three or more pathogens. The three or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by four or more pathogens. The three or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the four or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by five or more pathogens. The five or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the five or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by six or more pathogens. The six or more pathogens can be selected from the viruses, bacteria, parasites and fungi set forth herein. More particularly, the five or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. The present invention also provides a method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits co-infection by HIV and one or more viruses, bacteria, parasites or fungi. For example, decreasing co-infection of HIV and any of the viruses, including for example any families, genus, species, or group of viruses. As a further example, co-infection of HIV and a respiratory virus is provided herein. Respiratory viruses include picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, and adenoviruses. More specifically, the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV. Also provided is decreasing co-infection of HIV and a gastrointestinal virus. Gastrointestinal viruses include picornaviruses, filoviruses, flaviviruses, calciviruses and reoviruses. More specifically, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus. Further provided is a method of decreasing co-infection of HIV with a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus. More particularly, decreasing co-infection of HIV and a hepatitis virus, such as Hepatitis A, Hepatitis B or Hepatitis C is provided. It is noted that in a method of decreasing co-infection of HIV and Hepatitis C, the gene or gene product is not ERCCl, THAPl 1, COASY, NC0A6 or MAP3K14. Also provided is decreasing co- infection of HIV and a herpes virus, for example, HSV-I or HSV-2. In addition decreasing co- infection of HIV and tuberculosis is also provided. Further provided is decreasing co-infection of HIV and CMV, as well as decreasing co-infection of HIV and HPV.
As described herein, the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of two or more respiratory viruses. Therefore, the present invention provides methods of treating or preventing an unspecified respiratory infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more respiratory viruses. More particularly, the present invention provides a method of decreasing an unspecified respiratory infection in a subject comprising: a) diagnosing a subject with an unspecified respiratory infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more respiratory viruses selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses. As set forth above, in the methods of the present invention, the two or more respiratory viruses can be from the same family or from a different family of respiratory viruses. More specifically, the respiratory virus can be any strain of influenza, rhinovirus, adenovirus, parainfluenza virus or RSV. In this method, the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more respiratory viruses selected from the group consisting of a picornaviruses, an orthomyxoviruses, paramyxoviruses, coronaviruses, or adenoviruses.
As described herein, the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of two or more gastrointestinal viruses. Therefore, the present invention provides methods of treating or preventing an unspecified gastrointestinal infection in a subject by administering a composition that decreases activity or expression of a gene involved in the pathogenesis of two or more gastrointestinal viruses. More particularly, the present invention provides a method of decreasing an unspecified gastrointestinal infection in a subject comprising: a) diagnosing a subject with an unspecified gastrointestinal infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more gastrointestinal viruses selected from the group consisting of a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus. It is noted that if the gene or gene product is from Table 3, the two gastrointestinal viruses are not West Nile virus and Dengue fever virus. As set forth above, in the methods of the present invention, the two or more gastrointestinal viruses can be from the same family or from a different family of gastrointestinal viruses. More particularly, and not to be limiting, the gastrointestinal virus can be any strain of reovirus, adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, a rotavirus, an enterovirus, a Dengue fever virus, a yellow fever virus, or a West Nile virus. In this method, the composition can be a composition that inhibits infection by three or more, four or more, five or more; or six or more gastrointestinal viruses selected from the group consisting of a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus.
The present invention also provides a method of preventing or decreasing an unspecified pandemic or bioterror threat in a subject comprising: a) diagnosing a subject with an unspecified pandemic or bioterrorist inflicted infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more, three or more, four or more; or five or more viruses selected from the group consisting of a pox virus, an influenza virus, West Nile virus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus and a Dengue fever virus. It is noted that the present invention also provides a method wherein the composition administered is not a composition that decreases expression or activity of a gene set forth in Table 3 for West Nile virus and Dengue Fever virus. Also provided by the present invention is a method of managing secondary infections in a patient comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition can inhibit infection by HIV and one or more, two or more, three or more, four or more; or five or more secondary infections. As set forth above, the genes set forth in Tables 1, 2, 3 or 4 can be involved in the pathogenesis of three or more pathogens. Therefore, the present invention provides methods of treating or preventing an unspecified infection by administering a composition that decreases the activity or expression of a gene that is involved in the pathogenesis of three or more pathogens. Therefore, the present invention provides a method of decreasing infection in a subject comprising: a) diagnosing a subject with an unspecified infection and; b) administering a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition decreases infection by three or more pathogens. More specifically, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, CaI ici viruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
The infection can be a viral infection, a parasitic infection, a bacterial infection or a fungal infection, to name a few. As utilized herein, "an unspecified infection" is an infection that presents symptoms associated with an infection, but is not identified as specific infection. One of skill in the art, for example, a physician, a nurse, a physician's assistant, a medic or any other health practitioner would know how to diagnose the symptoms of infection even though the actual pathogen may not be known. For example, the patient can present with one or more symptoms, including, but not limited to, a fever, fatigue, lesions, weight loss, inflammation, a rash, pain (for example, muscle ache, headache, ear ache, joint pain, etc.), urinary difficulties, respiratory symptoms (for example, coughing, bronchitis, lung failure, breathing difficulties, bronchiolitis, airway obstruction, wheezing, runny nose, sinusitis, congestion, etc.), gastrointestinal symptoms (for example, nausea, diarrhea, vomiting, dehydration, abdominal pain, intestinal cramps, rectal bleeding, etc.). This can occur in the event of a bioterrorist attack or a pandemic. In this event, one of skill in the art would know to administer a composition that inhibits infection by decreasing the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 that is involved in the pathogenesis of several pathogens. Similarly, if there is a threat of an unspecified infection, for example, a threat of a bioterrorist attack, a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4 can be administered prophylactically to a subject to prevent an unspecified infection in a subject. By "treat," "treating," or "treatment" is meant a method of reducing the effects of an existing infection. Treatment can also refer to a method of reducing the disease or condition itself rather than just the symptoms. The treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease or the symptoms of the disease. Treatment can range from a positive change in a symptom or symptoms of viral infection to complete amelioration of the viral infection as detected by art-known techniques. For example, a disclosed method is considered to be a treatment if there is about a 10% reduction in one or more symptoms of the disease in a subject with the disease when compared to native levels in the same subject or control subjects. Thus, the reduction can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
The methods of the present invention can also result in a decrease in the amount of time that it normally takes to see improvement in a subject. For example, a decrease in infection can be a decrease of hours, a day, two days, three days, four days, five days, six days, seven days, eight days, nine days, ten days, eleven days, twelve days, thirteen days, fourteen days, fifteen days or any time in between that it takes to see improvement in the symptoms, viral load or any other parameter utilized to measure improvement in a subject. For example, if it normally takes 7 days to see improvement in a subject not taking the composition, and after administration of the composition, improvement is seen at 6 days, the composition is effective in decreasing infection. This example is not meant to be limiting as one of skill in the art would know that the time for improvement will vary depending on the infection.
As utilized herein, by "prevent," "preventing," or "prevention" is meant a method of precluding, delaying, averting, obviating, forestalling, stopping, or hindering the onset, incidence, severity, or recurrence of infection. For example, the disclosed method is considered to be a prevention if there is about a 10% reduction in onset, incidence, severity, or recurrence of infection, or symptoms of infection (e.g., inflammation, fever, lesions, weight loss, etc.) in a subject exposed to an infection when compared to control subjects exposed to an infection that did not receive a composition for decreasing infection. Thus, the reduction in onset, incidence, severity, or recurrence of infection can be about a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to control subjects. For example, and not to be limiting, if about 10% of the subjects in a population do not become infected as compared to subjects that did not receive preventive treatment, this is considered prevention.
Also provided is a method of decreasing infection in a subject comprising: a) administering a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4, in a subject with an unspecified infection; b) diagnosing the type of infection in the subject and; c) administering a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 for the diagnosed infection. Also provided is a method wherein the diagnosed infection is HIV and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 1. Further provided is a method wherein the diagnosed infection is West Nile Virus and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 2. Further provided is a method wherein the diagnosed infection is West Nile Virus or Dengue fever virus and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 3. Further provided is a method wherein the diagnosed infection is Hepatitis C and the composition is not a composition that decreases the expression or activity of a gene or gene product set forth in Table 4.
Further provided is a method of treating viral infection comprising: a) diagnosing a subject with a viral infection; and b) removing a drug from the subject that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4, if the viral infection is not a viral infection that is inhibited by a composition that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4. As mentioned above, upon recognizing that a subject has an infection or the symptoms of an infection, for example, in the case of a bioterrorist attack or a pandemic, given that a gene or gene product set forth in Table 1, 2, 3 or 4 can be involved in the pathogenesis of several pathogens, a practitioner can prescribe or administer a composition that decreases the expression or activity of the gene or gene product. After administration, the practitioner, who can be the same practitioner or a different practitioner, can diagnose the type of infection in a subject. This diagnosis can be a differential diagnosis where the practitioner distinguishes between infections by comparing signs or symptoms and eliminates certain types of infection before arriving at the diagnosis for a specific infection, or a diagnosis based on a test that is specific for a particular infection. Once a specific infection is diagnosed, if the gene or gene product is involved in the pathogenesis of this infection, the practitioner can prescribe or administer a composition that decreases the expression or activity of that gene or gene product. This can be the same composition administered prior to diagnosis of the specific infection or a different composition that decreases expression or activity.
Also provided is a method of preventing infection in a subject comprising administering to a subject susceptible to an unspecified infection a composition that decreases the expression or activity of a gene or gene product set forth in Table 1 , 2, 3 or 4. The composition can be administered in response to a lethal outbreak of an infection. For example, the infection can be a pandemic or a bioterrorist created infection. If there is a threat of an unspecified infection, such as a viral infection, a bacterial infection, a parasitic infection or an infection by a chimeric pathogenic agent, to name a few, a composition can be administered prophylactically to a subject to prevent an unspecified infection in a subject. The threat can also come in the form of a toxin. One of skill in the art would know to administer a composition that inhibits infection by decreasing the expression or activity of any one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4 that is involved in the pathogenesis of two or more, three or more, four or more; or five or more pathogens.
Such prophylactic use can decrease the number of people in a population that are infected, thus preventing further spread of a pandemic or decreasing the effects of a bioterrorist attack.
In the methods of the present invention, the composition can comprise one or more of, a chemical, a compound, a small molecule, an inorganic molecule, an aptamer, an organic molecule, a drug, a protein, a cDNA, a peptide, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an LNA, an antisense nucleic acid or a ribozyme that decreases the expression or activity of a gene or gene product set forth in Table 1, 2, 3 or 4. The composition can be administered before or after infection. The decrease in infection in a subject need not be complete as this decrease can be a 10% , 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any other percentage decrease in between as long as a decrease occurs. This decrease can be correlated with amelioration of symptoms associated with infection. These compositions can be administered to a subject alone or in combination with other therapeutic agents described herein, such as anti-viral compounds, antibacterial agents, antifungal agents, antiparasitic agents, anti-inflammatory agents, anti-cancer agents, etc. Examples of viral infections, bacterial infections, fungal infections parasitic infections are set forth above. The compounds set forth herein or identified by the screening methods set forth herein can be administered to a subject to decrease infection by any pathogen or infectious agent set forth herein. Any of the compounds set forth herein or identified by the screening methods of the present invention can also be administered to a subject to decrease infection by any pathogen, now known or later discovered in which a gene or gene product set forth in Table 1 , 2, 3 or 4 is involved. Various delivery systems for administering the therapies disclosed herein are known, and include encapsulation in liposomes, microparticles, microcapsules, expression by recombinant cells, receptor-mediated endocytosis (Wu and Wu, J. Biol. Chem. 1987, 262:4429-32), and construction of therapeutic nucleic acids as part of a retroviral or other vector. Methods of introduction include, but are not limited to, mucosal, topical, intradermal, intrathecal, intratracheal, via nebulizer, via inhalation, intramuscular, otic delivery (ear), eye delivery (for example, eye drops), intraperitoneal, vaginal, rectal, intravenous, subcutaneous, intranasal, and oral routes. The compounds can be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (for example, oral mucosa, rectal, vaginal and intestinal mucosa, etc.) and can be administered together with other biologically active agents. Administration can be systemic or local. Pharmaceutical compositions can be delivered locally to the area in need of treatment, for example by topical application or local injection.
Pharmaceutical compositions are disclosed that include a therapeutically effective amount of a RNA, DNA, antisense molecule, ribozyme, an LNA, siRNA, shRNA molecule, miRNA molecule, aptamer, drug, protein, small molecule, peptide inorganic molecule, organic molecule, antibody or other therapeutic agent, alone or with a pharmaceutically acceptable carrier. Furthermore, the pharmaceutical compositions or methods of treatment can be administered in combination with (such as before, during, or following) other therapeutic treatments, such as other antiviral agents, antibacterial agents, antifungal agents and antiparasitic agents.
For all of the administration methods disclosed herein, each method can optionally comprise the step of diagnosing a subject with an infection or diagnosing a subject in need of prophylaxis or prevention of infection.
Delivery systems
The pharmaceutically acceptable carriers useful herein are conventional. Remington 's Pharmaceutical Sciences, by Martin, Mack Publishing Co., Easton, PA, 15th Edition (1975), describes compositions and formulations suitable for pharmaceutical delivery of the therapeutic agents herein disclosed. In general, the nature of the carrier will depend on the mode of administration being employed. For instance, parenteral formulations usually include injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, sesame oil, glycerol, ethanol, combinations thereof, or the like, as a vehicle. The carrier and composition can be sterile, and the formulation suits the mode of administration. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
The composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder. For solid compositions (for example powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, sodium saccharine, cellulose, magnesium carbonate, or magnesium stearate. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Embodiments of the disclosure including medicaments can be prepared with conventional pharmaceutically acceptable carriers, adjuvants and counterions as would be known to those of skill in the art.
The amount of therapeutic agent effective in decreasing or inhibiting infection can depend on the nature of the pathogen and its associated disorder or condition, and can be determined by standard clinical techniques. Therefore, these amounts will vary depending on the type of virus, bacteria, fungus, parasite or other pathogen. For example, the dosage can be anywhere from 0.01 mg/kg to 100 mg/kg. Multiple dosages can also be administered depending on the type of pathogen, and the subject's condition. In addition, in vitro assays can be employed to identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems. The disclosure also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. Instructions for use of the composition can also be included. Instructions for the use of a composition to decrease infection or toxicity can also be included. For example instructions for decreasing infection by an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, a Hepatitis C virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE, WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus can be provided.
In an example in which a nucleic acid is employed to reduce infection, such as an antisense or siRNA molecule, the nucleic acid can be delivered intracellularly (for example by expression from a nucleic acid vector or by receptor-mediated mechanisms), or by an appropriate nucleic acid expression vector which is administered so that it becomes intracellular, for example by use of a retroviral vector (see U.S. Patent No. 4,980,286), or by direct injection, or by use of microparticle bombardment (such as a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (for example Joliot et al., Proc. Natl. Acad. ScL USA 1991, 88:1864-8). siRNA carriers also include, polyethylene glycol (PEG), PEG-liposomes, branched carriers composed of histidine and lysine (HK polymers), chitosan-thiamine pyrophosphate carriers, surfactants (for example, Survanta and Infasurf), nanochitosan carriers, and D5W solution. The present disclosure includes all forms of nucleic acid delivery, including synthetic oligos, naked DNA, plasmid and viral delivery, integrated into the genome or not.
As mentioned above, vector delivery can be via a viral system, such as a retroviral vector system which can package a recombinant retroviral genome (see e.g., Pastan et al., Proc. Natl. Acad. Sci. U.S.A. 85:4486, 1988; Miller et al., MoI. Cell Biol. 6:2895, 1986). The recombinant retrovirus can then be used to infect and thereby deliver to the infected cells a nucleic acid, for example an antisense molecule or siRNA. The exact method of introducing the altered nucleic acid into mammalian cells is, of course, not limited to the use of retroviral vectors. Other techniques are widely available for this procedure including the use of adenoviral vectors (Mitani et al., Hum. Gene Ther. 5:941-948, 1994), adeno-associated viral (AAV) vectors
(Goodman et al., Blood 84: 1492- 1500, 1994), lentiviral vectors (Naidini et al., Science 272:263- 267, 1996), and pseudotyped retroviral vectors (Agrawal et al., Exper. Hematol. 24:738-747, 1996). Other nonpathogenic vector systems such as the foamy virus vector can also be utilized (Park et al. "Inhibition of simian immunodeficiency virus by foamy virus vectors expressing siRNAs." Virology. 2005 Sep 20). It is also possible to deliver short hairpin RNAs (shRNAs) via vector delivery systems in order to inhibit gene expression (See Pichler et al. "In vivo RNA interference-mediated ablation of MDRl P-glycoprotein." Clin Cancer Res. 2005 Jun 15;1 1(12):4487-94; Lee et al. "Specific inhibition of HIV-I replication by short hairpin RNAs targeting human cyclin Tl without inducing apoptosis." FEBS Lett. 2005 Jun 6;579(14):3100- 6.).
Physical transduction techniques can also be used, such as liposome delivery and receptor-mediated and other endocytosis mechanisms (see, for example, Schwartzenberger et al., Blood 87:472-478, 1996) to name a few examples. This invention can be used in conjunction with any of these or other commonly used gene transfer methods.
Transgenic Cells and Non-Human Mammals
The present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, 2, 3 or 4, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a fungus or a parasite. Exemplary transgenic non-human mammals include, but are not limited to, ferrets, fish, guinea piags, chinchilla, mice, monkeys, rabbits, rats, chickens, cows, and pigs. Such knock-out animals are useful for reducing the transmission of viruses from animals to humans and for further validating a target. In the transgenic animals of the present invention one or both alleles of a gene set forth in Table 1, 2, 3 or 4 can be functionally deleted. By "decreased susceptibility" is meant that the animal is less susceptible to infection or experiences decreased infection by a pathogen as compared to an animal that does not have one or both alleles of a a gene set forth in Table 1, 2, 3 or 4 functionally deleted. The animal does not have to be completely resistant to the pathogen. For example, the animal can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% or any percentage in between less susceptible to infection by a pathogen as compared to an animal that does not have a functional deletion of a gene set forth in Table 1, 2, 3 or 4. Furthermore, decreasing infection or decreasing susceptibility to infection includes decreasing entry, replication, pathogenesis, insertion, lysis, or other steps in the replication strategy of a virus or other pathogen into a cell or subject, or combinations thereof. Therefore, the present invention provides a non- human transgenic mammal comprising a functional deletion of a gene set forth in Table 1 , 2, 3 or 4, wherein the mammal has decreased susceptibility to infection by a pathogen, such as a virus, a bacterium, a parasite or a fungus. A functional deletion is a mutation, partial or complete deletion, insertion, or other variation made to a gene sequence that inhibits production of the gene product or renders a gene product that is not completely functional or non-functional. Functional deletions can be made by insertional mutagenesis (for example via insertion of a transposon or insertional vector), by site directed mutagenesis, via chemical mutagenesis, via radiation or any other method now known or developed in the future that results in a transgenic animal with a functional deletion of a gene set forth in Table 1, 2, 3 or 4. More particularly, the present invention provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 1, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not HIV. Also provided is a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 2, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not West Nile Virus. Further provided is non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 3, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not West Nile Virus or Dengue Fever Virus. The present invention also provides a non-human transgenic mammal comprising a functional deletion of a gene set forth in Table 4, wherein the mammal has decreased susceptibility to infection by a pathogen, wherein the pathogen is not hepatitis C. Alternatively, a nucleic acid sequence such as siRNA, antisense, LNA, a morpholino or another agent that interferes with a gene set forth in Table 1 , 2, 3 or 4 mRNA expression can be delivered. The expression of the sequence used to knock-out or functionally delete the desired gene can be regulated by an appropriate promoter sequence. For example, constitutive promoters can be used to ensure that the functionally deleted gene is not expressed by the animal. In contrast, an inducible promoter can be used to control when the transgenic animal does or does not express the gene of interest. Exemplary inducible promoters include tissue- specific promoters and promoters responsive or unresponsive to a particular stimulus (such as light, oxygen, chemical concentration, such as a tetracycline inducible promoter). The transgenic animals of the present invention that comprise a functionally deleted a gene set forth in Table 1 , 2, 3 or 4 can be examined during exposure to various pathogens. Comparison data can provide insight into the life cycles of pathogens. Moreover, knock-out animals or functionally deleted (such as birds or pigs) that are otherwise susceptible to an infection (for example influenza) can be made to resist infection, conferred by disruption of the gene. If disruption of the gene in the transgenic animal results in an increased resistance to infection, these transgenic animals can be bred to establish flocks or herds that are less susceptible to infection.
Transgenic animals, including methods of making and using transgenic animals, are described in various patents and publications, such as WO 01/43540; WO 02/1981 1; U.S. Pub. Nos: 2001-0044937 and 2002-00661 17; and U.S. Pat. Nos: 5,859,308; 6,281,408; and 6,376,743; and the references cited therein.
The transgenic animals of this invention also include conditional gene knockdown animals produced, for example, by utilizing the SIRIUS-Cre system that combines siRNA for specific gene-knockdown, Cre-loxP for tissue-specific expression and tetracycline-on for inducible expression. These animals can be generated by mating two parental lines that contain a specific siRNA of interest gene and tissue-specific recombinase under tetracycline control. See Chang et al. "Using siRNA Technique to Generate Transgenic Animals with Spatiotemporal and Conditional Gene Knockdown." American Journal of Pathology 165: 1535-1541 (2004) which is hereby incorporated in its entirety by this reference regarding production of conditional gene knockdown animals.
The present invention also provides cells including an altered or disrupted gene set forth in Table 1, 2, 3 or 4 that are resistant to infection by a pathogen. These cells can be in vitro, ex vivo or in vivo cells and can have one or both alleles altered. These cells can also be obtained from the transgenic animals of the present invention. Such cells therefore include cells having decreased susceptibility to a virus or any of the other pathogens described herein, including bacteria, parasites and fungi.
Since the genes set forth herein are involved in viral infection, also provided herein are methods of overexpressing any of the genes set forth in Table 1 in host cells. Overexpression of these genes can provide cells that increase the amount of virus produced by the cell, thus allowing more efficient production of viruses. Also provided is the overexpression of the genes set forth herein in avian eggs, for example, in chicken eggs.
Methods of screening agents, such as a chemical, a compound, a small or large molecule, an organic molecule, an inorganic molecule, a peptide, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an siRNA, an shRNAs, an miRNA, an antisense nucleic acid or a ribozyme set forth using the transgenic animals described herein are also provided.
Screening for Resistance to Infection
Also provided herein are methods of screening host subjects for resistance to infection by characterizing a nucleotide sequence or amino acid sequence of a host gene set forth in Table 1, 2, 3 or 4. The nucleic acid or amino acid sequence of a subject can be isolated, sequenced, and compared to the wildtype sequence of a gene set forth in Table 1 , 2, 3 or 4. The greater the similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more susceptible that person is to infection, while a decrease in similarity between that subject's nucleic acid sequence or amino acid sequence and the wildtype sequence, the more resistant that subject can be to infection. Such screens can be performed for any gene set forth in Table 1, 2, 3 or 4 for any species.
Assessing the genetic characteristics of a population can provide information about the susceptibility or resistance of that population to viral infection. For example, polymorphic analysis of alleles in a particular human population, such as the population of a particular city or geographic area, can indicate how susceptible that population is to infection. A higher percentage of alleles substantially similar to a wild-type gene set forth in Table 1, 2, 3 or 4 can indicate that the population is more susceptible to infection, while a large number of polymorphic alleles that are substantially different than a wild-type gene sequence can indicate that a population is more resistant to infection. Such information can be used, for example, in making public health decisions about vaccinating susceptible populations.
The present invention also provides a method of screening a cell for a variant form of a gene set forth in Table 1, 2, 3 or 4. A variant can be a gene with a functional deletion, mutation or alteration in the gene such that the amount or activity of the gene product is altered. These cells containing a variant form of a gene can be contacted with a pathogen to determine if cells comprising a naturally occurring variant of a gene set forth in Table 1, 2, 3 or 4 differs in their resistance to infection. For example, cells from an animal, for example, a chicken, can be screened for a variant form of a gene set forth in Table 1, 2, 3 or 4. If a naturally occurring variant is found and chickens possessing a variant form of the gene in their genome are less susceptible to infection, these chickens can be selectively bred to establish flocks that are resistant to infection. By utilizing these methods, flocks of chickens that are resistant to avian flu or other pathogens can be established. Similarly, other animals can be screened for a variant form of a gene set forth in Table 1, 2, 3 or 4. If a naturally occurring variant is found and animals possessing a variant form of the gene in their genome are less susceptible to infection, these animals can be selectively bred to establish populations that are resistant to infection. These animals include, but are not limited to, cats, dogs, fish, livestock (for example, cattle, horses, pigs, sheep, goats, etc.), laboratory animals (for example, mouse,monkey, rabbit, rat, gerbil, guinea pig, etc.) and avian species (for example, flocks of chickens, geese, turkeys, ducks, pheasants, pigeons, doves etc.). Therefore, the present application provides populations of animals that comprise a naturally occurring variant of a gene set forth in Table 1, 2, 3 or 4 that results in decreased susceptibility to viral infection, thus providing populations of animals that are less susceptible to viral infection. Similarly, if a naturally occurring variant is found and animals possessing a variant form of the gene in their genome are less susceptible to bacterial, parasitic or fungal infection, these animals can be selectively bred to establish populations that are resistant to bacterial, parasitic or fungal infection.
Screening Methods
The present invention provides a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection of a cell by a pathogen comprising: a) contacting a compound with a gene product set forth in Table 1, 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating the binding with a decrease in infection by the pathogen.
The present invention provides a method of identifying an agent that decreases infection of a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4; and b) detecting the level and/or activity of the gene product produced by the cellular gene, a decrease or elimination of the gene product and/or gene product activity indicating an agent with antipathogenic activity. Also provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; b) contacting the cell with a pathogen, wherein the pathogen is not HIV; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
Further provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 2; b) contacting the cell with a pathogen, wherein the pathogen is not West Nile Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
Further provided is a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 3; b) contacting the cell with a pathogen, wherein the pathogen is not West Nile Virus or Dengue Fever Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
The present invention also provides a method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 4; b) contacting the cell with a pathogen, wherein the pathogen is not hepatitis C virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection. This method can further comprise measuring the level of expression and/or activity of the gene product. The level of infection can be determined by determining the level of replication of the pathogen.
The present invention also provides a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection by three or more pathogens comprising: a) contacting a compound with a gene product set forth in Table 1, 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens. This method can further comprise optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens. This method can be cell based or an in vivo assay. The three or more pathogens can be any three or more pathogens set forth herein. For example, the three or more pathogens can be respiratory pathogens selected from the group consisting of picornaviruses, orthomyxoviruses, paramyxoviruses, coronaviruses or adenoviruses. In another example, the three or more pathogens can be gastrointestinal pathogens selected from filoviruses, flaviviruses, calciviruses and reoviruses. The three or more pathogens can also be a combination of respiratory and gastrointestinal viruses. In another example, the three or more pathogens can be selected from the group consisting of : an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus. The cell population used in the method can be the same cell population for each pathogen or can be different cell populations. Typically, the agent would be administered to a different cell population for each pathogen assayed. For example, and not to be limiting, if the pathogens are viruses, a cell population is contacted with the agent and a first virus, another cell population is contacted with the agent and second virus, a third cell population is contacted with the agent and a third virus etc. in order to determine whether the agent inhibits infection by three or more viruses. Since the cell type will vary depending on whether or not a given virus can infect the cell, one of skill in the art would know how to pair the cell type with the virus in order to perform the assay. This method can further comprise measuring the level of expression and/or activity of the gene product set forth in Table 1, 2, 3 or 4. This method can further comprise associating the level of infection with the level of expression and/or activity a gene product set forth in Table 1, 2, 3 or 4. In the screening methods disclosed herein, the level of infection can be measured, for example, by measuring viral replication. In the methods of the present invention, if the agent has previously been identified as an agent that decreases or inhibits the level and/or activity of a gene product set forth in Table 1, 2, 3 or 4, this can indicate a decrease in infection. Previous identification of the agent can occur via experimentation, literature, chemical databases, catalogues, or any other means that allow one of skill in the art to associate the agent as an inhibitor of the gene or gene product. A decrease in infection as compared to infection in a cell that was not contacted with the agent known to decrease or inhibit the level and/or activity of the gene product can be sufficient to identify the agent as an agent that decreases or inhibits infection.
The methods described above can be utilized to identify any agent with an activity that decreases infection, prevents infection or promotes cellular survival after infection with a pathogen(s). Therefore, the cell can be contacted with a pathogen before, or after being contacted with the agent. The cell can also be contacted concurrently with the pathogen and the agent. The agents identified utilizing these methods can be used to inhibit infection in cells either in vitro, ex vivo or in vivo.
In the methods of the present invention any cell that can be infected with a pathogen can be utilized. The cell can be prokaryotic or eukaryotic, such as a cell from an insect, fish, crustacean, mammal, bird, reptile, yeast or a bacterium, such as E. coli. The cell can be part of an organism, or part of a cell culture, such as a culture of mammalian cells or a bacterial culture. The cell can also be in a nonhuman subject thus providing in vivo screening of agents that decrease infection by a pathogen. Cells susceptible to infection are well known and can be selected based on the pathogen of interest.
The test agents or compounds used in the methods described herein can be, but are not limited to, chemicals, small molecules, inorganic molecules, organic molecules, drugs, proteins, cDNAs, aptamers, large molecules, antibodies, morpholinos, triple helix molecule, peptides, siRNAs, shRNAs, miRNAs, LNAs, antisense RNAs, ribozymes or any other compound. The compound can be random or from a library optimized to bind to a gene or gene product set forth in Table 1, 2, 3 or 4. Drug libraries optimized for the proteins in the class of proteins provided herein can also be screened or tested for binding or activity. Compositions identified with the disclosed approaches can be used as lead compositions to identify other compositions having even greater antipathogenic activity. For example, chemical analogs of identified chemical entities, or variants, fragments or fusions of peptide agents, can be tested for their ability to decrease infection using the disclosed assays. Such analogs can be purchased or synthesized via routine methods known to those of skill in the art. Candidate agents can also be tested for safety in animals and then used for clinical trials in animals or humans.
In the methods described herein, once the cell containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4 has been contacted with an agent, the level of infection can be assessed by measuring an antigen or other product associated with a particular infection. For example, the level of viral infection can be measured by real-time quantitative reverse transcription-poly merase chain reaction (RT-PCR) assay (See for example, Payungporn et al. "Single step multiplex real-time RT-PCR for H5N1 influenza A virus detection." J Virol Methods. Sep 22, 2005; Landolt et Ia. "Use of real-time reverse transcriptase polymerase chain reaction assay and cell culture methods for detection of swine influenza A viruses" Am J Vet Res. 2005 Jan;66(l):l 19-24). If there is a decrease in infection then the composition is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between.
In the methods set forth herein, the level of the gene product can be measured by any standard means, such as by detection with an antibody specific for the protein. The nucleic acids set forth herein and fragments thereof can be utilized as primers to amplify nucleic acid sequences, such as a gene transcript of a gene set forth in Table 1, 2, 3 or 4 by standard amplification techniques. For example, expression of a gene transcript can be quantified by real time PCR using RNA isolated from cells. A variety of PCR techniques are familiar to those skilled in the art. For a review of PCR technology, see White (1997) and the publication entitled "PCR Methods and Applications" (1991, Cold Spring Harbor Laboratory Press), which is incorporated herein by reference in its entirety for amplification methods. In each of these PCR procedures, PCR primers on either side of the nucleic acid sequences to be amplified are added to a suitably prepared nucleic acid sample along with dNTPs and a thermostable polymerase such as Taq polymerase, Pfu polymerase, or Vent polymerase. The nucleic acid in the sample is denatured and the PCR primers are specifically hybridized to complementary nucleic acid sequences in the sample. The hybridized primers are extended. Thereafter, another cycle of denaturation, hybridization, and extension is initiated. The cycles are repeated multiple times to produce an amplified fragment containing the nucleic acid sequence between the primer sites. PCR has further been described in several patents including U.S. Pat. Nos. 4,683,195, 4,683,202 and 4,965,188. Each of these publications is incorporated herein by reference in its entirety for PCR methods. One of skill in the art would know how to design and synthesize primers that amplify any of the nucleic acid sequences set forth herein or a fragment thereof.
A detectable label may be included in an amplification reaction. Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein (6-FAM), 2',7'-dimethoxy-4',5'-dichloro-6- carboxyfluorescein (JOE), 6-carboxy-X-rhodamine (ROX), 6-carboxy-2',4',7',4,7- hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N',N'-tetramethyl-6- carboxyrhodamine (TAMRA), radioactive labels, e.g., 32 P, 35 S, 3 H; etc. The label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label. The label may be conjugated to one or both of the primers. Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.
The sample nucleic acid, e.g. amplified fragment, can be analyzed by one of a number of methods known in the art. The nucleic acid can be sequenced by dideoxy or other methods. Hybridization with the sequence can also be used to determine its presence, by Southern blots, dot blots, etc.
In the methods of the present invention, the level of gene product can be compared to the level of the gene product in a control cell not contacted with the compound. The level of gene product can be compared to the level of the gene product in the same cell prior to addition of the compound. Activity or function, can be measured by any standard means, such as by enzymatic assays that measure the conversion of a substrate to a product or binding assays that measure the binding of a gene product set forth in Table 1, 2, 3 or 4 to another protein, for example.
Moreover, the regulatory region of a gene set forth in Table 1 , 2, 3 or 4 can be functionally linked to a reporter gene and compounds can be screened for inhibition of reporter gene expression. Such regulatory regions can be isolated from genomic sequences and identified by any characteristics observed that are characteristic for regulatory regions of the species and by their relation to the start codon for the coding region of the gene. As used herein, a reporter gene encodes a reporter protein. A reporter protein is any protein that can be specifically detected when expressed. Reporter proteins are useful for detecting or quantitating expression from expression sequences. Many reporter proteins are known to one of skill in the art. These include, but are not limited to, β-galactosidase, luciferase, and alkaline phosphatase that produce specific detectable products. Fluorescent reporter proteins can also be used, such as green fluorescent protein (GFP), cyan fluorescent protein (CFP), red fluorescent protein (RFP) and yellow fluorescent protein (YFP). Viral infection can also be measured via cell-based assays. Briefly, cells (20,000 to
2,500,000) are infected with the desired pathogen, and the incubation continued for 2-7 days. The incubation time is routinely determined by one of skill in the art who would know the incubation times for a particular pathogen. The antiviral agent can be applied to the cells before, during, or after infection with the pathogen. The amount of virus and agent administered can be determined by skilled practitioners. In some examples, several different doses of the potential therapeutic agent can be administered, to identify optimal dose ranges. Following transfection, assays are conducted to determine the resistance of the cells to infection by various agents.
For example, if analyzing viral infection, the presence of a viral antigen can be determined by using antibody specific for the viral protein then detecting the antibody. In one example, the antibody that specifically binds to the viral protein is labeled, for example with a detectable marker such as a fluorophore. In another example, the antibody is detected by using a secondary antibody containing a label. The presence of bound antibody is then detected, for example using microscopy, flow cytometry and ELISA. Similar methods can be used to monitor bacterial, protozoal, or fungal infection (except that the antibody would recognize a bacterial, protozoal, or fungal protein, respectively).
Alternatively, or in addition, the ability of the cells to survive viral infection is determined, for example, by performing a cell viability assay, such as trypan blue exclusion. Plaque assays can be utilized as well.
The amount of protein in a cell, can be determined by methods standard in the art for quantitating proteins in a cell, such as Western blotting, ELISA, ELISPOT, immunoprecipitation, immunofluorescence (e.g., FACS), immunohistochemistry, immunocytochemistry, etc., as well as any other method now known or later developed for quantitating protein in or produced by a cell.
The amount of a nucleic acid in a cell can be determined by methods standard in the art for quantitating nucleic acid in a cell, such as in situ hybridization, quantitative PCR, RT-PCR, Taqman assay, Northern blotting, ELISPOT, dot blotting, etc., as well as any other method now known or later developed for quantitating the amount of a nucleic acid in a cell.
The ability of an antiviral agent to prevent or decrease infection by a virus, for example, any of the viruses listed above, can be assessed in an animal model. Several animal models for viral infection are known in the art. For example, mouse HIV models are disclosed in Sutton et al. (Res. Initiat Treat. Action, 8:22-4, 2003) and Pincus et al. (AIDS Res. Hum. Retroviruses 19:901-8, 2003); guinea pig models for Ebola infection are disclosed in Parren et al. (J. Virol. 76:6408-12, 2002) and Xu et al. (Nat. Med. 4:37-42, 1998); chimpanzee models for HCV are disclosed (Lanford et al., ILAR J. 2001 ;42(2):117-26); cynomolgus monkey (Macaca fascicularis) models for influenza infection are disclosed in Kuiken et al. (Vet. Pathol. 40:304- 10, 2003); mouse models for RSV are also disclosed (Sudo et al. Antivir Chem Chemother. 1999 May; 10(3): 135-9) mouse models for herpes are disclosed in Wu et al. (Cell Host Microbe 22:5(l):84-94. 2009); mouse models for rhinovorus are disclosed (Bartlett et al. Nat Med. 2008 Feb; 14(2): 199-204); pox models are disclosed in Smee et al. (Nucleosides Nucleotides Nucleic Acids 23(l-2):375-83, 2004) and in Bray et al. (J. Infect. Dis. 181(1): 10-19); animal models for Dengue are available (Shresta et al. J Virol. 2006 October; 80(20): 10208-1021 ; Bente and Rico-Hesse, Drug Discovery Today: Disease Models 3(1) 2006: 97-103; Martin et al. Microbiol Immunol. 2009 Apr;53(4):216-23); and Franciscella tularensis models are disclosed in Klimpel et al. (Vaccine 26(52): 6874-82, 2008). Other animal models for influenza infection are also available. These include, but are not limited to, a cotton rat model disclosed by Ottolini et al. (J. Gen. Virol, 86(Pt 10): 2823-30, 2005), as well as ferret and mouse models disclosed by Maines et al. (J. Virol. 79(18): 11788-11800, 2005). One of skill in the art would know how to select an animal model for assessing the in vivo activity of an agent for its ability to decrease infection by viruses, bacteria, fungi and parasites. One of skill in the art would know how to select an animal model for assessing the in vivo activity of an agent for its ability to decrease infection by viruses, bacteria, fungi and parasites. Such animal models can also be used to test agents for the ability to ameliorate symptoms associated with viral infection. In addition, such animal models can be used to determine the LD50 and the ED50 in animal subjects, and such data can be used to determine the in vivo efficacy of potential agents. LD50 is an index of toxicity (lethal dose 50%), the amount of the substance that kills 50% of the test population of experimental animals when administered as a single dose. ED50 is the dose of a drug that is pharmacologically effective for 50% of the population exposed to the drug or a 50% response in a biological system that is exposed to the drug. Animal models can also be used to assess antibacterial, antifungal and antiparasitic agents.
Animals of any species, including, but not limited to, birds, ferrets, cats, mice, rats, rabbits, fish (for example, zebrafish or koi) guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, can be used to generate an animal model of viral infection, bacterial infection, fungal infection or parasitic infection if needed. For example, for a model of viral infection, the appropriate animal is inoculated with the desired virus, in the presence or absence of the antiviral agent. The amount of virus and agent administered can be determined by skilled practitioners. In some examples, several different doses of the potential therapeutic agent (for example, an antiviral agent) can be administered to different test subjects, to identify optimal dose ranges. The therapeutic agent can be administered before, during, or after infection with the virus. Subsequent to the treatment, animals are observed for the development of the appropriate viral infection and symptoms associated therewith. A decrease in the development of the appropriate viral infection, or symptoms associated therewith, in the presence of the agent provides evidence that the agent is a therapeutic agent that can be used to decrease or even inhibit viral infection in a subject. For example, a virus can be tested which is lethal to the animal and survival is assessed. In other examples, the weight of the animal or viral titer in the animal can be measured. Similar models and approaches can be used for bacterial, fungal and parasitic infections.
In the methods of the present invention, the level of infection can be associated with the level of gene expression and/or activity, such that a decrease or elimination of infection associated with a decrease or elimination of gene expression and/or activity indicates that the agent is effective against the pathogen. For example, the level of infection can be measured in a cell after administration of siRNA that is known to inhibit a gene product set forth in Table 1, 2, 3 or 4. If there is a decrease in infection then the siRNA is an effective agent that decreases infection. This decrease does not have to be complete as the decrease can be a 10%, 20%, 30%, 40%, 50%, 60%. 70%, 80%, 90%, 100% decrease or any percentage decrease in between. In the event that the compound is not known to decrease expression and/or activity of a gene product set forth in Table 1, 2, 3 or 4, the level of expression and/or activity of can be measured utilizing the methods set forth above and associated with the level of infection. By correlating a decrease in expressionand/or activity with a decrease in infection, one of skill in the art can confirm that a decrease in infection is effected by a decrease in expression and/or activity of a gene or gene product set forth in Table 1, 2, 3 or 4. Similarly, the level of infection can be measured in a cell, utilizing the methods set forth above and known in the art, after administration of a chemical, small molecule, drug, protein, cDNA, antibody, aptamer, shRNA, miRNA, morpholino, antisense RNA, LNA, ribozyme or any other compound. If there is a decrease in infection, then the chemical, small molecule, drug, protein, cDNA, antibody, shRNA, miRNA, morpholino, antisense RNA, ribozyme or any other compound is an effective antpathogenic agent.
The genes and nucleic acids of the invention can also be used in polynucleotide arrays. Polynucleotide arrays provide a high throughput technique that can assay a large number of polynucleotide sequences in a single sample. This technology can be used, for example, to identify samples with reduced expression of as compared to a control sample. This technology can also be utilized to determine the effects of reduced expression of a gene set forth in Table 1, 2, 3 or 4 on other genes. In this way, one of skill in the art can identify genes that are upregulated or downregulated upon reducing expression of a gene set forth in Table 1, 2, 3 or 4. Similarly, one of skill in the art can identify genes that are upregulated or downregulated upon increased expression of a gene set forth in Table 1, 2, 3, or 4. This allows identification of other genes that are upregulated or downregulated upon modulation of expression that can be targets for therapy, such as antiviral therapy, antibacterial therapy, antiparasitic therapy or antifungal therapy.
To create arrays, single- stranded polynucleotide probes can be spotted onto a substrate in a two-dimensional matrix or array. Each single-stranded polynucleotide probe can comprise at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or 30 or more contiguous nucleotides selected from nucleotide sequences set forth under GenBank Accession Nos. herein and other nucleic acid sequences that would be selected by one of skill in the art depending on what genes, in addition to one or more of the genes set forth in Table 1 , 2, 3 or 4 are being analyzed.
The array can also be a microarray that includes probes to different polymorphic alleles of these genes. A polymorphism exists when two or more versions of a nucleic acid sequence exist within a population of subjects. For example, a polymorphic nucleic acid can be one where the most common allele has a frequency of 99% or less. Different alleles can be identified according to differences in nucleic acid sequences, and genetic variations occurring in more than 1% of a population (which is the commonly accepted frequency for defining polymorphism) are useful polymorphisms for certain applications. The allelic frequency (the proportion of all allele nucleic acids within a population that are of a specified type) can be determined by directly counting or estimating the number and type of alleles within a population. Polymorphisms and methods of determining allelic frequencies are discussed in Hartl, D.L. and Clark, A.G., Principles of Population Genetics, Third Edition (Sinauer Associates, Inc., Sunderland Massachusetts, 1997), particularly in chapters 1 and 2.
These microarrays can be utilized to detect polymorphic alleles in samples from subjects. Such alleles may indicate that a subject is more susceptible to infection or less susceptible to infection. For example, microarrays can be utilized to detect polymorphic versions of genes set forth in Table 1 , 2, 3 or 4 that result in decreased gene expression and/or decreased activity of the gene product to identify subjects that are less susceptible to viral infection. In addition, the existence of an allele associated with decreased expression in a healthy individual can be used to determine which genes are likely to have the least side effects if the gene product is inhibited or bound or may be selected for in commercial animals and bred into the population.
The substrate can be any substrate to which polynucleotide probes can be attached, including but not limited to glass, nitrocellulose, silicon, and nylon. Polynucleotide probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions. Techniques for constructing arrays and methods of using these arrays are described in EP No. 0 799 897; PCT No. WO 97/29212; PCT No. WO 97/27317; EP No. 0 785 280; PCT No. WO 97/02357; U.S. Pat. Nos. 5,593,839; 5,578,832; EP No. 0 728 520; U.S. Pat. No. 5,599,695; EP No. 0 721 016; U.S. Pat. No. 5,556,752; PCT No. WO 95/22058; and U.S. Pat. No. 5,631,734. Commercially available polynucleotide arrays, such as Affymetrix GeneChip.TM., can also be used. Use of the GeneChip.™. to detect gene expression is described, for example, in Lockhart et al., Nature Biotechnology 14:1675 (1996); Chee et al., Science 274:610 (1996); Hacia et al., Nature Genetics 14:441, 1996; and Kozal et al., Nature Medicine 2:753, 1996.
The present invention also provides a method of identifying an agent that can decrease infection by three or more pathogens comprising: a) administering the agent to three or more cell populations containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4; b) contacting the three or more cell populations with a pathogen selected wherein each population is contacted with a different pathogen; and c) determining the level of infection, a decrease or elimination of infection by three or more pathogens indicating that the agent is an agent that decreases infection by three or more pathogens. In the screening methods set forth herein, the three or more pathogens can be three or more respiratory viruses selected from the one or more families from group consisting of: picornaviruses, an orthomyxoviruses, a paramyxoviruses, a coronaviruses, or an adenoviruses. The three or more pathogens can be three or more gastrointestinal viruses selected from one or more families from the group consisting of: flaviviruses, filoviruses, calciviruses or reoviruses. In another example, the three or more pathogens can be three or more viruses selected from gastrointestinal viruses and respiratory viruses. In another example, the three or more pathogens can be selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, West Nile virus, Chikungunya virus or a Dengue fever virus. The three or more pathogens can also be selected from the group consisting of: HIV, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus. In another example, the three or more pathogens can be selected from the group consisting of: influenza, a pox virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, hantavirus, Rift Valley Fever virus Ebola virus, Marburg virus or Dengue Fever virus
Also provided is a method of identifying a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and can decrease infection by three or more pathogens comprising: a) contacting a compound with a gene product set forth in Table 1 , 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens. This method can further comprise optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens. A set forth above, as utilized herein "three or more" includes four or more, five or more, six or more, seven or more, etc. This assay can be cell based or in vivo based. Methods of making compounds
The present invention provides_a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; c) contacting the cell with an infectious pathogen, wherein the pathogen is not HIV; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
Also provided is a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 2; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile Virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
Further provided is a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 3; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile virus or Dengue fever virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product.
The present invention also provides_a method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 4; c) contacting the cell with an infectious pathogen, wherein the pathogen is not hepatitis C; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection; and e) associating the agent with decreasing expression or activity of the gene product. The association can be made by measuring the level of expression and/or activity of the gene product. As set forth above, the association can also be made by one of skill in the art if there is a priori knowledge, for example, via experimentation, literature, chemical databases or catalogues that the agent decreases expression and or activity of a gene or gene product. The level of infection can be determined by determining the level of replication of the pathogen. Further provided is a method of making a compound that decreases infection in a cell by a pathogen, comprising: a) optimizing a compound to bind a gene product set forth in Table 1, 2, 3 or 4; b) administering the compound to a cell containing a cellular gene encoding the gene product; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating the making of a compound that decreases infection in a cell by a pathogen. This method can further synthesizing therapeutic quantities of the compound. The method can further comprise packaging of the therapeutic quantities.
The present invention also provides a method of synthesizing a compound that binds to a gene product set forth in Table 1 , 2, 3 or 4 and decreases infection by a pathogen comprising: a) contacting a library of compounds with a gene product set forth in Table 1, 2, 3 or 4; b) associating binding with a decrease in infection; and c) synthesizing derivatives of the compounds from the library that bind to the gene product.
The present invention also provides a business method to reduce the cost of drug discovery of drugs that can reduce infection by a pathogen comprising: screening, outside of the United States, for drugs that reduce infection by binding to or reducing the function of a gene product set forth in Table 1, 2, 3 or 4; and b) importing drugs that reduce infection into the United States. Also provided is a method of making drugs comprising directing the synthesis of drugs that reduce infection by binding to or reducing the function of a gene or gene product set forth in Table 1, 2, 3 or 4. EXAMPLES siRNA studies
Any of the genes set forth in Table 1, 2, 3 or 4 is further analyzed by contacting cells comprising a gene set forth in Table 1, 2, 3 or 4 with siRNA that specifically targets the gene product of the gene, and any pathogen set forth herein to identify the gene as a gene involved in pathogenic infection (for example, and not to be limiting, an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Ban- Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, West Nile virus, Chikungunya virus or a Dengue fever virus). A decrease in viral infection indicates that the gene is a gene that is involved in pathogenic infection. This process can be performed for all of the genes set forth herein with any of the viruses, bacteria, parasites or fungi set forth herein. siRNA Transfections can be performed as follows: Pools of 4 duplexed siRNA molecules targeting a gene of interest are reconstituted to a final working concentration of 5OuM as directed by the manufacturer (Qiagen). Twenty-four hours prior to transfection, cells are plated in 6-well dishes at 3x105 cells per well, such that at the time of transfection, the cells are approximately 30% confluent. Prior to transfection, the cells are washed once with IX phosphate buffered saline, and the medium replaced with approximately 1.8ml antibiotic- free medium. siRNA aliquots are diluted with Opti-MEM and RNAseOUT (Invitrogen), lOOul and IuI per transfection, respectively. In a separate tube, transfection reagent Lipofectamine2000 (Invitrogen) or Oligofectamine (Invitrogen) are diluted in Opti-MEM as directed by the manufacturer. Following a 5 minute incubation at room temperature, the diluted siRNA is added to the transfection reagent mixture, and incubated for an additional 20 minutes prior to adding to independent wells of the 6-well dishes. Transfections are incubated at 37°C for 48 hours without changing the medium.
Virus Infections: Following 48-hour transfection, medium is aspirated from 6-well plates. Viruses are diluted in the appropriate medium and 500ul of either virus-free medium or virus dilution is added to each well, and adsorption is allowed to occur at the appropriate temperature for 1 hour. Following adsorption, inoculum is aspirated off the cells, cells are washed once with IX phosphate buffered saline, and 2ml growth medium is added to the cells. The infected cells are incubated for 72 hours at the appropriate temperature prior to harvesting samples for viral titration.
Viral Genomic Extractions: Seventy-hours after inoculating cells, medium is harvested from 6-well dishes and centrifuged for 2 minutes at 10,000 rpm to remove any cellular debris. 200ul of clarified medium is added to 25ul Proteinase K, to which 200ul PureLink96 Viral RNA/DNA lysis buffer (Invitrogen) is added according to the manufacturer. Samples were processed and viral genomic RNA or DNA is extracted using an epMotion 5075 robotics station (Eppendorf) and the PureLink96 Viral RNA/DNA kit (Invitrogen). cDNA and Quantitative Real-Time PCR Reactions: 3ul of extracted viral RNA is converted to cDNA using M-MLV reverse transcriptase (Invitrogen) and AmpliTaq Gold PCR buffer (Applied Biosystems). MgCl2, dNTPs and RNAseOUT (Invitrogen) are added to achieve a final concentration of 5mM, ImM and 2U/ul, respectively. Random hexamers (Applied
Biosystems) are added to obtain 2.5mM final concentration. Reactions are incubated at 42°C for 1 hour, followed by heat inactivation of the reverse transcriptase at 92°C for 10 minutes. Quantitative real-time PCR reactions are set up in lOul volumes using IuI of template cDNA or extracted viral DNA using virus-specific TaqMan probes (Applied Biosystems) and RealMasterMix (Eppendorf). 2-step reactions are allowed to proceed through 40 to 50 cycles on an ep RealPlex thermocycler (Eppendorf). Quantitative standards for real-time PCR are constructed by cloning purified amplicons into pCR2-TOPO (Invitrogen) and sequenced as necessary. The amount of viral replication in the cells contacted with siRNA to the gene of interest is calculated and compared to the amount of viral replication in control cells that did not receive siRNA targeting the gene of interest.
Compounds are assessed as follows. Duplicate experiments are performed as follows. Prior to compound exposure cells are seeded into 12-well tissue culture plates and allowed to adhere to the vessel surface. The compounds are serially diluted and lOOμl is added to the growth medium in triplicate wells to obtain the indicated final concentration of compound or solvent. Cells are incubated in the presence of the test compound for about 24 hours at 37°C under 5% CO2.
Following the pre-treatment period, cells are examined under a phase-contrast microscope to determine any cytotoxic effects at the treatment dosages. The pre-treatment medium is then aspirated off the cells, and the specified virus, diluted in serum- and compound- free growth medium, is added to the cells. Virus is allowed to adsorb to the cells at 37°C under 5% CO2. Following adsorption, the viral inoculum is aspirated off the cells and replaced with growth medium supplemented with 2% FBS, 1% L-glutamine, 1% penicillin/streptomycin and the respective concentration of test compound. Cells are incubated under the same conditions for an additional 72 hours. As stated above, incubation period can vary depending on the pathogen. 200μl samples are harvested from each well and viral RNA or DNA is extracted using the PureLink 96 Viral RNA/DNA extraction kit (Invitrogen) as directed by the manufacturer. Viral nucleic acid is subjected to reverse transcription to generate cDNA, which is subsequently used as the template for quantitative real-time TaqMan PCR to titrate viral replication using a Mastercycler ep realplex2 (Eppendorf). The experiment described above can be performed to determine the effect of any of the compounds set forth herein on any pathogenic infection, for example, and not to be limiting, a pox virus, BVDV, HIV, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, CaI ici viruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
Any of the experiments set forth herein can optionally comprise the step of assessing toxicity of any of the compounds set forth herein via any of the toxicity measurement methods described herein, or via any of the toxicity measurement methods known to one of skill in the art, such as, for example, the CytoTox-Glo assay (see Niles, A. et al. (2007) Anal. Biochem. 366, 197-206) or the Cell-Titer-Glo assay from Promega.

Claims

What is claimed is:
1. A method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) selected from the group consisting of EDNRA, BCR, CA2, CHRNA7, CXCR4, EGFR, F2, GRI A2, HDACl, KCNJl 1, MMP9, PDE8A, PLKl , POLE, SRD5A2, TUBA8 and DPP4, wherein the pathogen is not HIV.
2. A method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table
1 , wherein the pathogen is not HIV.
3. A method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table
2, wherein the pathogen is not West Nile Virus.
4. The method of claim 3, wherein the gene(s) or gene product(s) are selected from the group consisting of CACNAlA, PDE3B and TUBB4.
5. A method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table
3, wherein the pathogen is not West Nile Virus or Dengue Fever Virus.
6. The method of claim 5, wherein the gene(s) or gene product(s) is ADRAlD.
7. A method of inhibiting infection in a cell by a pathogen comprising decreasing expression or activity of one or more gene(s) or gene product(s) set forth in Table
4, wherein the pathogen is not hepatitis C virus.
8. The method of claim 7, wherein the gene(s) or gene product(s) are selected from the group consisting of STAT and TBXA2R.
9. The method of any of claims 1-8, wherein infection is decreased by decreasing the replication of the pathogen.
10. The method of any of claims 1-9, wherein the pathogen is a respiratory virus.
1 1. The method of claim 10, wherein the respiratory virus is a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, or an adenovirus.
12. The method of claim 1 1, wherein the respiratory virus is selected from the group consisting of influenza virus, parainfluenza virus, adenovirus, rhinovirus and RSV.
13. The method of any of claims 1-9, wherein the pathogen is a gastrointestinal virus.
14. The method of claim 13, wherein the gastrointestinal virus is a picornavirus, a filovirus, an adenovirus, a calicivirus or a reovirus.
15. The method of claim 14, wherein the gastrointestinal virus is selected from the group consisting of: a reovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus and an enterovirus.
16. The method of claim 1 or 2, wherein the pathogen is a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, West Nile virus, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
17. The method of claim 3 or 4, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, a hepatitis E virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, Chikungunya virus or a Dengue fever virus.
18. The method of claim 5 or 6, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis C virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A5 LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies or Chikungunya virus.
19. The method of claim 7 or 8, wherein the pathogen is an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, Epstein Barr Virus, Human Papilloma Virus, CMV5 a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus.
20. The method of any of claims 1-19, wherein expression or activity of the gene or gene product is decreased by contacting the cell with a composition comprising a chemical, a compound, an LNA, an aptamer, a small molecule, a drug, a protein, a cDNA, an antibody, a morpholino, a triple helix molecule, an miRNA, an siRNA, an shRNA, an antisense nucleic acid or a ribozyme.
21. The method of claim 20, wherein decreasing expression comprises decreasing translation of an mRNA encoding a gene product set forth in Table 1 , 2, 3 or 4.
22. The method claim 20, wherein the composition comprises an antisense nucleic acid that specifically hybridizes and decreases expression or activity of the gene product.
23. The method of claim 20, wherein the composition comprises an siRNA that decreases expression or activity of the gene product.
24. The method of claim 20, wherein the composition comprises an antibody that specifically binds to a protein.
25. The method of claims 1-24, wherein the cell is an in vitro, an ex vivo or an in vivo cell.
26. The method of any of claims 1-25, wherein the cell is in a subject.
27. The method of claim 26, wherein an effective amount of a composition that decreases the expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4 is administered to the subject.
28. A method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by two or more respiratory viruses selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus and an adenovirus.
29. The method of claim 28, wherein the two or more respiratory viruses are selected from the group consisting of an influenza virus, a parainfluenza virus, an adenovirus, a rhinovirus and an RSV virus.
30. A method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more gastrointestinal viruses selected from the group consisting of: a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus.
31. The method of claim 30, wherein the two or more gastrointestinal viruses are selected from the group consisting of: a reovirus, an adenovirus, a Norwalk virus, an Ebola virus, a Marburg virus, an adenovirus, a rotavirus, a Dengue fever virus and an enterovirus.
32. A method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4, wherein the composition inhibits infection by one or more pathogens selected from the group consisting of: a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, an adenovirus, and inhibits infection by one or more pathogens selected from the group consisting of: a flavivirus, a filovirus, a calcivirus or a reovirus.
33. A method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , 2, 3 or 4 wherein the composition inhibits infection by three or more pathogens.
34. The method of claim 33, wherein the pathogen is a virus.
35. The method of claim 34, wherein the three or more viruses are selected from the group consisting of: an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, a Hepatitis C virus, Epstein Barr Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus.
36. A method of decreasing infection in a subject comprising administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1, 2, 3 or 4, wherein the composition inhibits infection by an HIV virus and a hepatitis virus.
37. The method of claim 36, wherein the hepatitis virus is hepatitis B or hepatitis C.
38. A method of decreasing an unspecified respiratory infection in a subject comprising: a) diagnosing a subject with an unspecified respiratory infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more respiratory viruses selected from the group consisting of a picornavirus, an orthomyxovirus, a paramyxovirus, a coronavirus, or an adenovirus.
39. A method of decreasing an unspecified gastrointestinal infection in a subject comprising: a) diagnosing a subject with an unspecified gastrointestinal infection; and b) administering to the subject an effective amount of a composition that decreases expression or activity of one or more gene(s) or gene product(s) set forth in Table 1 , Table 2, Table 3 or Table 4, wherein the composition inhibits infection by two or more gastrointestinal viruses selected from the group consisting of a flavivirus, an adenovirus, a filovirus, a calcivirus or a reovirus.
40. A method of identifying a compound that binds to a gene product set forth in Table 1, 2 , 3 or 4 and can decrease infection of a cell by a pathogen comprising: a) contacting a compound with a gene product set forth in Table 1, 2, 3 or
4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by the pathogen.
41. The method of claim 40, further comprising optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection.
42. The method of claim 40-41, wherein the assay is a cell based assay.
43. The method of claim 40-41, wherein the assay is an in vivo assay.
44. A method of identifying an agent that decreases infection of a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1 , 2, 3 or 4; and b)detecting the level and/or activity of the gene product produced by the cellular gene, a decrease or elimination of the gene product and/or gene product activity indicating an agent with antipathogenic activity.
14
45. The method of claim 44, wherein the activity is binding between a gene product set forth in Table 1, 2, 3 or 4 and another cellular protein or binding between a gene product set forth in Table 1, 2, 3 or 4 and a pathogenic (i.e. non-host) protein .
46. A method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; b) contacting the cell with a pathogen, wherein the pathogen is not HIV; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
47. A method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 2; b) contacting the cell with a pathogen, wherein the pathogen is not West Nile Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
48. A method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 3;
— -" b) contacting the cell with a pathogen, wherein the pathogen is not West
Nile Virus or Dengue Fever Virus; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
49. A method of identifying an agent that decreases infection in a cell by a pathogen comprising: a) administering the agent to a cell containing a cellular gene encoding a gene product set forth in Table 4; b) contacting the cell with a pathogen, wherein the pathogen is not Hepatitis C; and c) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection.
50. The method of claims 46-49, further comprising measuring the level of expression and/or activity of the gene product.
51. The method of claim 50, wherein the level of infection is determined by determining the level of replication of the pathogen.
52. A method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 1 ; c) contacting the cell with an infectious pathogen, wherein the pathogen is not HIV; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection. e) associating the agent with decreasing expression or activity of the gene product.
53. A method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 2; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile Virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection. e) associating the agent with decreasing expression or activity of the gene product.
54. A method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 3; c) contacting the cell with an infectious pathogen, wherein the pathogen is not West Nile Virus or Dengue Fever Virus; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection. e) associating the agent with decreasing expression or activity of the gene product.
55. A method of making a compound that decreases infection of a cell by a pathogen, comprising: a) synthesizing a compound; b) administering the compound to a cell containing a cellular gene encoding a gene product set forth in Table 4; c) contacting the cell with an infectious pathogen, wherein the pathogen is not hepatitis C; d) determining the level of infection, a decrease or elimination of infection indicating that the agent is an agent that decreases infection. e) associating the agent with decreasing expression or activity of the gene product.
56. The method of claims 52-55, comprising making the association by measuring the level of expression and/or activity of the gene product.
57. The method of claims 52-55, wherein the level of infection is determined by determining the level of replication of the pathogen.
58. A method of making a compound that decreases infection in a cell by a pathogen, comprising: a) optimizing a compound to bind a gene product set forth in Table 1, 2 , 3 or 4; b) administering the compound to a cell containing a cellular gene encoding the gene product; c) contacting the cell with an infectious pathogen; d) determining the level of infection, a decrease or elimination of infection indicating the making of a compound that decreases infection in a cell by a pathogen.
59. The method of claim 58, wherein the level of infection is determined by determining the level of replication of the pathogen.
60. A method of making a compound that decreases infection in a cell by a pathogen comprising further synthesizing therapeutic quantities of the compound made in claim 58.
61. A method of synthesizing a compound that binds to a gene product set forth in Table 1, 2, 3 or 4 and decreases infection by a pathogen comprising: a) contacting a library of compounds with a gene product set forth in Table 1, 2, 3 or 4; b) associating binding with a decrease in infection and; c) synthesizing derivatives of the compounds from the library that bind to the gene product.
62. A business method to reduce the cost of drug discovery of drugs that can reduce infection by a pathogen comprising: a) screening, outside of the United States, for drugs that reduce infection by binding to or reducing the function of a gene product set forth in Table 1, 2, 3 or 4; and b) importing drugs that reduce infection into the United States.
63. A method of making drugs comprising directing the synthesis of drugs that reduce infection by binding to or reducing the function of a gene or gene product set forth in Table 1, 2, 3 or 4.
64. A method of identifying an agent that can decrease infection by three or more pathogens comprising: a) administering the agent to three or more cell populations containing a cellular gene encoding a gene product set forth in Table 1, 2, 3 or 4; b) contacting the three or more cell populations with a pathogen selected wherein each population is contacted with a different pathogen; and c) determining the level of infection, a decrease or elimination of infection by three or more pathogens indicating that the agent is an agent that decreases infection by three or more pathogens.
65. A method of identifying a compound that binds to a gene product set forth in Table 1 , 2, 3 or 4 and can decrease infection by three or more pathogens comprising a) contacting a compound with a gene product set forth in Table 1 , 2, 3 or 4; b) detecting binding of the compound to the gene product; and c) associating binding with a decrease in infection by three or more pathogens.
66. The method of claim 65, further comprising optimizing a compound that binds the gene product in an assay that determines the functional ability to decrease infection by three or more pathogens.
67. The method of claim 66, wherein the assay is a cell based assay.
68. The method of claim 67, wherein the assay is an in vivo assay.
69. The method of claim 67 or 68, wherein the three or more pathogens are selected from the group consisting of an HIV virus, a pox virus, a herpes virus, an RSV virus, an influenza virus, a hepatitis B virus, a Hepatitis C virus, Epstein Ban- Virus, Human Papilloma Virus, CMV, a rhinovirus, an adenovirus, measles virus, Marburg virus, Ebola virus, Rift Valley Fever Virus, LCM, Junin virus, Machupo virus, Guanarito virus, Lassa Fever virus, Hantavirus, SARS virus, Nipah virus, Caliciviruses, Hepatitis A, LaCrosse, California encephalitis, VEE, EEE5WEE, Japanese Encephalitis Virus, Kyasanur Forest Virus, Yellow Fever, Rabies, a Dengue Fever virus, West Nile virus or Chikungunya virus.
70. A method of decreasing the toxicity of a toxin in a cell comprising administering to the cell an effective amount of a composition that decreases expression or activity of one or more gene(s) or a gene product (s) set forth in Table 1 , 2, 3 or 4.
71. The method of claim 70, wherein the toxin is a ricin toxin, a c. perfringens toxin, Clostridium botulinum toxin, a C. difficile toxin or a staphylococcal toxin.
72. The method of claim 71, wherein the cell is in vitro, ex vivo, or in vivo.
PCT/US2010/000916 2009-03-27 2010-03-26 Mammalian genes involved in infection WO2010110914A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21117809P 2009-03-27 2009-03-27
US61/211,178 2009-03-27

Publications (2)

Publication Number Publication Date
WO2010110914A2 true WO2010110914A2 (en) 2010-09-30
WO2010110914A3 WO2010110914A3 (en) 2011-05-19

Family

ID=42781722

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/000916 WO2010110914A2 (en) 2009-03-27 2010-03-26 Mammalian genes involved in infection

Country Status (1)

Country Link
WO (1) WO2010110914A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR3015986A1 (en) * 2013-12-30 2015-07-03 Lvmh Rech APTAMERS INHIBITING THE ENZYMATIC ACTIVITY OF MMP-9 PROTEIN
WO2015184164A1 (en) * 2014-05-30 2015-12-03 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl peptidase iv (dpp4) animals
WO2019204571A1 (en) * 2018-04-20 2019-10-24 Texas Tech University System Self-attenuated prophylactic and therapeutic vaccines against pathogens
US10898449B2 (en) 2016-12-20 2021-01-26 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US11033512B2 (en) 2017-06-26 2021-06-15 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and silicone acrylic hybrid polymer
CN113444785A (en) * 2021-06-28 2021-09-28 甘肃农业大学 SSc-miR-122-5p related to piglet C-type clostridium perfringens infectious diarrhea and application thereof
WO2021262799A1 (en) * 2020-06-23 2021-12-30 Flagship Pioneering, Inc. Anti-viral compounds and methods of using same
US11337932B2 (en) 2016-12-20 2022-05-24 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and polysiloxane or polyisobutylene
US11648213B2 (en) 2018-06-20 2023-05-16 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070087008A1 (en) * 2004-02-24 2007-04-19 The Government Of The United States Of America As Rab9a, rab11a, and modulators thereof related to infectious disease

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070087008A1 (en) * 2004-02-24 2007-04-19 The Government Of The United States Of America As Rab9a, rab11a, and modulators thereof related to infectious disease

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CAMARGOS, R.S. E. ET AL.: 'Protective role of ETA endothelin receptors during the acute phase of Trypanosoma cruzi infection in rats.' MICROBES AND INFECTION. vol. 6, no. 7, June 2004, pages 650 - 656 *
GRANDER, W. ET AL.: 'Bosentan treatment of portopulmonary hypertension related to liver cirrhosis owing to hepatitis C.' EUROPEAN JOURNAL OF CLINICAL INVESTIGATION. vol. 36, September 2006, pages 67 - 70 *
MARCHANT, D. ET AL.: 'Bosentan enhances viral load via endothelin-1 receptor type-A-mediated p38 mitogen-activated protein kinase activation while improving cardiac function during coxsackievirus-induced myocarditis.' CIRCULATION RESEARCH. vol. 104, February 2009, pages 813 - 821 *
ROFFE, E. ET AL.: 'Endothelin-1 receptors play a minor role in the protection against acute Trypanosoma cruzi infection in mice.' BRAZILIAN JOURNAL OF MEDICAL AND BIOLOGICAL RESEARCH. vol. 40, no. 3, March 2007, pages 391 - 399 *
SITBON, 0. ET AL.: 'Bosentan for the treatment of human immunodeficiency virus-associated pulmonary arterial hypertension.' AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE. vol. 170, August 2004, pages 1212 - 1217 *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015101637A3 (en) * 2013-12-30 2015-08-27 L V M H Recherche Aptamers inhibiting the enzymatic activity of the mmp-9 protein
US9902961B2 (en) 2013-12-30 2018-02-27 Lvmh Recherche Aptamers inhibiting the enzymatic activity of the MMP-9 protein
FR3015986A1 (en) * 2013-12-30 2015-07-03 Lvmh Rech APTAMERS INHIBITING THE ENZYMATIC ACTIVITY OF MMP-9 PROTEIN
US11013220B2 (en) 2014-05-30 2021-05-25 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl-peptidase IV (DPP4) animals
WO2015184164A1 (en) * 2014-05-30 2015-12-03 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl peptidase iv (dpp4) animals
US9497945B2 (en) 2014-05-30 2016-11-22 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl peptidase IV (DPP4) animals
US9532555B2 (en) 2014-05-30 2017-01-03 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl peptidase IV (DPP4) animals
CN106413394A (en) * 2014-05-30 2017-02-15 再生元制药公司 Humanized dipeptidyl peptidase IV (DPP4) animals
US10212923B2 (en) 2014-05-30 2019-02-26 Regeneron Pharmaceuticals, Inc. Humanized dipeptidyl-peptidase IV (DPP4) animals
CN111118047B (en) * 2014-05-30 2023-09-22 再生元制药公司 Humanized dipeptidyl peptidase IV (DPP 4) animals
CN111118047A (en) * 2014-05-30 2020-05-08 再生元制药公司 Humanized dipeptidyl peptidase IV (DPP4) animals
US10898449B2 (en) 2016-12-20 2021-01-26 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US10980753B2 (en) 2016-12-20 2021-04-20 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
US11337932B2 (en) 2016-12-20 2022-05-24 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and polysiloxane or polyisobutylene
US11033512B2 (en) 2017-06-26 2021-06-15 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine and silicone acrylic hybrid polymer
US11389526B2 (en) 2018-04-20 2022-07-19 Texas Tech University System Self-attenuated prophylactic and therapeutic vaccines against pathogens
WO2019204571A1 (en) * 2018-04-20 2019-10-24 Texas Tech University System Self-attenuated prophylactic and therapeutic vaccines against pathogens
US11648213B2 (en) 2018-06-20 2023-05-16 Lts Lohmann Therapie-Systeme Ag Transdermal therapeutic system containing asenapine
WO2021262799A1 (en) * 2020-06-23 2021-12-30 Flagship Pioneering, Inc. Anti-viral compounds and methods of using same
CN113444785A (en) * 2021-06-28 2021-09-28 甘肃农业大学 SSc-miR-122-5p related to piglet C-type clostridium perfringens infectious diarrhea and application thereof
CN113444785B (en) * 2021-06-28 2022-06-21 甘肃农业大学 SSc-miR-122-5p related to piglet C-type clostridium perfringens infectious diarrhea and application thereof

Also Published As

Publication number Publication date
WO2010110914A3 (en) 2011-05-19

Similar Documents

Publication Publication Date Title
WO2011008956A2 (en) Mammalian genes involved in infection
WO2010110914A2 (en) Mammalian genes involved in infection
US10202615B2 (en) Mammalian genes involved in toxicity and infection
WO2011146527A2 (en) Mammalian genes involved in infection
WO2010134939A2 (en) Mammalian genes involved in infection
JP6980762B2 (en) Use of VCP inhibitors and oncolytic viruses in the preparation of antitumor agents
WO2010039778A2 (en) Mammalian genes involved in infection
WO2015085905A1 (en) Non-coding rna of salmonella and identification and use thereof
JP2019531699A (en) Diagnosis and treatment method of cancer by expression state and mutation state of NRF2 and downstream target gene of the gene
RU2674147C2 (en) Methods of treating colorectal cancer
WO2006047673A2 (en) Mammalian genes involved in infection
JP2013520197A (en) Methods and compositions and miRNA inhibitors and targets for detecting and treating cancers associated with miRNA
KR102194746B1 (en) Markers associated with wnt inhibitors
Zheng et al. Circular RNA circ-ABCB10 promotes non-small cell lung cancer proliferation and inhibits cell apoptosis through repressing KISS1.
US8247451B2 (en) ADAM10 and its uses related to infection
US20230149507A1 (en) Treatment of alt cancers
Wang et al. METTL1/WDR4-mediated tRNA m7G modification and mRNA translation control promote oncogenesis and doxorubicin resistance
WO2013066442A2 (en) Mammalian genes and gene products involved in infection
US20130067609A1 (en) Mammalian Genes Involved in Tularemia and Other Infections
WO2012045067A2 (en) Mammalian genes involved in infection
CN113637709B (en) Application of KIF5B gene as target spot in inhibiting foot-and-mouth disease virus replication
WO2015085903A1 (en) Non-coded rna of in-vivo infected microorganisms, parasitic microorganisms, symbiotic microorganisms and identification and application thereof
KR20160135362A (en) Combination of egfr inhibitor and mek inhibitor for use in the treatment of nras mutated cancer
Nau et al. Varenicline prevents SARS-CoV-2 infection in vitro and in rhesus macaques
US20140220690A1 (en) Mammalian genes involved in infection

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10756503

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10756503

Country of ref document: EP

Kind code of ref document: A2