EP1001967A1 - Behandlungsmethode eines lignocellulosischen materials oder eines expandierten minerals zur formung eines fertigproduktes - Google Patents

Behandlungsmethode eines lignocellulosischen materials oder eines expandierten minerals zur formung eines fertigproduktes

Info

Publication number
EP1001967A1
EP1001967A1 EP98935519A EP98935519A EP1001967A1 EP 1001967 A1 EP1001967 A1 EP 1001967A1 EP 98935519 A EP98935519 A EP 98935519A EP 98935519 A EP98935519 A EP 98935519A EP 1001967 A1 EP1001967 A1 EP 1001967A1
Authority
EP
European Patent Office
Prior art keywords
irf
sequence
hsv
reactivation
polynucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98935519A
Other languages
English (en)
French (fr)
Other versions
EP1001967A4 (de
Inventor
Shelley L. Berger
Nigel W. Fraser
Jeffrey J. Leary
Ruth Tal-Singer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
SmithKline Beecham Ltd
Wistar Institute of Anatomy and Biology
Original Assignee
SmithKline Beecham Ltd
Wistar Institute of Anatomy and Biology
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 SmithKline Beecham Ltd, Wistar Institute of Anatomy and Biology filed Critical SmithKline Beecham Ltd
Publication of EP1001967A1 publication Critical patent/EP1001967A1/de
Publication of EP1001967A4 publication Critical patent/EP1001967A4/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • This invention relates to newly identified attributes of cellular and viral polynucleotides and polypeptides, the uses of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides.
  • This invention also relates to inhibiting the biosynthesis, action or interaction of such polynucleotides and/or polypeptides and to the use of such inhibitors in therapy, particularly therapy for viral reactivation, especially Herpesvirus reactivation, as well as for prophylaxis.
  • HSV latent Herpes simplex virus
  • the virus can undergo sporadic reactivation to produce recurrent mucocutaneous lesions at peripheral sites innervated by the infected ganglia (reviewed in Fraser et al., 1991, Roizman, 1991, Stevens, 1989).
  • Reactivation stimuli range from direct mechanical or pharmacological insults to the neuron and surrounding tissue to systemic changes in immune modulators and neurotransmitters (Fraser et al., 1991, Fraser & Valyi-Nagy, 1993, Hill, 1985). The earliest molecular events in neuronal cells that trigger reactivation of HSV remain unclear.
  • the present invention provides cellular factors with a putative role in the reactivation process of Herpesviruses, particularly HSV-1, HSV-2, varicella zoster virus (herein “VZV”), Epstein Barr virus (herein “EBV”) and human cytomegalovirus (herein “HCMV”).
  • This invention was made, in part, using differential display RT-PCR (DDRT- PCR), which allows the visualization and subsequent isolation of cDNAs corresponding to mRNAs displaying altered expression in different cell populations (Liang et al, 1995, Liang & Pardee, 1992). Using this method, levels of gene expression in TG populations derived from various time points following explantation were compared.
  • any factors modulated during the first 4 hours, the period in which viral gene expression is first detected are believed to be important in the initial stimulation of latent viral genomes.
  • DDRT-PCR has also been used to identify cellular genes modulated by SV40 and EBV transformation (Sompayrac et al, 1996, Yan et al, 1996). However, it is a novel approach to study viral reactivation.
  • This invention demonstrates the isolation of forty eight differentially-displayed cDNAs representing transcripts whose levels were altered within the first 4 hours following explantation of latently infected TG.
  • Five cDNAs were identical to murine TIS7, whose sequence has been shown to be related to interferons (IFNs) (Skup et al, 1982, Tirone & Shooter, 1989, Varnum et al, 1989). Rapid induction of IFNs ⁇ and ⁇ in neuronal cells of TG explants was also detected.
  • IFN-1 Interferon Regulatory Factor -1
  • TNF- ⁇ tumor necrosis factor -a
  • Figure 1 shows PCR differential-display of cDNA derived from latently-infected mouse TG following explantation. Autoradiograph of radiolabeled DDRT-PCR products. Arrows denote PCR products representing (Left Panel) band #64 amplified with 3' primer #2 and 5' primer #7, (Right Panel) band #56 amplified with 3' primer #2 and 5' primer #3.
  • Figure 2 shows sequences of DDRT-PCR products. cDNAs isolated from differential-display gels were reamplified using primers that included the T7 promoter sequence, and PCR products sequenced. Sequences were analyzed by BLAST searches.
  • Figure 3 shows confirmation of differential display.
  • RNA was prepared from uninfected TG explants at 0, 1, 2, and 4 h p.e.
  • Complementary DNA from differentially displayed band #56 was reamplified by PCR using 3' primers that included the T7 promoter. PCR products were used as templates to prepare riboprobes labeled with 32" P- UTP, and added to each RNA sample. Following hybridization at 37°C and RNase digestion, samples were separated by PAGE.
  • P The input probe (P) and protected fragments were visualized using phosphorimager screens.
  • Panel B The intensity of each protected fragment was quantitated using Imagequant software. Fold induction was expressed as the ratio between each band to the 0 time point.
  • RT-PCR was used to detect (Panel A) IFN- ⁇ , (Panel B) IRF-1, (Panel C) IFN ⁇ R, (Panel D) IRF-2, and each was compared to the level of cyclophilin mRNA.
  • Duplicate samples of TG explant RNA from 0, 1, 2, and 4 h post- explantation were analysed. Products were separated by agarose gel electrophoresis, followed by fluorimager scanning and analysis using Imagequant software. The relative amount of cDNA is expressed in these graphs in arbitrary units representing the ratio between the intensity of the PCR-product band to the intensity of cyclophilin. The ratio at the 0 time point is designated as 1.
  • L indicates latent.
  • Figure 5 shows immunostaining of interferon protein in TG following.
  • Latently- infected or uninfected BALB/c mice were sacrificed, TG were excised, and incubated in culture media for 0-24 h p.e.
  • Paraffin-embedded sections were processed as described in Methods and reacted with rabbit polyclonal antisera against IFN ⁇ and ⁇ .
  • E 0 h p.e. of uninfected TG,
  • F 4 h p.e. of uninfected TG.
  • FIG. 6 shows RT-PCR detection of TNF- ⁇ , cyclophilin, and ⁇ -actin transcripts in murine TG cultured for varying times post explantation.
  • RNA from latently-infected TG explants was prepared and analyzed by RT-PCR for (Panel A) TNF- ⁇ , and (Panel B) ⁇ - actin, and cyclophilin, as described in Materials and Methods. Products were visualized by ethidium bromide staining as shown in the inserts.
  • the graphs represent the ratio between the PCR product band and the cyclophilin band. The ratio at the time of explantation (0) was determined as 1. Experiments were done in duplicate in four separate experiments.
  • Figure 7 shows the locations and sequences of IRF-1 consensus binding sites in the genome of HSV- 1 , with reference to the nucleotide numbering system of GENEMBL entry HE1CG.
  • FIG 8 shows that IRF-1 binds specifically to HSV-1 LAT DNA consensus sequence.
  • IRF-1 binds specifically to HSV-1 LAT DNA consensus sequence.
  • In vitro translated human IRF-1 was incubated with 32-p_ ⁇ a b e ] e( j LAT probe in the presence or absence of antisera specific for IRF- 1 or IRF-2.
  • Samples were analyzed by electromobility shift assay (EMS A). Probe alone was used as control.
  • Figure 9 shows that IRF-2 binds specifically to LAT DNA. Competition experiments were carried out with mutant LAT oligonucleotides.
  • FIG. 10 shows that TIS7 and IRF-1 are induced by hyperthermia measured by heat shock protein 70 (HSP70) induction.
  • HSP70 heat shock protein 70
  • This invention provides cellular proteins from murine trigeminal ganglia induced by the stress of injury in viral infection and/or reactivation.
  • polynucleotides which bind such polypeptides, or which alter the biological activity of such polypeptides.
  • the invention provides polynucleotides having the DNA sequences given herein.
  • the invention also relates to novel oligonucleotides derived from the sequences given herein which can act as PCR primers in the process herein described to determine, for exaple, whether or not the genes identified herein in whole or in part are expressed in an infected tissue or individual. It is recognised that such sequences will also have utility in diagnosis of the stage of infection and type of infection the pathogen has attained.
  • the proteins so identified are also useful as targets in screens designed to identify antiviral compounds.
  • a polypeptide and/or polynucleotide of the invention for therapeutic or prophylactic purposes, for example, as an antiviral agent or a vaccine.
  • polypeptides and/or polypeptide of the invention for therapeutic or prophylactic purposes.
  • inhibitors or activators to such polypeptides and/or polynucleotides useful as antiviral agents.
  • antibodies against such polypeptides and polypeptide-polynucleotide complexes are provided.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the above polypeptide, polynucleotide, inhibitor or activator of the invention and a pharmaceutically acceptable carrier.
  • the invention provides the use of an activator or inhibitor of the invention as an antiviral agent.
  • the invention further relates to the manufacture of a medicament for such uses, particularly a medicament to treat HSV-1 and/or HSV-2 infection or reactivation.
  • composition comprising IRF-1, particularly used as a screening target for the development of prophylactic and therapeutic compounds, such as compounds that interfere with HSV (herein "HSV” means HSV-1 and or HSV-2) or VZV reactivation.
  • HSV HSV-1 and or HSV-2
  • VZV reactivation a method for inhibiting the binding of IRF-1 to a polynucleotide, such as a DNA element (for example, ISRE's), particulary DNA comprising or near a viral origins of replication.
  • a polynucleotide such as a DNA element (for example, ISRE's), particulary DNA comprising or near a viral origins of replication.
  • a further aspect of the invention is a composition comprising an IRF- 1 : IRF-BP complex.
  • reactivation means viral reactivation from latency
  • compositions comprising IRF- 1 binding site consensus sequences in HSV oriL and oriS and HSV long terminal repeats.
  • Vector(s) of the invention refers, among others, to a polypeptide comprising the amino acid sequence any or a combination of the factors involved in viral infection and/or reactivation disclosed herein, such as IRF-1, TIS7, IFN- ⁇ and IFN- ⁇ , or an allelic variant thereof; also included are polynucleotides encoding any of these polypeptides.
  • Biological Activity of the Receptor refers to the metabolic or physiologic function of said Factors of the invention including similar activities or improved activities or these activities with decreased undesirable side-effects. Also included are antigenic and immunogenic activities of said Factors of the invention.
  • Factor gene(s) refers to a polynucleotide comprising the nucleotide sequence encoding a Factor of the invention or allelic variants thereof and/or their complements.
  • Antibodies as used herein includes polyclonal and monoclonal antibodies, chimeric, single chain, and humanized antibodies, as well as Fab fragments, including the products of an Fab or other immunoglobulin expression library. "Isolated” means altered “by the hand of man” from the natural state. If an
  • isolated composition or substance occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living animal is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated”, as the term is employed herein.
  • Polynucleotide generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • Polynucleotides include, without limitation single- and double- stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the term polynucleotide also includes DNAs or RNAs containing one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons.
  • Modified bases include, for example, tritylated bases and unusual bases such as inosine.
  • polynucleotide embraces chemically, enzymatically or metabolically modified forms of polynucleotides as typically found in nature, as well as the chemical forms of DNA and RNA characteristic of viruses and cells.
  • Polynucleotide also embraces relatively short polynucleotides, often referred to as oligonucleotides.
  • Polypeptide refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres.
  • Polypeptide refers to both short chains, commonly referred to as peptides, oligopeptides or oligomers, and to longer chains, generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids.
  • Polypeptides include amino acid sequences modified either by natural processes, such as posttranslational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
  • Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP- ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • Variant is a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide respectively, but retains essential properties.
  • a typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide.
  • Changes in the nucleotide sequence of the variant may or may not alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
  • a typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions in any combination.
  • a substituted or inserted amino acid residue may or may not be one encoded by the genetic code.
  • a variant of a polynucleotide or polypeptide may be a naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally.
  • Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
  • Identity is a measure of the identity of nucleotide sequences or amino acid sequences. In general, the sequences are aligned so that the highest order match is obtained. “Identity” per se has an art-recognized meaning and can be calculated using published techniques.
  • identity is well known to skilled artisans (Carillo, H., and Lipton, D., SIAM J Applied Math (1988) 48: 1073). Methods commonly employed to determine identity or similarity between two sequences include, but are not limited to, those disclosed in Guide to Huge Computers, Martin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo, H., and Lipton, D., SIAM J Applied Math (1988) 48: 1073. Methods to determine identity and similarity are codified in computer programs.
  • Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, GCG program package (Devereux, J., et al, Nucleic Acids Research (1984) 12(1):387), BLASTP, BLASTN, FASTA (Atschul, S.F. et al, J Molec Biol (1990) 215:403).
  • a polynucleotide having a nucleotide sequence having at least, for example, 95% "identity" to a reference nucleotide sequence of SEQ ID NO: 1 is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence of SEQ ID NO: 1.
  • a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • These mutations of the reference sequence may occur at the 5 or 3 terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • a polypeptide having an amino acid sequence having at least, for example, 95% "identity" to a reference amino acid sequence of is intended that the amino acid sequence of the polypeptide is identical to the reference sequence except that the polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the reference amino acid sequence.
  • up to 5% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 5% of the total amino acid residues in the reference sequence may be inserted into the reference sequence.
  • alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • “Individual(s)” means a eukaryote, particularly a mammal, and especially a human, particularly one infected with a virus or believed to be infected by a virus.
  • Plasmids are designated by a lower case p preceded and/or followed by capital letters and/or numbers.
  • the starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures.
  • equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
  • “Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA.
  • the various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan.
  • For analytical purposes typically 1 ⁇ g of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 ⁇ l of buffer solution.
  • For the purpose of isolating DNA fragments for plasmid construction typically 5 to 50 ⁇ g of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37 C are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylamide gel to isolate the desired fragment.
  • Size separation of the cleaved fragments is performed using 8 percent polyacrylamide gel described by Goeddel, D. et al, (1980) Nucleic Acids Res., 8:4057.
  • Oligonucleotides refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized. Such synthetic oligonucleotides have no 5' phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated. "Ligation” refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al, supra., p.
  • ligation may be accomplished using known buffers and conditions with 10 units to T4 DNA ligase ("ligase”) per 0.5 ⁇ g of approximately equimolar amounts of the DNA fragments to be ligated.
  • ligase DNA ligase
  • a "replicon” is any genetic element (e.g., plasmid, chromosome, virus) that functions as an autonomous unit of DNA replication in vivo; i.e., capable of replication under its own control.
  • a “vector” is a replicon, such as a plasmid, phage, or cosmid, to which another DNA segment may be attached so as to bring about the replication of the attached segment.
  • a “double-stranded DNA molecule” refers to the polymeric form of deoxyribonucleotides (bases adenine, guanine, thymine, or cytosine) in a double-stranded helix, both relaxed and supercoiled. This term refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms.
  • this term includes double-stranded DNA found, inter alia, in linear DNA molecules (e.g., restriction fragments), viruses, plasmids, and chromosomes.
  • sequences may be described herein according to the normal convention of giving only the sequence in the 5' to 3' direction along the nontranscribed strand of DNA (i.e., the strand having the sequence homologous to the mRNA).
  • a DNA "coding sequence of” or a "nucleotide sequence encoding" a particular protein is a DNA sequence which is transcribed and translated into a polypeptide when placed under the control of appropriate regulatory sequences.
  • a “promoter sequence” is a DNA regulatory region capable of binding RNA polymerase in a cell and initiating transcription of a downstream (3' direction) coding sequence.
  • the promoter sequence is bound at the 3' terminus by a translation start codon (e.g., ATG) of a coding sequence and extends upstream (5' direction) to include the minimum number of bases or elements necessary to initiate transcription at levels detectable above background.
  • a transcription initiation site (conveniently defined by mapping with nuclease SI), as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase.
  • Eukaryotic promoters will often, but not always, contain "TATA” boxes and “CAT” boxes.
  • Prokaryotic promoters contain Shine-Dalgarno sequences in addition to the -10 and -35 consensus sequences.
  • DNA "control sequences” refers collectively to promoter sequences, ribosome binding sites, polyadenylation signals, transcription termination sequences, upstream regulatory domains, enhancers, and the like, which collectively provide for the expression (i.e., the transcription and translation) of a coding sequence in a host cell.
  • a control sequence "directs the expression" of a coding sequence in a cell when RNA polymerase will bind the promoter sequence and transcribe the coding sequence into mRNA, which is then translated into the polypeptide encoded by the coding sequence.
  • a "host cell” is a cell which has been transformed or transfected, or is capable of transformation or transfection by an exogenous DNA sequence.
  • a cell has been "transformed" by exogenous DNA when such exogenous DNA has been introduced inside the cell membrane.
  • Exogenous DNA may or may not be integrated (covalently linked) into chromosomal DNA making up the genome of the cell.
  • the exogenous DNA may be maintained on an episomal element, such as a plasmid.
  • a stably transformed or transfected cell is one in which the exogenous DNA has become integrated into the chromosome so that it is inherited by daughter cells through chromosome replication. This stability is demonstrated by the ability of the eukaryotic cell to establish cell lines or clones comprised of a population of daughter cell containing the exogenous DNA.
  • a “clone” is a population of cells derived from a single cell or common ancestor by mitosis.
  • a “cell line” is a clone of a primary cell that is capable of stable growth in vitro for many generations.
  • a "heterologous" region of a DNA construct is an identifiable segment of DNA within or attached to another DNA molecule that is not found in association with the other molecule in nature.
  • DETAILED DESCRIPTION OF THE INVENTION Applicants have discovered that cellular interferon-related genes are induced in tissues in response to certain stimuli as described herein.
  • the tissue studied is primarily nerve tissue (ganglia) where HSV and other Herpesviruses establish latent infections. These stimuli also induce reactivation of HSV and other Herpesviruses from latency into active replication.
  • IRF-1 an important cellular regulatory protein
  • HSV and other Herpesviruses replication through the identification of IRF-1 binding sites in the DNA sequence of HSV and other Herpesviruses DNA.
  • IRF-1 and/or the IRF-1 regulatory pathway, are essential for reactivation of HSV and other Herpesviruses from latency, and that pharmacological interference with this pathway will interfere with HSV and other Herpesvirus reactivation resulting in arrest of clinical disease.
  • the invention provides various targets for screening for therapeutic compounds using the methods described herein.
  • targets include, for example, IRF-1.
  • a compound interfering with HSV and other Herpesvirus reactivation by inhibiting the binding of IRF-1 to viral DNA elements (ISRE's) may be found using such screens and are useful in antiviral therapy, especially against HSV and other Herpesviruses.
  • IRF-1 IRF-BP complex
  • IRF-1 and IRF-BP can be used to screen for compounds interfering with HSV reactivation by, among other modes of action, freezing or mimicking the binding of IRF-1 to it's inhibitor factor IRF-BP.
  • Such compounds are useful in antiviral therapy, especially against HSV.
  • target include any element in the IRF-1 and interferon genetic regulatory pathway which subsequently impacts HSV reactivation from latency. These compounds are useful to screen for antiviral compounds. Such compounds are useful in antiviral therapy, especially against HSV.
  • Yet another target are polynucleotides derived from or comrpising an IRF-1 binding site consensus sequences in HSV LAT/ICPO, oriL and oriS play a significant role in HSV replication. A preferred embodiment of this sequence is set forth in SEQ ID NO: 1. Such compounds are useful in antiviral therpy, especially against HSV.
  • DDRT differential display RT-PCR
  • IFN-related gene transcripts including IFN- ⁇ , and the interferon regulatory factor- 1 (IRF-1) were induced following explantation of murine trigeminal ganglia (Tal-Singer et al. 1998).
  • IRF-1 interferon regulatory factor-1
  • IRF-1 is a transcriptional activator and tumor supressor protein which regulates interferon genes transcription such as IL-1 beta (via Interleukin Converting Enzyme (ICE) pathways) (Tamura et al, 1996), and MHC-I expression (Drew et al, 1995, Hobart et al, 1997). It is activated by interferons and the STATl pathway (Pine et al, 1994). It binds to cellular proteins such as TFIIB, and NFKB (Drew et al, 1995, Wang et al, 1996). Its DNA binding and transcriptional activation functions are inhibited when it is bound to IRF-BP protein (Kondo, 1996). Its known functions involve the inflammatory response and regulation of the cell cycle via induction of p21 (Tanaka et al, 1996).
  • ICE Interleukin Converting Enzyme
  • Targets include any element in the IRF-1 and interferon genetic regulatory pathway which subsequently impacts HSV reactivation from latency. These compounds are useful to screen for antiviral compounds. Such compounds are useful in antiviral therapy, especially against HSV.
  • Yet another target are polynucleotides derived from or comrpising an IRF-1 binding site consensus sequences in HSV LAT/ICPO, oriL and oriS which play a significant role in HSV replication.
  • a preferred embodiment of this sequence is set forth in SEQ ID NO:l.
  • Such compounds are useful in antiviral therpy, especially against HSV.
  • murine trigeminal ganglia explants were analyzed by differential display RT-PCR (DDRT).
  • IRF-1 is a transcriptional activator and tumor supressor protein which regulates interferon genes transcription such as IL- 1 beta (via Interleukin Converting Enzyme (ICE) pathways) (Tamura et al, 1996), and MHC-I expression (Drew et al, 1995, Hobart et al, 1997)]. It is activated by interferons and the STATl pathway (Pine et al, 1994). It binds to cellular proteins such as TFIIB, and NFkB (Drew et al, 1995, Wang et al, 1996). Its DNA binding and transcriptional activation functions are inhibited when it is bound to IRF-BP protein (Kondo, 1996). Its known functions involve the inflammatory response and regulation of the cell cycle via induction of p21 (Tanaka et al, 1996).
  • ICE Interleukin Converting Enzyme
  • IRF-1 is a member of a multigene family which recognizes the same promoter consensus sequence (herein referred to as "ISRE” or "ICS"). Sequence analysis using Findpatterns function of the GCG software (Genetics Computer Group, Madison, Wisconsin). HSV-1 complete genome sequence (Genbank locus HE1CG accession # X14112) was searched for the core consensus IRF-1 site in interferon promoters using the hexamer AAGTGA (Tanaka & Taniguchi, 1992). Sequence matches are listed in Table 1 by sequence location, and gene name. Ten identical matches were found in in HSV-1 strain 17+ sequence (see Figure 7).
  • Binding Site HI is within regions previously identified as origin binding protein (OBP or UL9) Binding Site HI. Also, of great interest are previous observations that, in addition to binding OBP, Binding Site HI interacts with yet unknown cellular proteins (Dabrowski et al., 1991; Dabrowski et al, 1994). Taken together, these observations suggest that IRF-1 may bind to regulatory elements in the viral genome such as the origins of replication, and ICPO/LAT, and thereby upregulate viral replication or gene expression. Interestingly, functional ERF binding sites recently were identified in the herpesvirus Epstein-Barr virus (EBV) (Schaffer et al, 1997; Nonkwelo et al, 1997).
  • EBV herpesvirus Epstein-Barr virus
  • IRF-1 and IRF-2 bind directly to consensus sequence sites in the EBV Type I latency promoter of EBNA-1 (Qp), and IRF factors may play a primary role in transcriptional regulation of EBNA-1 in cell culture (Schaffer et al, 1997; Nonkwelo et al, 1997).
  • IRF-1 consensus sequences include, for example, the following sequences in Table 1. Highly conserved nucleotides surrounding the core are bold and underlined.
  • Epstein Barr Virus Ebv.Gb_Vi 1 AAGTGA
  • Applicants have also used, for example, DDRT-PCR to identify genes differentially expressed following the stress caused by explantation of latently-infected murine TG.
  • Mice were infected by the corneal route with HSV-1, and at 4 weeks post infection, mice were sacrificed and the TG were explanted.
  • RNA was prepared from TG at various times following explantation, followed by DDRT-PCR.
  • TIS7 is induced by the stress of explantation. Five differentially displayed bands were identified as overlapping regions of murine TIS7 (Lim et al, 1987, Varnum et al, 1989) ( Figure 2) and the results were confirmed by quantitative RPA and RT-PCR ( Figure 3).
  • the TIS (TPA-inducible sequences) family members are early response genes (Walz et al, 1976) which are induced rapidly and transiently in Swiss 3T3 cells by the tumor promoter mitogen tetradecanoyl phorbol acetate (TPA) (Lim et al, 1987), or by serum (Herschman, 1991).
  • TIS genes also have been identified in rat PC 12 cells, following induction with NGF, TPA, epidermal growth factor and depolarization (Kujubu et al, 1987). Moreover, TIS induction has been detected in primary astrocyte cultures following mitogen induction (Arenalder et al, 1989). Thus, a family of ⁇ S genes appear to constitute a common pathway or response to many cell stimulatory agents or physical stimuli.
  • TIS7/PC4 genes The pattern of induction previously observed for TIS7/PC4 genes in these systems revealed an increase in the levels of RNA or protein between 2-4 hours post stimulus (Varnum et al, 1994), similar to our observation in the mouse explant model ( Figure 2).
  • Applicants have shown that another TIS transcript, TIS28 or c-fos, was induced rapidly following explantation (Tal-Singer et al, 1997, Valyi-Nagy et al, 1991), and again, the kinetics match those previously observed following mitogen induction (Arenalder et al, 1989).
  • rat PC4 highly related to murine TIS7
  • c-fos are both induced by activation of the oncogenenic protein-tyrosine kinase v-fps, encoded in the retrovirus Fujinami sarcoma virus (Jahner & Hunter, 1991). It would be of interest to determine whether a similar kinase, either of viral or cellular origin, is activated in explanted ganglia or in other HSV reactivation systems.
  • Interferon ⁇ is induced by the stress of explantation.
  • TIS7 was originally identified as a gene induced in murine 3T3 cells following infection with Newcastle disease virus (Skup et al, 1982). Nucleotide sequence analysis revealed some conservation with human IFN- ⁇ and rat IFN- ⁇ (Tirone & Shooter, 1989). The present invention provides that, like TIS7, IFN- ⁇ and IFN- ⁇ are induced in neuronal cells within the first hour following explantation.
  • IFN- ⁇ expression is modulated at the transcriptional level by multiple regulatory factors that bind upstream of the initiation site, such as the activators IRF-1 and NFkB, and the repressor IRF-2 (reviewed in Tanaka & Taniguchi, 1992).
  • the activators IRF-1 and NFkB and the repressor IRF-2 (reviewed in Tanaka & Taniguchi, 1992).
  • IRF- 1 was induced by the stress of explantation.
  • IRF-2 nor the IFN receptor were induced ( Figure 4). Since the induction of IRF-1 followed the same temporal pattern as that of IFN, IFN- alpha and beta are believed to be induced via an IRF-1 dependent pathway in explanted TG cells (Kawakami et al, 1995).
  • IFN induction is a consequence of the stress of explantation, and did not result from viral gene expression, or from incubation in the presence of serum factors.
  • IFN is known to possess antiviral properties, and indeed is induced by HSV infection (Gobi et al, 1988a, Green et al, 1981, Stanwick et al, 1982b).
  • IFN is an inhibitor of activation of HSV immediate early (IE) genes in vitro by VP16 (LaMarco & Mc Knight, 1989).
  • IE immediate early
  • Applicants recently found induction of both Early and IE HSV genes during the first hours of viral reactivation (Tal- Singer et al, 1997).
  • the absence of viral IE gene expression prior to Early gene expression during the first hours following explantation of TG may be a result of the inhibitory effect of IFN on a neuron-specific VP16 homologue.
  • IFN blocks both HSV morphogenesis and release of viral particles from infected cells (Chatterjee et al, 1985). Therefore, an interesting relationship between IFN and reactivating HSV may involve one round of viral replication, allowing the virus to travel from TG neurons to the site of recrudescence in corneal epithelium. Immediate induction of IFN may prevent the spread of reactivating virus within the nervous system by inhibiting release of viral particles and activating host defense mechanisms such as natural killer cells (Habu et al, 1984). Moreover, in another virus system, neuroblastoma cells expressing high levels of IFN-b support persistent rabies virus infections (Honda et al, 1985). This suggests that the IFN response may be involved in ensuring the viability of infected host cells.
  • IFN may inhibit viral reactivation in neurons and, in a few cells, other cellular factors override its effects by inducing high levels of viral gene expression.
  • Walev et al. Wang et al, 1995
  • TNF-a treatment induced the efficiency of reactivation in that study.
  • TIS7 The relationship of TIS7, IFN, and viral responses. TIS7 and interferons are induced by viral infection (Skup et al, 1982, Tanaka & Taniguchi, 1992) and, as shown in this study, by the stress of explantation. The precise function of TIS7 in the cell is yet to be elucidated, it is however clear that it plays a role in cellular growth and differentiation (Guardavaccaro et al, 1995, Herschman et al, 1994). Furthermore, it is known that TIS7 has no antiviral activity (Tirone & Shooter, 1989) which is a property of interferons (Tanaka & Taniguchi, 1992).
  • PCR differential- display represents an excellent method to screen for species of RNA which are transcriptionally regulated in TG following explantation. Genes identified using this method can be studied in other HSV reactivation systems, resulting in a database of specific genes which may be involved in the reactivation process.
  • TIS7 induction was shown in neurons at the protein level, using immunohistochemistry of TG sections.
  • IRF- 1 binding in the viral genome Figure 7 (there were significant matches to the IRF-1 consensus binding site). The disclosure of these sites in no way limits the manner in which cellular factors interact with virus sequences during reactivation. Applicants have also demonstrated that IRF- 1 binding sites occur in potentially critical regions of the viral genome, and thus are believed to be crucial in the reactivation process.
  • Identified herein are cellular genes induced in response to stimuli that reactivate virus and methods for screening compounds to treat disease based on this observation. Therapies based on this observation are also provided.
  • the invention provides that these genes are potential causative agents in reactivation, but this model in no way limit the scope of the invention.
  • any polynucleotide of the present invention may be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA.
  • the DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
  • the coding sequence which encodes the polypeptide may be identical to the coding sequence shown or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encoding the same polypeptide.
  • the present invention includes variants of the hereinabove described polynucleotides which encode fragments, analogues and derivatives of the polypeptide characterized by the deduced amino acid sequence given herein.
  • the variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non- naturally occurring variant of the polynucleotide.
  • the present invention includes polynucleotides encoding the same polypeptide characterized by the deduced amino acid sequence given herein as well as variants of such polynucleotides which variants encode for a fragment, derivative or analogue of the polypeptide.
  • Such nucleotide variants include deletion variants, substitution variants and addition or insertion variants.
  • the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence characterized by the DNA sequence disclosed herein.
  • allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide.
  • the polynucleotide which encodes for the mature polypeptide may include only the coding sequence for the mature polypeptide or the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotein sequence.
  • polynucleotide encoding a polypeptide encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
  • the present invention therefore includes polynucleotides, wherein the coding sequence for the mature polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell.
  • the polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide.
  • the polynucleotides may also encode for a proprotein which is the mature protein plus additional 5' amino acid residues.
  • a mature protein having a prosequence is a proprotein and may be an inactive form of the protein.
  • the polynucleotide of the present invention may code for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence).
  • leader sequence the amino acid sequences provided herein show a methionine residue at the NH 2 -terminus. It is appreciated, however, that during post-translational modification of the peptide, this residue may be deleted. Accordingly, this invention contemplates the use of both the methionine-containing and the methionineless amino terminal variants of each protein disclosed herein.
  • the polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence at either the 5 ' or 3' terminus of the gene which allows for purification of the polypeptide of the present invention.
  • the marker sequence may be a hexa-histidine tag supplied by the pQE series of vectors (supplied commercially by Quiagen Inc.) to provide for purification of the polypeptide fused to the marker in the case of a bacterial host.
  • the present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 50% and preferably at least 70% identity between the sequences.
  • the present invention particularly relates to Streptococcal polynucleotides which hybridize under stringent conditions to the hereinabove-described polynucleotides .
  • stringent conditions means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
  • the polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which retain substantially the same biological function or activity as the polypeptide characterised by the deduced amino acid sequence given herein.
  • fragment when referring to the polypeptide characterized by the deduced amino acid sequence herein, means a polypeptide which retains essentially the same biological function or activity as such polypeptide.
  • an analogue includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.
  • the polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
  • the fragment, derivative or analogue of the polypeptide characterized by the deduced amino acid sequence herein may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the polypeptid
  • polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated.
  • Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • the present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
  • polypeptide of the invention by recombinant techniques by expressing a polynucleotide encoding said polypeptide in a host and recovering the expressed product.
  • polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector.
  • the vector may be, for example, in the form of a plasmid, a cosmid, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the genes.
  • the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • Suitable expression vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA.
  • any other vector may be used as long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by procedures known in the art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoters there may be mentioned: LTR or SV40 promoter, the E. coli. lac or trp, the phage lambda PL promoter and other promoters known to control expression of genes in eukaryotic or prokaryotic cells or their viruses.
  • the expression vector also contains a ribosome binding site for translation initiation and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
  • the gene can be placed under the control of a promoter, ribosome binding site (for bacterial expression) and, optionally, an operator (collectively referred to herein as "control" elements), so that the DNA sequence encoding the desired protein is transcribed into RNA in the host cell transformed by a vector containing this expression construction.
  • the coding sequence may or may not contain a signal peptide or leader sequence.
  • the polypeptides of the present invention can be expressed using, for example, the E. coli tac promoter or the protein A gene (spa) promoter and signal sequence. Leader sequences can be removed by the bacterial host in post-translational processing. See, e.g., U.S. Patent Nos. 4,431,739; 4,425,437; 4,338,397.
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers.
  • Two appropriate vectors are PKK232-8 and PCM7.
  • Particular named bacterial promoters include lad, lacZ, T3, T7, gpt, lambda PR, PL and trp.
  • Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • regulatory sequences which allow for regulation of the expression of the protein sequences relative to the growth of the host cell.
  • Regulatory sequences are known to those of skill in the art, and examples include those which cause the expression of a gene to be turned on or off in response to a chemical or physical stimulus, including the presence of a regulatory compound.
  • Other types of regulatory elements may also be present in the vector, for example, enhancer sequences.
  • An expression vector is constructed so that the particular coding sequence is located in the vector with the appropriate regulatory sequences, the positioning and orientation of the coding sequence with respect to the control sequences being such that the coding sequence is transcribed under the "control" of the control sequences (i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence).
  • control i.e., RNA polymerase which binds to the DNA molecule at the control sequences transcribes the coding sequence.
  • Modification of the coding sequences may be desirable to achieve this end. For example, in some cases it may be necessary to modify the sequence so that it may be attached to the control sequences with the appropriate orientation; i.e., to maintain the reading frame.
  • control sequences and other regulatory sequences may be ligated to the coding sequence prior to insertion into a vector, such as the cloning vectors described above.
  • the coding sequence can be cloned directly into an expression vector which already contains the control sequences and an appropriate restriction site.
  • recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly- expressed gene to direct transcription of a downstream structural sequence.
  • the heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium.
  • the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • the vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence may be employed to transform an appropriate host to permit the host to express the protein.
  • the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • a promoter operably linked to the sequence.
  • Bacterial pET-3 vectors (Stratagene), pQE70, pQE60, pQE-9 (Qiagen), pbs, pDIO, phagescript, psiX174, pbluescript SK, pbsks, pNH8A, pNHl ⁇ a, pNH18A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia).
  • Eukaryotic pBlueBacIII (Invitrogen), pWLNEO, " pSV2CAT, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia).
  • any other plasmid or vector may be used as long as they are replicable and viable in the host.
  • Examples of recombinant DNA vectors for cloning and host cells which they can transform include the bacteriophage 1 (E. coli), pBR322 (E. coli), pACYC177 (E. coli), pKT230 (gram-negative bacteria), pGV1106 (gram-negative bacteria), pLAFRl (gram- negative bacteria), pME290 (non-£. coli gram-negative bacteria), pHV14 (E.
  • Polypeptides can be expressed in host cells under the control of appropriate promoters.
  • Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
  • Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al, Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), the disclosure of which is hereby incorporated by reference.
  • the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well known to those skilled in the art.
  • the polypeptide of the present invention may be produced by growing host cells transformed by an expression vector described above under conditions whereby the polypeptide of interest is expressed. The polypeptide is then isolated from the host cells and purified. If the expression system secretes the polypeptide into growth media, the polypeptide can be purified directly from the media. If the polypeptide is not secreted, it is isolated from cell lysates or recovered from the cell membrane fraction. Where the polypeptide is localized to the cell surface, whole cells or isolated membranes can be used as an assayable source of the desired gene product. Polypeptide expressed in bacterial hosts such as E. coli may require isolation from inclusion bodies and refolding.
  • polypeptide can be recovered and purified from recombinant cell cultures by methods including ammonium sulphate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue.
  • the polypeptide may be used as an antigen for vaccination of a host to produce specific antibodies which have anti-bacterial action. This invention also contemplates the use of the DNA encoding the antigen as a component in a DNA vaccine as discussed more fully below.
  • the polypeptides or cells expressing them can be used as an immunogen to produce antibodies thereto.
  • These antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the term antibodies also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
  • Antibodies generated against the polypeptides of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
  • Polypeptide derivatives include antigenically or immunologically equivalent derivatives which form a particular aspect of this invention.
  • 'antigenically equivalent derivative' encompasses a polypeptide or its equivalent which will be specifically recognised by certain antibodies which, when raised to the protein or polypeptide according to the present invention, interfere with the interaction between pathogen and mammalian host.
  • 'immunologically equivalent derivative' as used herein encompasses a peptide or its equivalent which when used in a suitable formulation to raise antibodies in a vertebrate, the antibodies act to interfere with the interaction between pathogen and mammalian host.
  • derivatives which are slightly longer or slightly shorter than the native protein or polypeptide fragment of the present invention may be used.
  • polypeptides in which one or more of the amino acid residues are modified may be used.
  • Such peptides may, for example, be prepared by substitution, addition, or rearrangement of amino acids or by chemical modification thereof. All such substitutions and modifications are generally well known to those skilled in the art of peptide chemistry.
  • the polypeptide such as an antigenically or immunologically equivalent derivative or a fusion protein thereof is used as an antigen to immunize a mouse or other animal such as a rat or chicken.
  • the fusion protein may provide stability to the polypeptide.
  • the antigen may be associated, for example by conjugation , with an immunogenic carrier protein for example bovine serum albumin (BSA) or keyhole limpet haemocyanin (KLH).
  • BSA bovine serum albumin
  • KLH keyhole limpet haemocyanin
  • a multiple antigenic peptide comprising multiple copies of the protein or polypeptide, or an antigenically or immunologically equivalent polypeptide thereof may be sufficiently antigenic to improve immunogenicity so as to obviate the use of a carrier.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, Nature. 256:495-497(1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72(1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
  • the hybridomas are screened to select a cell line with high binding affinity and favorable cross reaction with other Streptococcal species using one or more of the original polypeptide and/or the fusion protein.
  • the selected cell line is cultured to obtain the desired Mab.
  • Hybridoma cell lines secreting the monoclonal antibody are another aspect of this invention.
  • phage display technology could be utilised to select antibody genes with binding activities towards the polypeptide either from repertoires of PCR amplified v- genes of lymphocytes from humans screened for possessing anti-Fbp or from naive libraries (McCafferty, J. et al, Nature 348:552-554(1990), and Marks, J. et al, Biotechnology 10:779-783(1992)).
  • the affinity of these antibodies can also be improved by chain shuffling (Clackson, T. et al, Nature 352:624-628(1991)).
  • the antibody should be screened again for high affinity to the polypeptide and/or fusion protein.
  • a fragment of the final antibody may be prepared.
  • the antibody may be either intact antibody of M r approx 150,000 or a derivative of it, for example a Fab fragment or a Fv fragment as described in Skerra, A and Pluckthun, A., Science 240:1038-1040 (1988). If two antigen binding domains are present each domain may be directed against a different epitope - termed 'bispecific' antibodies.
  • the antibody of the invention may be prepared by conventional means for example by established monoclonal antibody technology (Kohler, G. and Milstein, C. (supra. (1975) or using recombinant means e.g. combinatorial libraries, for example as described in Huse, W.D. et al. Science 246:1275-1281 (1989).
  • the antibody is prepared by expression of a DNA polymer encoding said antibody in an appropriate expression system such as described above for the expression of polypeptides of the invention.
  • the choice of vector for the expression system will be determined in part by the host, which may be a prokaryotic cell, such as E. coli (preferably strain B) or Streptomyces sp. or a eukaryotic cell, such as a mouse C127, mouse myeloma, human HeLa, Chinese hamster ovary, filamentous or unicellular fungi or insect cell.
  • the host may also be a transgenic animal or a transgenic plant (for example, as described in Hiatt, A. et al, Nature 340:76-78(1989).
  • Suitable vectors include plasmids, bacteriophages, cosmids and recombinant viruses, derived from, for example, baculoviruses and vaccinia.
  • the Fab fragment may also be prepared from its parent monoclonal antibody by enzyme treatment, for example using papain to cleave the Fab portion from the Fc portion.
  • the antibody or derivative thereof is modified to make it less immunogenic in the patient.
  • the patient is human the antibody may most preferably be Tiumanised'; where the complimentarity determining region(s) of the hybridoma-derived antibody has been transplanted into a human monoclonal antibody , for example as described in Jones, P. et al, Nature 321:522-525 (1986), or Tempest et al, Biotechnology 9:266-273 (1991).
  • the modification need not be restricted to one of fiumanisation' ; other primate sequences (for example Newman, R. et al, Biotechnology 10: 1455-1460 (1992)) may also be used.
  • the humanised monoclonal antibody, or its fragment having binding activity form a particular aspect of this invention.
  • This invention provides a method of screening drugs to identify those which interfere with the proteins selected as targets herein, which method comprises measuring the interference of the activity of the protein by a test drug. For example if the protein selected has a catalytic activity, after suitable purification and formulation the activity of the enzyme can be followed by its ability to convert its natural substrates. By incorporating different chemically synthesised test compounds or natural products into such an assay of enzymatic activity one is able to detect those additives which compete with the natural substrate or otherwise inhibit enzymatic activity.
  • the invention also relates to inhibitors identified thereby.
  • a polynucleotide of the invention in genetic immunisation will preferably employ a suitable delivery method such as direct injection of plasmid DNA into muscles (Wolff et al, Hum. Mol. Genet. 1 :363 (1992); Manthorpe et al, Hum. Gene Ther. 4:419 (1963)), delivery of DNA complexed with specific protein carriers ( Wu et al, Biol. Chem. 264: 16985 (1989)), coprecipitation of DNA with calcium phosphate (Benvenisty & Reshef, Proc. Nat! Acad. Sci. USA.
  • a suitable delivery method such as direct injection of plasmid DNA into muscles (Wolff et al, Hum. Mol. Genet. 1 :363 (1992); Manthorpe et al, Hum. Gene Ther. 4:419 (1963)), delivery of DNA complexed with specific protein carriers ( Wu et al, Biol. Chem. 264: 16985
  • Suitable promoters for muscle transfection include CMV, RSV, SRa, actin, MCK, alpha globin, adenovirus and dihydrofolate reductase.
  • the active agent i.e., the polypeptide, polynucleotide or inhibitor of the invention
  • the active agent may be administered to a patient as an injectable composition, for example as a sterile aqueous dispersion, preferably isotonic.
  • the composition may be formulated for topical application for example in the form of ointments, creams, lotions, eye ointments, eye drops, ear drops, mouthwash, impregnated dressings and sutures and aerosols, and may contain appropriate conventional additives, including, for example, preservatives, solvents to assist drug penetration, and emollients in ointments and creams.
  • Such topical formulations may also contain compatible conventional carriers, for example cream or ointment bases, and ethanol or oleyl alcohol for lotions.
  • suitable conventional carriers for example cream or ointment bases, and ethanol or oleyl alcohol for lotions.
  • Such carriers may constitute from about 1% to about 98% by weight of the formulation; more usually they will constitute up to about 80% by weight of the formulation.
  • the daily dosage level of the active agent will be from 0.01 to 10 mg/kg, typically around 1 mg/kg.
  • the physician in any event will determine the actual dosage which will be most suitable for an individual patient and will vary with the age, weight and response of the particular patient.
  • the above dosages are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • a vaccine composition is conveniently in injectable form.
  • Conventional adjuvants may be employed to enhance the immune response.
  • a suitable unit dose for vaccination is 0.5-5ug/kg of antigen, and such dose is preferably administered 1-3 times and with an interval of 1-3 weeks. Within the indicated dosage range, no adverse toxicologicals effects are expected with the compounds of the invention which would preclude their administration to suitable patients. Screening Assays
  • a Factor of the invention may be employed in a screening process for compounds which bind the receptor and which activate (agonists) or inhibit activation of (antagonists) the receptor polypeptide of the present invention.
  • polypeptides of the invention may also be used to assess the binding of small molecule substrates and ligands in, for example, cells, cell-free preparations, chemical libraries, and natural product mixtures. These substrates and ligands may be natural substrates and ligands or may be structural or functional mimetics. See Coligan et al, Current Protocols in Immunology l(2):Chapter 5 (1991).
  • Factors of the invention are responsible for many biological functions, including many pathologies. Accordingly, it is desirous to find compounds and drugs which stimulate a Factor of the invention on the one hand and which can inhibit the function of a Factor of the invention on the other hand.
  • agonists are employed for therapeutic and prophylactic purposes for such conditions as Herpesvirus infections, such as HSV, VZV, HCMV and EBV infections.
  • Antagonists may be employed for a variety of therapeutic and prophylactic purposes for such conditions as Herpesvirus infection, such as HSV, VZV, HCMV and EBV infections.
  • screening procedures involve producing appropriate cells which express the receptor polypeptide of the present invention on the surface thereof. Such cells include cells from mammals, yeast, Drosophila or E.
  • Cells expressing the receptor are then contacted with a test compound to observe binding, or stimulation or inhibition of a functional response.
  • the assays may simply test binding of a candidate compound wherein adherence to the cells bearing the receptor is detected by means of a label directly or indirectly associated with the candidate compound or in an assay involving competition with a labeled competitor. Further, these assays may test whether the candidate compound results in a signal generated by activation of the receptor, using detection systems appropriate to the cells bearing the receptor at their surfaces. Inhibitors of activation are generally assayed in the presence of a known agonist and the effect on activation by the agonist by the presence of the candidate compound is observed.
  • cDNA, protein and antibodies to a Factor of the invention may also be used to configure assays for detecting the effect of added compounds on the production of mRNA and protein from a Factor of the invention in cells.
  • an ELISA may be constructed for measuring secreted or cell associated levels of protein feom a Factor of the invention using monoclonal and polyclonal antibodies by standard methods known in the art, and this can be used to discover agents which may inhibit or enhance the production of a Factor of the invention (also called antagonist or agonist, respectively) from suitably manipulated cells or tissues. Standard methods for conducting screening assays are well understood in the art.
  • Examples of potential antagonists of a Factor of the invention include antibodies or, in some cases, oligonucleotides or proteins which are closely related to the ligand of a Factor of the invention, e.g., a fragment of the ligand, or small molecules which bind to the receptor but do not elicit a response, so that the activity of the receptor is prevented.
  • Prophylactic and Therapeutic Methods include antibodies or, in some cases, oligonucleotides or proteins which are closely related to the ligand of a Factor of the invention, e.g., a fragment of the ligand, or small molecules which bind to the receptor but do not elicit a response, so that the activity of the receptor is prevented.
  • This invention provides methods of treating abnormal conditions related to both an excess of and insufficient amounts of the biological activity of a Factor of the invention. If the activity of a Factor of the invention is in excess, several approaches are available.
  • One approach comprises administering to a subject an inhibitor compound (antagonist) as hereinabove described along with a pharmaceutically acceptable carrier in an amount effective to inhibit activation by blocking binding of ligands to a Factor of the invention, or by inhibiting a second signal, and thereby alleviating the abnormal condition.
  • soluble forms of a Factor of the invention polypeptides still capable of binding the ligand in competition with endogenous Factor of the invention may be administered. Typical embodiments of such competitors comprise fragments of the polypeptide from a Factor of the invention.
  • expression of the gene encoding endogenous Factor of the invention can be inhibited using expression blocking techniques.
  • Known such techniques involve the use of antisense sequences, either internally generated or separately administered. See, for example, O'Connor, J Neurochem (1991) 56:560 in Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression. CRC Press, Boca Raton, FL (1988).
  • oligonucleotides which form triple helices with the gene can be supplied. See, for example, Lee et al, Nucleic Acids Res (1979) 3:173; Cooney et al, Science (1988) 241 :456; Dervan et al, Science (1991) 251 : 1360. These oligomers can be administered per se or the relevant oligomers can be expressed in vivo.
  • a therapeutically effective amount of a compound which activates Factor of the invention i.e., an agonist as described above, in combination with a pharmaceutically acceptable carrier, to thereby alleviate the abnormal condition.
  • gene therapy may be employed to effect the endogenous production of a Factor of the invention by the relevant cells in the subject.
  • a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed above.
  • the retroviral expression construct may then be isolated and introduced into a packaging cell transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral particles containing the gene of interest.
  • These producer cells may be administered to a subject for engineering cells in vivo and expression of the polypeptide in vivo.
  • gene therapy see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics, T Strachan and A P Read, BIOS Scientific Publishers Ltd (1996). Diagnostic Assays
  • This invention also relates to the use of a Factor of the invention polynucleotides for use as diagnostic reagents. Detection of a differentially expressed Factor gene associated with a viral infection and/or reactivation, as compared to a normal individual, will provide a diagnostic tool that can add to or define a diagnosis of a disease or susceptibility to a disease which results from under-expression, over-expression or altered expression of a Factor gene. Nucleic acids for diagnosis may be obtained from a subject's cells, such as from blood, urine, saliva, tissue biopsy or autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR or other amplification techniques prior to analysis.
  • RNA or cDNA may also be used in similar fashion. Deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to labeled Factor gene-derived nucleotide sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase digestion or by differences in melting temperatures. DNA sequence differences may also be detected by alterations in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents, or by direct DNA sequencing. See, e.g., Myers et al, Science (1985) 230:1242.
  • nuclease protection assays such as RNase and S 1 protection or the chemical cleavage method. See Cotton et al., Proc Natl Acad Sci USA (1985) 85: 4397-4401.
  • an array of oligonucleotides probes comprising Factor gene nucleotide sequence or fragments thereof can be constructed to conduct efficient screening of e.g., genetic mutations.
  • Array technology methods are well known and have general applicability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability. (See for example: M.Chee et al. Science, Vol 274, pp 610-613 (1996)).
  • the diagnostic assays offer a process for diagnosing or determining a susceptibility to
  • Herpesvirus infection such as HSV, VZV, HCMV and EBV infection, through detection of mutation or difference in expression in the Factor of gene as compared to normal individuals by the methods described.
  • Herpesvirus infection such as HSV, VZV, HCMV and EBV infection
  • Assay techniques that can be used to determine levels of a protein, such as an a Factor of the invention, in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. The examples below are carried out using standard techniques, which are well known and routine to those of skill in the art, except where otherwise described in detail. The examples illustrate, but do not limit the invention. EXAMPLES Example 1: Infection of mice and reactivation experiments. 4 to 6 week old female BALB/c BYJ mice were obtained from Jackson
  • mice were anesthetized with intraperitoneal injection of ketamine (87mg kg)/xylazine (13 mg/kg), then, after corneal scarification, were inoculated in the eye with 10 4 PFU of HSV-1 17 + (Brown et al, 1973).
  • mice were sacrificed by cervical dislocation and TG were isolated. Groups of 6-10 explanted TG were incubated in Dulbecco's Modified Eagle medium supplemented with 5% fetal bovine serum at 37°C for 0, 1, 2, 4 or 24 hours post-explant (p.e.). In a single experiment, TG were explanted in the absence of serum.
  • mice were infected following light scarification of both ear pinnae with 10 5 PFU of HSV-1 SC-16 (Harbour et al. 1981).
  • transient hyperthermia was induced as described previously (Sawtell et al. 1992). Briefly, mice were placed in a 43°C water bath for 10-12 minutes, then dried and placed in a warm incubator (34°C) for approximately 30 min to prevent hypothermia. At varying times post treatment, mice were sacrificed by cervical dislocation and TG were removed.
  • Example 2 Extraction of RNA. Ganglia used for RNA preparation were snap frozen in liquid nitrogen.
  • Example 3 Differential Display RT-PCR.
  • RNA was prepared from 300 ng RNA from latently- infected TG at 0, 1, 2, and 4 hours p.e. using the Differential Display Kit (TM) (Display Systems Biotech, Inc., Los Angeles, CA), as described by manufacturer. Primers presented in this study are listed in Table 2. Briefly, RNA from each sample was incubated, with one of nine downstream primers containing 1 IT residues and 2 nucleotide anchors (AA, AC, AG, CA, CC, CG, GA, GC, GG), for one hour at 40°C, followed by 5 min at 95°C to inactivate the M-MuLV enzyme. cDNA was stored at -70"C.
  • TM Differential Display Kit
  • cDNA was subjected to PCR amplification with Display Taq (TM) (Display Systems Biotech) using the original downstream primer, one of 24 10-mer 5' primers, and a- 33 P-dATP (Warthoe, 1995).
  • PCR conditions were 35 cycles of 30 sec denaturation at 94"C, 60 sec primer annealing at 40"C, and 60 sec extension at 72"C, employing a Perkin Elmer Cetus Gene Amp PCR System thermocycler. A final extension reaction was then performed for 5 min 72°C .
  • Radiolabeled reaction products were subjected to high resolution polyacrylamide/urea gel electrophoresis as described (Liang & Pardee, 1992). Gels were dried on Whatman filters and analyzed by autoradiography. Differentially-displayed PCR bands were cut out from the filter paper and dissolved in DEPC-treated water (Ambion) for 30 min at room temperature followed by 10 min at lOO'C.
  • Example 4 Reamplification PCR.
  • each differentially displayed band was reamplified in 4 individual PCR reactions.
  • T7- T11VVN 3' primers were used, where VV was the original DDRT nucleotide anchor, T7 was a 23 nucleotide portion of the T7 promoter (TAATACGACTC ACTATAGGGCCC), and N was A,G,T, or C.
  • the Reamplification reactions included the original upstream primers, 2mM dNTP and the Stoffel fragment of Taq Polymerase (Perkin Elmer Cetus). Reactions were performed under the original DDRT-PCR conditions. PCR products were separated by agarose electrophoresis and the most prominent product among the four parallel reactions was isolated for automated sequencing and further confirmation.
  • Isolated reamplification bands were used as templates for synthesis of 32 P labeled ⁇ boprobes with the MAXIsc ⁇ ptTM kit (Ambion , Austin, TX) as described by manufacturer RNase protection assays (RPA) were performed using HybspeedTM RPA kit (Ambion), and 0 5- 1 mg total RNA from a second, new set of latently-infected and uninfected TG explants
  • Mouse ⁇ -actm ⁇ boprobes provided with the MAXIsc ⁇ ptTM kit were used as controls
  • Probes and protected fragments were analyzed by denaturing polyacrylamide gel electrophoresis (PAGE) and phospho ⁇ maging
  • Example 6 PCR amplification of cDNA.
  • the second confirmatory PCR using specific primer sets was performed as follows cDNA was generated from 2 mg of total RNA using Superscript Preamphfication kit priming with ohgo (dT) and random hexamers (Gibco BRL) Reactions were performed in 25-ml volumes containing 4% of cDNA, 200 mM each deoxynucleoside t ⁇ phosphate (Pharmacia), 1 mM of each primer , 1 25 U of AmphTaq GoldTM (Perkin Elmer) with PCR Buffer A (Fisher) Primer pairs used are described m Table 3
  • Ganglia used for immunohistochemistry were fixed for 24 h with 4% paraformaldehyde in PBS then immersed in 70% ethanol/ 150 mM NaCl for 24 h and embedded in paraffin wax, and 6 mm serial sections were cut and processed as described elsewhere (Randazzo et al, 1995).
  • Rabbit polyclonal antisera to HSV-1 (Dako Corporation, Carpinteria, CA) was used for detection of replicating virus as described (Adams et al, 1984, Kesari et al, 1995).
  • Rabbit polyclonal anti-mouse interferon-a/b (Lee Biomolecular Research, San Diego, CA), and rabbit polyclonal anti-mouse TNF-a (Genzyme Diagnostics, Cambridge, MA) were used to probe for cytokines.
  • Rabbit polyclonal anti-TIS7 was a generous gift from B. Varnum, Amgen, CA. Antigen-expressing cells were detected by an indirect avidin-biotin immunoperoxidase method (Vectastain ABC kit, Vector Labs, Burlingham, CA), with 3,3'-diaminobenzadine (DAB) as the chromagen (Trojanowski et al, 1993).
  • RNA was prepared from latently-infected TG at different time points (0, 1, 2, and 4 hours) following explantation into culture media.
  • Complementary DNA was amplified using a set of arbitrary PCR primers.
  • the PCR products were resolved by PAGE and visualized by autoradiography. Every pair of primers (216 primer combinations) identified a limited number of target sequences within the pool of cDNAs. Thus, a typical reaction generated 50-200 distinct radiolabeled PCR products between 50 and 600 bp in length.
  • the majority of PCR products were present at identical levels in samples derived from different time points ( Figure 1). However, over 100 differentially-displayed PCR products were detected and isolated.
  • RNA corresponding to the isolated bands was differentially expressed in either uninfected or latently-infected TG explants.
  • Reamplified PCR products were used as probes in quantitative RNase protection assays (RPA).
  • RNA corresponding to band #56 was induced by 2 hours following explantation of uninfected ( Figure 3 A) and infected (results not shown) explants.
  • Phosphorimager quantitation Figure 3B
  • Similar results were obtained using probes generated from band #64 (results not shown).
  • RNA transcripts corresponding to differentially-displayed bands #56, and #64 were clearly induced in TG by explantation.
  • cDNA prepared from a different set of latently infected TG explants was subjected to PCR using primers specific for TIS7 (Table 3. Each PCR reaction yielded single product bands whose expected sizes corresponded to TIS7 cDNA.
  • TIS7 detected with these primers was rapidly induced in both infected ( Figure 3C) and uninfected explants (not shown), confirming the RPA results ( Figure 3A).
  • TIS7 expression returned to basal levels ( Figure 3C).
  • We also confirmed TIS7 induction in neuronal cells by immunostaining using affinity purified polyclonal antisera directed against TIS7 (not shown).
  • induction of TIS7 following explantation of ganglia was detected by three independent methods: DDRT- PCR, RPA, and immunostaining.
  • HSV latency occurs primarily in neuronal cells which innervate the cornea (Cook et al, 1974, Ramakrishnan et al, 1994, Ramakrishnan et al, 1996). These neurons represent approximately 2% of the neuronal population in the TG (Arvidson, 1977, Marfurt et al, 1989).
  • TG sections from latently-infected and uninfected explants were analyzed by immunohistochemistry. IFN ⁇ / ⁇ protein expression was not detected at 0 h p.e., and was induced at 4, 8, and 24 h p.e.
  • IFN Interferon Regulatory Factor -1
  • IRF Interferon Regulatory Factor -1
  • ICSs interferon consensus sequences
  • IRF-1 is an activator of IFN-b, whereas IRF-2 functions as a repressor (Watanabe et al, 1991).
  • IRF-1 transcription was induced within the first hour p.e., and its profile of induction was strikingly similar to IFN- ⁇ ( Figure 4A).
  • IFN ⁇ R or IRF-2 Figure 4 C, 4D.
  • IFN ⁇ R IFN- ⁇ receptor
  • IRF-2 Figure 4 C, 4D
  • TNF-a Soluble tumor necrosis factor (TNF)-a enhances the reactivation frequency and replication of HSV-1 during explant reactivation (Walev et al, 1995).
  • RT-PCR was performed using primers specific for TNF- ⁇ . TNF- ⁇ transcripts were induced rapidly following explant ( Figure 6). However, we were unable to detect TNF- ⁇ , a secreted factor, in TG sections by immunostaining.
  • Example 15 Oligonucleotides, probes, and electromobility shift assays.
  • DNA binding assays were carried out as described previously (Frazier et al., 1996). The reaction was in 20 ul total volume containing 2 ul (20 ug) BSA (NEB) 0.2 ul (2 ug) sheared salmon sperm DNA9 ul buffer D (20 mM herpes, pH7.9, 100 mM KC1, 20% glycerol, 0.2 mM EDTA, 1 mM DTT, protein (usually 1-2 ul) and water. In samples where antibody incunbation was added, the reaction was incubated with unlabeled probe (100 fold) at room T for 20 minutes before the probe ( ⁇ 1 ng, 30K cpm) being added. Reaction was done at RT for 20 minutes.
  • Example 16 In vitro translated IRF-1 binds to LAT ISRE.
  • IRF-1 protein sample was generated by in vitro transcription and translation of a human IRF-1 DNA template (generous gift from Richard Pine, New York University) by using the rabbit reticulate lysate system (Promega) and incubated with LAT probe. As shown in figure 8, IRF-1 formed a complex LAT ISRE DNA sequence. This complex was supershifted with IRF- 1 specific antisera but not with IRF-2 antisera, indicating the specificity of the interaction. We have been unable to show in gel mobility shift assays competition experiments with oriL site III oligo, and oriS site I oligo possess IRF binding sites (not shown).
  • the LAT region ISRE was shown to be able to associate with IRF-2 in phorbol ester stimulated Jurkat cells extracts (Santa Cruz) in gel mobility shift assays (not shown).
  • the IRF-2/ISRE complex formed when the LAT probe was incubated with Jurkat cell extracts was competed by wild-type LAT and ISRE probes (Santa Cruz) but not by mutant ISRE mQpinr (Santa Cruz), mutated LAT probes LATmAT and LATmm that contain two and three nucleotide changes, respectively, or and oriS Site I oligo (figure 9).
  • Transient hyperthermia is known to cause reactivation of HSV-1 from latency (Sawtell et al, 1992).
  • IFN-related genes such as TIS7 and IRF-1 are induced by another reactivation stimulus.
  • samples from individual hyperthermia-treated mice were prepared.
  • Figure 10A cellular housekeeping genes were realtively similar in all the animals, whereas HSP70, TIS7, and IRF-1 transcription was induced within the first 1-2 hours following hyperthermia ( Figure 10 B, C, D). Similar results were obtained in two experiments using samples obtained from different mice (not shown). Our results suggest that the pathway leading to induction of TIS7 and IRF-1 is common to different reactivation stimuli.
  • IRF-1 interferon regulatory factor 1
  • IRF-2 interferon regulatory factor 2
  • LPS lipopolysaccharide
  • Interferon regulatory factor-2 physically interacts with NF-kappa B in vitro and inhibits NF-kappa B induction of major histocompatibility class I and beta 2-microglobulin geneexpression in transfected human neuroblastoma cells.
  • IFN regulatory factor- 1 plays a central role in the regulation of the expression of class I and II MHC genes in vivo. J. Immunol. 158, 4260-4269. Honda, Y., Kawai, A. & Matsumoto, S. (1985). Persistent infection of rabies virus (HEP- Flury strain) in human neuroblastoma cells capable of producing interferon. J. Gen. Virol. 66, 957-967.
  • Nerve injury enhances rat neuronal glutamate transporter expression: identification by differential display PCR. J. Neuroscience 15, 7872-8.
  • Tyrosine phosphorylated p91 binds to a single element in the ISGF2/IRF- 1 promoter tomediate induction by IFN alpha and IFN gamma, and is likely to autoregulate the p91 gene.
  • EMBO J 13
  • OBF-1 a novel B cell-specific coactivator that stimulates immunoglobulin promoter activity through association with octamer-binding proteins. Cell 80, 497-506.
  • TIS7 a gene induced in Swiss 3T3 cells by the tumor promoter tetradecanoyl phorbol acetate. Oncogene 4, 1263-1265.
  • IFN-beta element by IRF- 1 requires a posttranslational event in addition to IRF-1 synthesis. Nuc. Ac. Res. 19, 4421-8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
EP98935519A 1997-07-03 1998-07-01 Behandlungsmethode eines lignocellulosischen materials oder eines expandierten minerals zur formung eines fertigproduktes Withdrawn EP1001967A4 (de)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US5163397P 1997-07-03 1997-07-03
US51633P 1997-07-03
US5451597P 1997-08-01 1997-08-01
US54515P 1997-08-01
US8035298P 1998-04-01 1998-04-01
US80352P 1998-04-01
PCT/US1998/013733 WO1999001464A1 (en) 1997-07-03 1998-07-01 Componds and methods for treating and screening viral reactivation

Publications (2)

Publication Number Publication Date
EP1001967A1 true EP1001967A1 (de) 2000-05-24
EP1001967A4 EP1001967A4 (de) 2004-12-29

Family

ID=27367993

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98935519A Withdrawn EP1001967A4 (de) 1997-07-03 1998-07-01 Behandlungsmethode eines lignocellulosischen materials oder eines expandierten minerals zur formung eines fertigproduktes

Country Status (4)

Country Link
EP (1) EP1001967A4 (de)
JP (1) JP2002511877A (de)
CA (1) CA2295933A1 (de)
WO (1) WO1999001464A1 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005019478A1 (en) * 2003-08-18 2005-03-03 Irm Llc Methods and compositions for modulating herpesviral replication and transcription activator
US8802106B2 (en) 2009-10-30 2014-08-12 The Administrators Of The Tulane Educational Fund Peptide compositions and methods for inhibiting herpesvirus infection

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0355190A1 (de) * 1988-08-24 1990-02-28 Tadatsugu Dr. Taniguchi Genexpression regulierender Faktor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9901464A1 *

Also Published As

Publication number Publication date
EP1001967A4 (de) 2004-12-29
CA2295933A1 (en) 1999-01-14
WO1999001464A1 (en) 1999-01-14
JP2002511877A (ja) 2002-04-16

Similar Documents

Publication Publication Date Title
Zimmermann et al. Isolation and characterization of cDNA clones encoding the human carcinoembryonic antigen reveal a highly conserved repeating structure.
US8236507B2 (en) Modulators of TNF receptor associated factor (TRAF), their preparation and use
AU697535B2 (en) Haemopoietic maturation factor
US6740738B2 (en) Antibody against hypoxia proteins
EP0567575B1 (de) Cytokin-induziertes protein, tsg-6, seine dna und verwendung
WO1999062952A1 (en) A human zinc finger protein gene (bmzf2)
US20050197307A1 (en) Compounds and methods for treating and screening viral reactivation
WO1999001464A1 (en) Componds and methods for treating and screening viral reactivation
WO1993019179A1 (en) Ifn receptors recognition factors, protein sequences and methods of use thereof
CN112402592A (zh) Spink7蛋白在制备促进过度炎症创面愈合的药物中的应用
US7524928B2 (en) IFN receptor 1 binding proteins
US6605442B1 (en) Methods of testing drugs or agents that modulate the activity of receptor recognition factors
US7264945B2 (en) IFN receptor 1 binding proteins, DNA encoding them, and methods of modulating cellular response to interferons
WO1996023410A1 (en) UBIQUITIN CONJUGATING ENZYMES 7, 8 and 9
CA2264544A1 (en) Snrnp sm proteins
EP1224220A2 (de) Gen 1 der epo-primärantwort, eprg1
EP0556395B1 (de) Neuartige cytokine
US5945321A (en) Ubiquitin conjugating enzymes 7, 8 and 9
EP1780277A1 (de) IFN Rezeptor 1 bindende Proteine, dafür kodierende DNA und Verfahren zur Regulierung der zellulären Antwort auf Interferone
MXPA99006595A (en) Novel ifn receptor 1 binding proteins, dna encoding them, and methods of modulating cellular response to interferons
AU3508101A (en) Modulators of TNF receptor associated factor (TRAF), their preparation and use
MXPA96003897A (en) Factor 10 of fibroblas growth

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19991221

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): BE CH DE ES FR GB IT LI NL

A4 Supplementary search report drawn up and despatched

Effective date: 20041109

17Q First examination report despatched

Effective date: 20081219

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090501