EP0983078A1 - SIALOADHESIN FAMILY 4 cDNA - Google Patents

SIALOADHESIN FAMILY 4 cDNA

Info

Publication number
EP0983078A1
EP0983078A1 EP98923812A EP98923812A EP0983078A1 EP 0983078 A1 EP0983078 A1 EP 0983078A1 EP 98923812 A EP98923812 A EP 98923812A EP 98923812 A EP98923812 A EP 98923812A EP 0983078 A1 EP0983078 A1 EP 0983078A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
seq
identity
amino acid
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Withdrawn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
EP98923812A
Other languages
German (de)
French (fr)
Other versions
EP0983078A4 (en
Inventor
Kristine Kay Kikly
Connie Lynn Erickson-Miller
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 Corp
Original Assignee
SmithKline Beecham Corp
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 Corp filed Critical SmithKline Beecham Corp
Publication of EP0983078A1 publication Critical patent/EP0983078A1/en
Publication of EP0983078A4 publication Critical patent/EP0983078A4/en
Withdrawn legal-status Critical Current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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • This invention relates to newly identified polypeptides and polynucleotides encoding such polypeptides, to their use in therapy and in identifying compounds which may be agonists, antagonists and /or inhibitors which are potentially useful in therapy, and to production of such polypeptides and polynucleotides.
  • the drug discovery process is currently undergoing a fundamental revolution as it embraces 'functional genomics' , that is, high throughput genome- or gene-based biology. This approach is rapidly superceding earher approaches based on 'positional cloning' . A phenotype, that is a biological function or genetic disease, would be identified and this would then be tracked back to the responsible gene, based on its genetic map position.
  • the present invention relates to SAF-4, in particular SAF-4 polypeptides and SAF-4 polynucleotides, recombinant materials and methods for their production.
  • the invention relates to methods for using such polypeptides and polynucleotides, including the treatment of cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotr ⁇ phic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reper usion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormalities, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, hereinafter
  • the invention relates to methods for identifying agomsts and antagonists/inhibitors using the materials provided by the invention, and treating conditions associated with SAF-4 imbalance with the identified compounds.
  • the invention relates to diagnostic assays for detecting diseases associated with inappropriate SAF-4 activity or levels. Description of the Invention
  • the present invention relates to SAF-4 polypeptides.
  • SAF-4 polypeptides include isolated polypetides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99 % identity, to that of SEQ ID NO: 2 over the entire length of SEQ ID NO: 2.
  • polypeptides include those comprising the amino acid of SEQ ID NO:2.
  • peptides of the present invention include isolated polypeptides in which the amino acid sequence has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO: 2 over the entire length of SEQ ID NO: 2.
  • polypeptides include the polypeptide of SEQ ID NO:2.
  • peptides of the present invention include isolated polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 1.
  • Polypeptides of the present invention are believed to be members of the Sialoadhesin family of polypeptides. They are therefore of interest because the sialoadhesin family of proteins, sialoadhesin, CD33, CD22 and myelin-associated glycoprotein (MAG), are utilized as cellular interaction molecules. They bind specific carbohydrates in a sialic acid-dependent manner on target cells.
  • the extracellular domain is made up of various numbers of immunoglobulin-like domains of the N-like and C2-like subtypes and the intracellular portion has no known homology to any signalling motifs.
  • Sialoadhesin expression is restricted to macrophages, it has 17 Ig-like domains and the specific recognition sequence on target cells is ⁇ eu5Aca2,3Galbl3Gal ⁇ Ac.
  • Known target cells are developing myeloid cells in the bone marrow and lymphocytes in the spleen and lymph node (Crocker, P. R. , et al. EMBO J, 1994, 13:4490-4503).
  • CD22 is expressed only on B cells and has a and b isoforms with 5 and 7 Ig-like domains, respectively.
  • CD22 is known to bind T cells, B cells, monocytes, granulocytes and erythrocytes by recognizing Neu5Aca2,6Galbl,4Glc(NAc) in N-linked glycans (Crocker, P. R., et al. EMBO J, 1994, 13:4490-4503; Sta enkovic, I. and Seed, B. Nature, 1990, 345:74-77; Wilson, G. L., et al. J Exp Med, 1991, 173:137-146).
  • MAG Myelin- associated glycoprotein
  • L-MAG large MAG
  • S-MAG small MAG
  • CD33 is most relevant to SAF-4 because they are the most closely related of all the family members. CD33 is normally expressed on the developing myelomonocytic lineage. It is absent on early stem cells but is present on colony-forming units for granulocytes, erythrocytes, monocytes, and megakaryocytes (CFU-GEMM) and progenitors of granulocytes and mononuclear phagocytes (CFU-GM). It is downregulated by mature granulocytes but retained by mature monocytes and macrophages (Andrews, R. G., et al., Blood, 1983, 62:124; Griffin, J. D., et al., Leuk Res 1984, 8:521).
  • CD33 has two Ig-like domains and prefers to bind targets expressing NeuAca2,3Gal in N- and O-linked glycans. It maps to chromosome 19ql3.1-13.3, closely linking it in the genome with MAG and CD22 (Freeman, S. D., et al., Blood, 1995, 85:2005-2012).
  • CD33 has also been found to be expressed on about 85 % of leukemic myeloblasts in patients with acute myelogenous leukemia (AML) and is frequently used to differentiate AML from acute lymphoblastic leukemia (ALL).
  • AML acute myelogenous leukemia
  • ALL acute lymphoblastic leukemia
  • Monoclonal antibodies to CD33 have been used therapeutically to purge residual myeloblasts from autologous bone marrow grafts ex vivo for the treatment of AML (Robertson, M. J., et al., Blood, 1992 79:2229-2236). More recently, humanized monoclonal antibodies to CD33 have undergone evaluation in vivo for the treatment of AML (Caron, P. C, et al., Blood, 1994, 83:1760-1768).
  • SAF-4 activity or “SAF-4 polypeptide activity” or “biological activity of SAF-4" .
  • antigenic and immunogenic activities of said SAF-4 polypeptides in particular the antigenic and immunogenic activities of the polypeptide of SEQ ID NO:2.
  • a polypeptide of the present invention exhibits at least one biological activity of SAF-4.
  • polypeptides of the present invention may be in the form of the "mature" protein or may be a part of a larger protein such as a fusion protein. It is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production.
  • the present invention also includes include variants of the aforementioned polypetides, that is polypeptides that vary from the referents by conservative amino acid substitutions, whereby a residue is substituted by another with like characteristics. Typical such substitutions are among Ala, Nal, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr. Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 amino acids are substituted, deleted, or added in any combination.
  • Polypeptides of the present invention can be prepared in any suitable manner. Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art. In a further aspect, the present invention relates to SAF-4 polynucleotides.
  • polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to the amino acid sequence of SEQ ID ⁇ O:2, over the entire length of SEQ ID NO:2.
  • polypeptides which have at least 97% identity are highly preferred, whilst those with at least 98-99 % identity are more highly preferred, and those with at least 99 % identity are most highly preferred.
  • polynucleotides include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO:l encoding the polypeptide of SEQ ID NO:2.
  • Further polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence that has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO: 2, over the entire coding region.
  • polynucleotides which have at least 97 % identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99 % identity are most highly preferred.
  • polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to SEQ ID NO: 1 over the entire length of SEQ ID NO: 1.
  • polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identiy are more highly preferred, and those with at least 99 % identity are most highly preferred.
  • Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO: 1 as well as the polynucleotide of SEQ ID NO: 1.
  • the invention also provides polynucleotides which are complementary to all the above described polynucleotides.
  • the nucleotide sequence of SEQ ID NO:l shows homology with CD33 (Simmons, D., and Seed, B., JI 141:2797-2800, 1988).
  • the nucleotide sequence of SEQ ID NO:l is a cDNA sequence and comprises a polypeptide encoding sequence (nucleotide 51 to 1970) encoding a polypeptide of 639 amino acids, the polypeptide of SEQ ID NO:2.
  • the nucleotide sequence encoding the polypeptide of SEQ ID NO: 2 may be identical to the polypeptide encoding sequence contained in SEQ ID NO:l or it may be a sequence other than the one contained in SEQ ID NO: 1 , which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ID NO:2.
  • the polypeptide of the SEQ ID NO: 2 is structurally related to other proteins of the Sialoadhesin family, having homology and/or structural similarity withCD33 (Simmons, D., and Seed, B., JI 141:2797-2800, 1988).
  • Preferred polypeptides and polynucleotides of the present invention are expected to have, inter alia, similar biological functions/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides and polynucleotides of the present invention have at least one SAF-4 activity.
  • the present invention also relates to partial or other polynucleotide and polypeptide sequences which were first identified prior to the determination of the corresponding full length sequences of SEQ ID NO: 1 and SEQ ED NO:2.
  • the present invention provides for an isolated polynucleotide comprising:
  • nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity to SEQ ID NO: 3 over the entire length of SEQ ID NO: 3;
  • nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity, to SEQ ID NO: 3 over the entire length of SEQ ID NO:3;
  • nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO:4, over the entire length of SEQ ID NO:4; as well as the polynucleotide of SEQ ID NO: 3.
  • the present invention further provides for a polypeptide which:
  • (a) comprises an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99 % identity, to that of SEQ ID NO:4 over the entire length of SEQ ID NO:4;
  • (b) has an amino acid sequence which is at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO: 4 over the entire length of SEQ ID NO:4;
  • (c) comprises the amino acid of SEQ ID NO: 4.
  • (d) is the polypeptide of SEQ ID NO:4; as well as polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 3.
  • nucleotide sequence of SEQ ID NO: 3 and the peptide sequence encoded thereby are derived from EST (Expressed Sequence Tag) sequences. It is recognized by those skilled in the art that there will inevitably be some nucleotide sequence reading errors in EST sequences (see Adams, M.D. et al, Nature 377 (supp) 3, 1995).
  • nucleotide sequence of SEQ ID NO: 3 and the peptide sequence encoded therefrom are therefore subject to the same inherent limitations in sequence accuracy.
  • the peptide sequence encoded by SEQ ID NO: 3 comprises a region of identity or close homology and/or close structural similarity (for example a conservative amino acid difference) with the closest homologous or structurally similar protein.
  • Polynucleotides of the present invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of human primary dendritic cells, using the expressed sequence tag (EST) analysis (Adams, M.D., et al. Science (1991) 252:1651-1656; Adams, M.D. et al , Nature, (1992) 355:632-634; Adams, M.D. , et al. , Nature (1995) 377 Supp:3-174). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesized using well known and commercially available techniques.
  • EST expressed sequence tag
  • the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide in reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions.
  • a marker sequence which facilitates purification of the fused polypeptide can be encoded.
  • the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, Inc.) and described in Gentz et al. , Proc Natl Acad Sci USA (1989) 86:821-824, or is an HA tag.
  • the polynucleotide may also contain non-coding 5' and 3' sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ribosome binding sites and sequences that stabilize mRNA.
  • polypeptide variants which comprise the amino acid sequence of SEQ ID NO:2 and in which several, for instance from 5 to 10, 1 to 5, 1 to 3, 1 to 2 or 1, amino acid residues are substituted, deleted or added, in any combination.
  • Polynucleotides which are identical or sufficiently identical to a nucleotide sequence contained in SEQ ID NO: 1 may be used as hybridization probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides of the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding homologs and orthologs from species other than human) that have a high sequence similarity to SEQ ID NO: 1.
  • these nucleotide sequences are 70% identical, preferably 80% identical, more preferably 90% identical, most preferably 95 % identical to that of the referent.
  • the probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides. Particularly preferred probes will have between 30 and 50 nucleotides.
  • a polynucleotide encoding a polypeptide of the present invention may be obtained by a process which comprises the steps of screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof; and isolating full- length cDNA and genomic clones containing said polynucleotide sequence.
  • Such hybridization techniques are well known to the skilled artisan.
  • Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in 0. Ix SSC at about 65°C.
  • the present invention also includes polynucleotides obtainable by screening an appropriate library under stingent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof.
  • an isolated cDNA sequence will be incomplete, in that the region coding for the polypeptide is cut short at the 5 ' end of the cDNA. This is a consequence of reverse transcriptase, an enzyme with inherently low 'processivity' (a measure of the ability of the enzyme to remain attached to the template during the polymerization reaction), failing to complete a DNA copy of the mRNA template during 1st strand cDNA synthesis.
  • PCR Nucleic acid amplification
  • the products of this reaction can then be analysed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or carrying out a separate full-length PCR using the new sequence information for the design of the 5' primer.
  • Recombinant polypeptides of the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, in a further aspect, the present invention relates to expression systems which comprise a polynucleotide or polynucleotides of the present invention, to host cells which are genetically engineered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. For recombinant production, host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention.
  • polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al. , Basic Methods in Molecular Biology (1986) and Sambrook et al. , Molecular Cloning: A Laboratory Manual, 2nd Ed. , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
  • Preferred such methods include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection.
  • bacterial cells such as streptococci, staphylococci, E. coli, Streptomyces and Bacillus subtilis cells
  • fiingal cells such as yeast cells and Aspergillus cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells
  • plant cells include bacterial cells, such as streptococci, staphylococci, E. coli, Streptomyces and Bacillus subtilis cells
  • fiingal cells such as yeast cells and Aspergillus cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells
  • plant cells such as CHO, COS, HeLa, C127, 3T3, BHK, HE
  • expression systems can be used, for instance, chromosomal, episomal and virus-derived systems, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids.
  • the expression systems may contain control regions that regulate as well as engender expression.
  • any system or vector which is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used.
  • the appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook et al. , MOLECULAR CLONING, A LABORATORY MANUAL (supra).
  • Appropriate secretion signals may be incorporated into the desired polypeptide to allow secretion of the translated protein into the lumen of the endoplasmic reticulum, the periplasmic space or the extracellular environment. These signals may be endogenous to the polypeptide or they may be heterologous signals.
  • a polypeptide of the present invention is to be expressed for use in screening assays, it is generally preferred that the polypeptide be produced at the surface of the cell. In this event, the cells may be harvested prior to use in the screening assay. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the polypeptide. If produced intracellularly, the cells must first be lysed before the polypeptide is recovered.
  • Polypeptides of the present invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
  • This invention also relates to the use of polynucleotides of the present invention as diagnostic reagents. Detection of a mutated form of the gene characterized by the polynucleotide of SEQ ID NO: 1 which is associated with a dysfunction 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 the gene. Individuals carrying mutations in the gene may be detected at the DNA level by a variety of techniques.
  • 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 SAF-4 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 (ee, e.g., Myers et al. , Science (1985) 230: 1242). Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (see Cotton et al. , Proc NatlAcad Sci USA (1985) 85: 4397-4401).
  • an array of oligonucleotides probes comprising SAF-4 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 the Diseases through detection of mutation in the SAF-4 gene by the methods described.
  • diseases may be diagnosed by methods comprising determining from a sample derived from a subject an abnormally decreased or increased level of polypeptide or mRNA.
  • Decreased or increased expression can be measured at the RNA level using any of the methods well known in the art for the quantitation of polynucleotides, such as, for example, nucleic acid amplification, for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods.
  • Assay techniques that can be used to determine levels of a protein, such as a polypeptide of the present 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 and flow cytometric analysis.
  • the present invention relates to a diagonostic kit which comprises:
  • a polynucleotide of the present invention preferably the nucleotide sequence of SEQ ID NO: 1, or a fragment thereof ;
  • b a nucleotide sequence complementary to that of (a);
  • polypeptide of the present invention preferably the polypeptide of SEQ ID NO: 2 or a fragment thereof; or
  • kits may comprise a substantial component.
  • a kit will be of use in diagnosing a disease or suspectability to a disease, particularly cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotrophic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reperfusion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormahties, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, amongst others.
  • the nucleotide sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome.
  • the mapping of relevant sequences to chromosomes according to the present invention is an important first step in correlating those sequences with gene associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).
  • the differences in the cDNA or genomic sequence between affected and unaffected individuals can also be determined. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
  • polypeptides of the invention or their fragments or analogs thereof, or cells expressing them, can also be used as immunogens to produce antibodies immunospecific for polypeptides of the present invention.
  • immunospecific means that the antibodies have substantially greater affinity for the polypeptides of the invention than their affinity for other related polypeptides in the prior art.
  • Antibodies generated against polypeptides of the present invention may be obtained by administering the polypeptides or epitope-bearing fragments, analogs or cells to an animal, preferably a non-human animal, using routine protocols.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature (1975) 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al. , Immunology Today (1983) 4:72) and the EBN-hybridoma technique (Cole et al. , MONOCLONAL ANTIBODIES AND CANCER THERAPY, pp. 77-96, Alan R. Liss, Inc. , 1985).
  • the above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or to purify the polypeptides by affinity chromatography.
  • Antibodies against polypeptides of the present invention may also be employed to treat the Diseases, amongst others.
  • Antibodies against SAF-4 polypeptides may also be employed to subcharacterize cell populations during hematopoietic development, as a diagnostic marker to distinguish between different forms of cancer, to purge bone marrow ex vivo of cancer cells expressing SAF-4, as a tool to aid in the ex vivo expansion (proliferation and/or differentiation) of hematopoietic progenitor cells expressing SAF-4, as a stimulus in vivo for stem cell mobilization into the periphery, and as an in vivo chemoprotective agent.
  • the present invention relates to genetically engineered soluble fusion proteins comprising a polypeptide of the present invention, or a fragment thereof, and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclasses (IgG, IgM, IgA, IgE).
  • immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgGl , where fusion takes place at the hinge region.
  • the Fc part can be removed simply by incorporation of a cleavage sequence which can be cleaved with blood clotting factor Xa.
  • this invention relates to processes for the preparation of these fusion proteins by genetic engineering, and to the use thereof for drug screening, diagnosis and therapy.
  • soluble forms of SAF-4 polypeptides still capable of binding the ligand in competition with endogenous SAF-4 may be administered.
  • Typical embodiments of such competitors comprise fragments of the SAF-4 polypeptide.
  • One example is using the extracellular domain of SAF-4 fused to a human immunoglobulin Fc region which can then be employed to treat cancer, inflammation, autoimmunity and allergy, among others.
  • SAF-4/Fc polypeptides may also be employed to purge bone marrow ex vivo of cancer cells expressing SAF-4 ligands, as a tool to aid in the ex vivo expansion (proliferation and/or differentiation) of hematopoietic progenitor cells expressing SAF-4 ligands, as a stimulus in vivo for stem cell mobilization into the periphery, and as an in vivo chemoprotective agent.
  • a further aspect of the invention also relates to polynucleotides encoding such fusion proteins. Examples of fusion protein technology can be found in International Patent Application Nos. WO94/29458 and WO94/22914.
  • Another aspect of the invention relates to a method for inducing an immunological response in a mammal which comprises inoculating the mammal with a polypeptide of the present invention, adequate to produce antibody and/or T cell immune response to protect said animal from the Diseases hereinbefore mentioned, amongst others.
  • Yet another aspect of the invention relates to a method of inducing immunological response in a mammal which comprises, delivering a polypeptide of the present invention via a vector directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce antibody to protect said animal from diseases.
  • a further aspect of the invention relates to an immunological/vaccine formulation (composition) which, when introduced into a mammalian host, induces an immunological response in that mammal to a polypeptide of the present invention wherein the composition comprises a polypeptide or polynucleotide of the present invention.
  • the vaccine formulation may further comprise a suitable carrier. Since a polypeptide may be broken down in the stomach, it is preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection).
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation instonic with the blood of the recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use.
  • the vaccine formulation may also include adjuvant systems for enhancing the immunogenicity of the formulation, such as oil-in water systems and other systems known in the art. The dosage will depend on the specific activity of the vaccine and can be readily determined by routine experimentation.
  • Polypeptides of the present invention are responsible for many biological functions, including many disease states, in particular the Diseases hereinbefore mentioned. It is therefore desirous to devise screening methods to identify compounds which stimulate or which inhibit the function of the polypeptide. Accordingly, in a further aspect, the present invention provides for a method of screening compounds to identify those which stimulate or which inhibit the function of the polypeptide.
  • agonists or antagonists may be employed for therapeutic and prophylactic purposes for such Diseases as hereinbefore mentioned.
  • Compounds may be identified from a variety of sources, for example, cells, cell- free preparations, chemical libraries, and natural product mixtures. Such agonists, antagonists or inhibitors so-identified may be natural or modified substrates, ligands, receptors, enzymes, etc.
  • the screening method may simply measure the binding of a candidate compound to the polypeptide, or to cells or membranes bearing the polypeptide, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound.
  • the screening method may involve competition with a labeled competitor. Further, these screening methods may test whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells bearing the polypeptide.
  • 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.
  • Constitutively active polpypeptides may be employed in screening methods for inverse agomsts or inhibitors, in the absence of an agonist or inhibitor, by testing whether the candidate compound results in inhibition of activation of the polypeptide. Further, the screening methods may simply comprise the steps of mixing a candidate compound with a solution containing a polypeptide of the present invention, to form a mixture, measuring SAF-4 activity in the mixture, and comparing the SAF-4 activity of the mixture to a standard.
  • Fusion proteins such as those made from Fc portion and SAF-4 polypeptide, as hereinbefore described, can also be used for high-throughput screening assays to identify antagonists for the polypeptide of the present invention (see D. Bennett et al., J Mol Recognition, 8:52-58 (1995); and K. Johanson et al, J Biol Chem, 270(16):9459-9471 (1995)).
  • the polynucleotides, polypeptides and antibodies to the polypeptide of the present invention may also be used to configure screening methods for detecting the effect of added compounds on the production of mRNA and polypeptide in cells.
  • an ELISA assay may be constructed for measuring secreted or cell associated levels of polypeptide using monoclonal and polyclonal antibodies by standard methods known in the art. This can be used to discover agents which may inhibit or enhance the production of polypeptide (also called antagonist or agonist, respectively) from suitably manipulated cells or tissues.
  • the polypeptide may be used to identify membrane bound or soluble receptors, if any, through standard receptor binding techniques known in the art. These include, but are not limited to, ligand binding and crosslinking assays in which the polypeptide is labeled with a radioactive isotope (for instance, ⁇ I), chemically modified (for instance, biotinylated), or fused to a peptide sequence suitable for detection or purification, and incubated with a source of the putative receptor (cells, cell membranes, cell supernatants, tissue extracts, bodily fluids). Other methods include biophysical techniques such as surface plasmon resonance and spectroscopy. These screening methods may also be used to identify agonists and antagonists of the polypeptide which compete with the binding of the polypeptide to its receptors, if any. Standard methods for conducting such assays are well understood in the art.
  • polypeptide antagonists examples include antibodies or, in some cases, ohgonucleotides or proteins which are closely related to the ligands, substrates, receptors, enzymes, etc. , as the case may be, of the polypeptide, e.g. , a fragment of the ligands, substrates, receptors, enzymes, etc. ; or small molecules which bind to the polypetide of the present invention but do not elicit a response, so that the activity of the polypeptide is prevented.
  • the present invention relates to a screening kit for identifying agonists, antagonists, Ugands, receptors, substrates, enzymes, etc. for polypeptides of the present invention; or compounds which decrease or enhance the production of such polypeptides, which comprises:
  • kits may comprise a substantial component.
  • a polypeptide of the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by: (a) determining in the first instance the three-dimensional structure of the polypeptide;
  • the present invention provides methods of treating abnormal conditions such as, for instance, cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotrophic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reperfusion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormahties, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, related to either an excess of, or an under-expression of, SAF-4 polypeptide activity.
  • abnormal conditions such as, for instance, cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, C
  • One approach comprises administering to a subject in need thereof an inhibitor compound (antagonist) as hereinabove described, optionally in combination with a pharmaceutically acceptable carrier, in an amount effective to inhibit the function of the polypeptide, such as, for example, by blocking the binding of ligands, substrates, receptors, enzymes, etc. , or by inhibiting a second signal, and thereby alleviating the abnormal condition.
  • an inhibitor compound as hereinabove described
  • a pharmaceutically acceptable carrier in an amount effective to inhibit the function of the polypeptide, such as, for example, by blocking the binding of ligands, substrates, receptors, enzymes, etc. , or by inhibiting a second signal, and thereby alleviating the abnormal condition.
  • soluble forms of the polypeptides still capable of binding the ligand, substrate, enzymes, receptors, etc. in competition with endogenous polypeptide may be administered. Typical examples of such competitors include fragments of the SAF-4 polypeptide.
  • expression of the gene encoding endogenous SAF-4 polypeptide 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).
  • oligomers can be administered per se or the relevant oligomers can be expressed in vivo.
  • SAF-4 For treating abnormal conditions related to an under-expression of SAF-4 and its activity, several approaches are also available.
  • One approach comprises administering to a subject a therapeutically effective amount of a compound which activates a polypeptide of the present 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 SAF-4 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.
  • Another approach is to administer a therapeutic amount of a polypeptide of the present invention in combination with a suitable pharmaceutical carrier.
  • the present invention provides for pharmaceutical compositions comprising a therapeutically effective amount of a polypeptide, such as the soluble form of a polypeptide of the present invention, agonist antagonist peptide or small molecule compound, in combination with a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically acceptable carrier or excipient include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • the invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention.
  • Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
  • composition will be adapted to the route of administration, for instance by a systemic or an oral route.
  • Preferred forms of systemic administration include injection, typically by intravenous injection. Other injection routes, such as subcutaneous, intramuscular, or intraperitoneal, can be used.
  • Alternative means for systemic administration include transmucosal and transdermal administration using penetrants such as bile salts or fusidic acids or other detergents.
  • penetrants such as bile salts or fusidic acids or other detergents.
  • oral administration may also be possible. Administration of these compounds may also be topical and or localized, in the form of salves, pastes, gels, and the like.
  • the dosage range required depends on the choice of peptide or other compounds of the present invention, the route of administration, the nature of the formulation, the nature of the subject's condition, and the judgment of the attending practitioner. Suitable dosages, however, are in the range of 0.1-100 ⁇ g/kg of subject. Wide variations in the needed dosage, however, are to be expected in view of the variety of compounds available and the differing efficiencies of various routes of administration. For example, oral administration would be expected to require higher dosages than administration by intravenous injection. Variations in these dosage levels can be adjusted using standard empirical routines for optimization, as is well understood in the art. Polypeptides used in treatment can also be generated endogenously in the subject, in treatment modalities often referred to as "gene therapy" as described above.
  • cells from a subject may be engineered with a polynucleotide, such as a DNA or RNA, to encode a polypeptide ex vivo, and for example, by the use of a retroviral plasmid vector.
  • the cells are then introduced into the subject.
  • Polynucleotide and polypeptide sequences form a valuable information resource with which to identify further sequences of similar homology. This is most easily facilitated by storing the sequence in a computer readable medium and then using the stored data to search a sequence database using well known searching tools, such as GCC.
  • the present invention provides for a computer readable medium having stored thereon a polynucleotide comprising the sequence of SEQ ID NO: 1 and/or a polypeptide sequence encoded thereby.
  • 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. A variety of modifications may be made to DNA and RNA; thus, “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 ohgonucleotides.
  • 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 post-translational 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 may 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 to 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 post-translation 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 (see, for instance, PROTEINS - STR
  • Variant refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, 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.
  • 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 relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as the case may be, as determined by comparing the sequences.
  • identity also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences.
  • Identity can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A.M., ed.
  • Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in pubhcly available computer programs.
  • Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux, J. , et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S.F. et al., J. Molec. Biol 215: 403-410 (1990).
  • the BLAST X program is pubhcly available from NCBI and other sources (BLAST Manual, Altschul, S.
  • Gap Penalty 12 Gap Length Penalty: 4
  • a program useful with these parameters is pubhcly available as the "gap" program from Genetics Computer Group, Madison WI.
  • the aforementioned parameters are the default parameters for peptide comparisons (along with no penalty for end gaps).
  • Gap Penalty 50 Gap Length Penalty: 3
  • Polynucleotide embodiments further include an isolated polynucleotide comprising a polynucleotide sequence having at least a 50, 60, 70, 80, 85, 90, 95, 97 or 100% identity to the reference sequence of SEQ ID NO:l, wherein said polynucleotide sequence may be identical to the reference sequence of SEQ ID NO: 1 or may include up to a certain integer number of nucleotide alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ ID NO: 1 by the integer
  • n n is the number of nucleotide alterations
  • x n is the total number of nucleotides in
  • SEQ ID NO:l is 0.50 for 50%, 0.60 for 60% , 0.70 for 70%, 0.80 for 80%, 0.85 for 85 %, 0.90 for 90%, 0.95 for 95 %, 0.97 for 97% or 1.00 for 100% , and • is the symbol for the multiplication operator, and wherein any non-integer product of x n and y is rounded down to the nearest integer prior to subtracting it from x n .
  • Alterations of a polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 may create nonsense, missense or frameshift mutations in this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
  • a polynucleotide sequence of the present invention may be identical to the reference sequence of SEQ ID NO: 2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity.
  • Such alterations are selected from the group consisting of at least one nucleic acid deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5 ' or 3 ' terminal positions of the reference polynucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleic acids in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of nucleic acid alterations for a given percent identity is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or: n n ⁇ x n - (x n • y),
  • n n is the number of amino acid alterations
  • x n is the total number of amino acids in
  • y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85 % etc., • is the symbol for the multiplication operator, and wherein any non-integer product of x n and y is rounded down to the nearest integer prior to subtracting it from x n .
  • Polypeptide embodiments further include an isolated polypeptide comprising a polypeptide having at least a 50,60, 70, 80, 85, 90, 95, 97 or 100% identity to a polypeptide reference sequence of SEQ ID NO:2, wherein said polypeptide sequence may be identical to the reference sequence of SEQ ID NO: 2 or may include up to a certain integer number of amino acid alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of amino acid alterations is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID
  • n a is the number of amino acid alterations
  • x a is the total number of amino acids in
  • y is 0.50 for 50%, 0.60 for 60%, 0.70 for 70%, 0.80 for 80%, 0.85 for 85 % , 0.90 for 90% , 0.95 for 95 %, 0.97 for 97% or 1.00 for 100%, and • is the symbol for the multiplication operator, and wherein any non-integer product of x a and y is rounded down to the nearest integer prior to subtracting it from x a .
  • a polypeptide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity.
  • Such alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or: n a ⁇ x a - (x a - y),
  • n a is the number of amino acid alterations
  • x a is the total number of amino acids in SEQ ID NO:2
  • y is, for instance 0.70 for 70 % , 0.80 for 80 % , 0.85 for 85 % etc.
  • is the symbol for the multiplication operator, and wherein any non-integer product of x a and y is rounded down to the nearest integer prior to subtracting it from x a .
  • Fusion protein refers to a protein encoded by two, often unrelated, fused genes or fragments thereof.
  • EP-A-0 464 discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • an immunoglobulin Fc region as a part of a fusion protein is advantageous for use in therapy and diagnosis resulting in, for example, improved pharmacokinetic properties [see, e.g., EP-A 0232 262].
  • ADDRESSEE Ratner & Prestia
  • B STREET: P.O. Box 980
  • TELECOMMUNICATION INFORMATION (A) TELEPHONE: 610-407-0700 (B) TELEFAX: 610-407-0701
  • TCCGAGAGAA CAGGCCTGTC TCAGGCAGGC CCTGCGCCTC CTATGCGGAG ATGCTACTGC 60 CACTGCTGCT GTCCTCGCTG CTGGGCGGGT CCCAGGCTAT GGATGGGAGA TTCTGGATAC 120
  • GACTCATCTC AACGGCATTC TCCAACGGAG CGTTTCTGGG AATCGGCATC ACGGCTCTTC 1560 TTTTCCTCTG CCTGGCCCTG ATCATCATGA AGATTCTACC GAAGAGACGG ACTCAGACAG 1620
  • CTCTTTAAAA AACATCTGGC CAGGCAGCAC AGTGGCTCAC GCCTGTAATC CCAGCACTTT 2160 GGGAGGTTGA GGTGGGCAGA TCGCCTGAGG TCGGGAGTTC GAGACCAGCC TGGCCAACTT 2220
  • MOLECULE TYPE protein
  • TTATCAGCAT TTCACGTGAC AACACGCCAG CCCTGGAGCC CCAGCCCCAG GGAAATGTCC 660
  • GCAGCTGCTC CTCCCAGGCC AGCCCGGCCC CCTCTCTGCG CTGGTGGCTT GGGGAGGAGC 1320
  • GACTCATCTC AACGGCATTC TCCAACGGAG CGTTTCTGGG AATCGGCATC ACGGCTCTTC 1560

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Immunology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pulmonology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Diabetes (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Psychology (AREA)
  • Vascular Medicine (AREA)
  • Virology (AREA)
  • Dermatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)

Abstract

The SAF-4 polypeptides and polynucleotides and methods for producing such polypeptides by recombinant techniques are disclosed. Also disclosed are methods for utilizing SAF-4 polypeptides and polynucleotides in therapy, and diagnostic assays for such.

Description

Sialoadhesin Family 4 cDNA
Field of the Invention
This invention relates to newly identified polypeptides and polynucleotides encoding such polypeptides, to their use in therapy and in identifying compounds which may be agonists, antagonists and /or inhibitors which are potentially useful in therapy, and to production of such polypeptides and polynucleotides.
Background of the Invention
The drug discovery process is currently undergoing a fundamental revolution as it embraces 'functional genomics' , that is, high throughput genome- or gene-based biology. This approach is rapidly superceding earher approaches based on 'positional cloning' . A phenotype, that is a biological function or genetic disease, would be identified and this would then be tracked back to the responsible gene, based on its genetic map position.
Functional genomics relies heavily on the various tools of bioinformatics to identify gene sequences of potential interest from the many molecular biology databases now available. There is a continuing need to identify and characterize further genes and their related polypeptides/proteins, as targets for drug discovery.
Summary of the Invention
The present invention relates to SAF-4, in particular SAF-4 polypeptides and SAF-4 polynucleotides, recombinant materials and methods for their production. In another aspect, the invention relates to methods for using such polypeptides and polynucleotides, including the treatment of cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotrøphic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reper usion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormalities, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, hereinafter referred to as "the Diseases", amongst others. In a further aspect, the invention relates to methods for identifying agomsts and antagonists/inhibitors using the materials provided by the invention, and treating conditions associated with SAF-4 imbalance with the identified compounds. In a still further aspect, the invention relates to diagnostic assays for detecting diseases associated with inappropriate SAF-4 activity or levels. Description of the Invention
In a first aspect, the present invention relates to SAF-4 polypeptides. Such peptides include isolated polypetides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99 % identity, to that of SEQ ID NO: 2 over the entire length of SEQ ID NO: 2. Such polypeptides include those comprising the amino acid of SEQ ID NO:2.
Further peptides of the present invention include isolated polypeptides in which the amino acid sequence has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO: 2 over the entire length of SEQ ID NO: 2. Such polypeptides include the polypeptide of SEQ ID NO:2.
Further peptides of the present invention include isolated polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 1. Polypeptides of the present invention are believed to be members of the Sialoadhesin family of polypeptides. They are therefore of interest because the sialoadhesin family of proteins, sialoadhesin, CD33, CD22 and myelin-associated glycoprotein (MAG), are utilized as cellular interaction molecules. They bind specific carbohydrates in a sialic acid-dependent manner on target cells. The extracellular domain is made up of various numbers of immunoglobulin-like domains of the N-like and C2-like subtypes and the intracellular portion has no known homology to any signalling motifs. Sialoadhesin expression is restricted to macrophages, it has 17 Ig-like domains and the specific recognition sequence on target cells is Νeu5Aca2,3Galbl3GalΝAc. Known target cells are developing myeloid cells in the bone marrow and lymphocytes in the spleen and lymph node (Crocker, P. R. , et al. EMBO J, 1994, 13:4490-4503). CD22 is expressed only on B cells and has a and b isoforms with 5 and 7 Ig-like domains, respectively. CD22 is known to bind T cells, B cells, monocytes, granulocytes and erythrocytes by recognizing Neu5Aca2,6Galbl,4Glc(NAc) in N-linked glycans (Crocker, P. R., et al. EMBO J, 1994, 13:4490-4503; Sta enkovic, I. and Seed, B. Nature, 1990, 345:74-77; Wilson, G. L., et al. J Exp Med, 1991, 173:137-146). Myelin- associated glycoprotein (MAG) is expressed by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system and is thought to participate in the cell adhesion to axons. MAG has two alternatively spliced variants, large MAG (L-MAG) and small MAG (S-MAG) which are expressed either during embryonic development or in the adult, respectively. The alternative splicing results in the expression of the same extracellular domains but distinct intracellular domains (Pedraza, L. et al., JCB, 1990, 111:2651-2661).
CD33 is most relevant to SAF-4 because they are the most closely related of all the family members. CD33 is normally expressed on the developing myelomonocytic lineage. It is absent on early stem cells but is present on colony-forming units for granulocytes, erythrocytes, monocytes, and megakaryocytes (CFU-GEMM) and progenitors of granulocytes and mononuclear phagocytes (CFU-GM). It is downregulated by mature granulocytes but retained by mature monocytes and macrophages (Andrews, R. G., et al., Blood, 1983, 62:124; Griffin, J. D., et al., Leuk Res 1984, 8:521). CD33 has two Ig-like domains and prefers to bind targets expressing NeuAca2,3Gal in N- and O-linked glycans. It maps to chromosome 19ql3.1-13.3, closely linking it in the genome with MAG and CD22 (Freeman, S. D., et al., Blood, 1995, 85:2005-2012).
CD33 has also been found to be expressed on about 85 % of leukemic myeloblasts in patients with acute myelogenous leukemia (AML) and is frequently used to differentiate AML from acute lymphoblastic leukemia (ALL). Monoclonal antibodies to CD33 have been used therapeutically to purge residual myeloblasts from autologous bone marrow grafts ex vivo for the treatment of AML (Robertson, M. J., et al., Blood, 1992 79:2229-2236). More recently, humanized monoclonal antibodies to CD33 have undergone evaluation in vivo for the treatment of AML (Caron, P. C, et al., Blood, 1994, 83:1760-1768). These properties are hereinafter referred to as "SAF-4 activity" or "SAF-4 polypeptide activity" or "biological activity of SAF-4" . Also included amongst these activities are antigenic and immunogenic activities of said SAF-4 polypeptides, in particular the antigenic and immunogenic activities of the polypeptide of SEQ ID NO:2. Preferably, a polypeptide of the present invention exhibits at least one biological activity of SAF-4.
The polypeptides of the present invention may be in the form of the "mature" protein or may be a part of a larger protein such as a fusion protein. It is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production.
The present invention also includes include variants of the aforementioned polypetides, that is polypeptides that vary from the referents by conservative amino acid substitutions, whereby a residue is substituted by another with like characteristics. Typical such substitutions are among Ala, Nal, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr. Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 amino acids are substituted, deleted, or added in any combination.
Polypeptides of the present invention can be prepared in any suitable manner. Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art. In a further aspect, the present invention relates to SAF-4 polynucleotides. Such polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to the amino acid sequence of SEQ ID ΝO:2, over the entire length of SEQ ID NO:2. In this regard, polypeptides which have at least 97% identity are highly preferred, whilst those with at least 98-99 % identity are more highly preferred, and those with at least 99 % identity are most highly preferred. Such polynucleotides include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO:l encoding the polypeptide of SEQ ID NO:2. Further polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence that has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO: 2, over the entire coding region. In this regard, polynucleotides which have at least 97 % identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99 % identity are most highly preferred.
Further polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, to SEQ ID NO: 1 over the entire length of SEQ ID NO: 1. In this regard, polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identiy are more highly preferred, and those with at least 99 % identity are most highly preferred. Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO: 1 as well as the polynucleotide of SEQ ID NO: 1.
The invention also provides polynucleotides which are complementary to all the above described polynucleotides.
The nucleotide sequence of SEQ ID NO:l shows homology with CD33 (Simmons, D., and Seed, B., JI 141:2797-2800, 1988). The nucleotide sequence of SEQ ID NO:l is a cDNA sequence and comprises a polypeptide encoding sequence (nucleotide 51 to 1970) encoding a polypeptide of 639 amino acids, the polypeptide of SEQ ID NO:2. The nucleotide sequence encoding the polypeptide of SEQ ID NO: 2 may be identical to the polypeptide encoding sequence contained in SEQ ID NO:l or it may be a sequence other than the one contained in SEQ ID NO: 1 , which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ID NO:2. The polypeptide of the SEQ ID NO: 2 is structurally related to other proteins of the Sialoadhesin family, having homology and/or structural similarity withCD33 (Simmons, D., and Seed, B., JI 141:2797-2800, 1988).
Preferred polypeptides and polynucleotides of the present invention are expected to have, inter alia, similar biological functions/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides and polynucleotides of the present invention have at least one SAF-4 activity.
The present invention also relates to partial or other polynucleotide and polypeptide sequences which were first identified prior to the determination of the corresponding full length sequences of SEQ ID NO: 1 and SEQ ED NO:2.
Accordingly, in a further aspect, the present invention provides for an isolated polynucleotide comprising:
(a) a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity to SEQ ID NO: 3 over the entire length of SEQ ID NO: 3;
(b) a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity, to SEQ ID NO: 3 over the entire length of SEQ ID NO:3;
(c) the polynucleotide of SEQ ID NO: 3; or
(d) a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, even more preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO:4, over the entire length of SEQ ID NO:4; as well as the polynucleotide of SEQ ID NO: 3.
The present invention further provides for a polypeptide which:
(a) comprises an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99 % identity, to that of SEQ ID NO:4 over the entire length of SEQ ID NO:4;
(b) has an amino acid sequence which is at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95 % identity, most preferably at least 97-99% identity, to the amino acid sequence of SEQ ID NO: 4 over the entire length of SEQ ID NO:4;
(c) comprises the amino acid of SEQ ID NO: 4; and
(d) is the polypeptide of SEQ ID NO:4; as well as polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 3.
The nucleotide sequence of SEQ ID NO: 3 and the peptide sequence encoded thereby are derived from EST (Expressed Sequence Tag) sequences. It is recognized by those skilled in the art that there will inevitably be some nucleotide sequence reading errors in EST sequences (see Adams, M.D. et al, Nature 377 (supp) 3, 1995).
Accordingly, the nucleotide sequence of SEQ ID NO: 3 and the peptide sequence encoded therefrom are therefore subject to the same inherent limitations in sequence accuracy. Furthermore, the peptide sequence encoded by SEQ ID NO: 3 comprises a region of identity or close homology and/or close structural similarity (for example a conservative amino acid difference) with the closest homologous or structurally similar protein.
Polynucleotides of the present invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of human primary dendritic cells, using the expressed sequence tag (EST) analysis (Adams, M.D., et al. Science (1991) 252:1651-1656; Adams, M.D. et al , Nature, (1992) 355:632-634; Adams, M.D. , et al. , Nature (1995) 377 Supp:3-174). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesized using well known and commercially available techniques.
When polynucleotides of the present invention are used for the recombinant production of polypeptides of the present invention, the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide in reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions. For example, a marker sequence which facilitates purification of the fused polypeptide can be encoded. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, Inc.) and described in Gentz et al. , Proc Natl Acad Sci USA (1989) 86:821-824, or is an HA tag. The polynucleotide may also contain non-coding 5' and 3' sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ribosome binding sites and sequences that stabilize mRNA.
Further embodiments of the present invention include polynucleotides encoding polypeptide variants which comprise the amino acid sequence of SEQ ID NO:2 and in which several, for instance from 5 to 10, 1 to 5, 1 to 3, 1 to 2 or 1, amino acid residues are substituted, deleted or added, in any combination. Polynucleotides which are identical or sufficiently identical to a nucleotide sequence contained in SEQ ID NO: 1 , may be used as hybridization probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides of the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding homologs and orthologs from species other than human) that have a high sequence similarity to SEQ ID NO: 1. Typically these nucleotide sequences are 70% identical, preferably 80% identical, more preferably 90% identical, most preferably 95 % identical to that of the referent. The probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides. Particularly preferred probes will have between 30 and 50 nucleotides.
A polynucleotide encoding a polypeptide of the present invention, including homologs and orthologs from species other than human, may be obtained by a process which comprises the steps of screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof; and isolating full- length cDNA and genomic clones containing said polynucleotide sequence. Such hybridization techniques are well known to the skilled artisan. Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in 0. Ix SSC at about 65°C. Thus the present invention also includes polynucleotides obtainable by screening an appropriate library under stingent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof.
The skilled artisan will appreciate that, in many cases, an isolated cDNA sequence will be incomplete, in that the region coding for the polypeptide is cut short at the 5 ' end of the cDNA. This is a consequence of reverse transcriptase, an enzyme with inherently low 'processivity' (a measure of the ability of the enzyme to remain attached to the template during the polymerization reaction), failing to complete a DNA copy of the mRNA template during 1st strand cDNA synthesis. There are several methods available and well known to those skilled in the art to obtain full-length cDNAs, or extend short cDNAs, for example those based on the method of Rapid Amplification of cDNA ends (RACE) (see, for example, Frohman et al., PNAS USA 85, 8998-9002, 1988). Recent modifications of the technique, exemplified by the Marathon™' technology (Clontech Laboratories Inc.) for example, have significantly simplified the search for longer cDNAs. In the Marathon™ technology, cDNAs have been prepared from mRNA extracted from a chosen tissue and an 'adaptor' sequence ligated onto each end. Nucleic acid amplification (PCR) is then carried out to amplify the 'missing' 5' end of the cDNA using a combination of gene specific and adaptor specific oligonucleotide primers. The PCR reaction is then repeated using 'nested' primers, that is, primers designed to anneal within the amplified product (typically an adaptor specific primer that anneals further 3' in the adaptor sequence and a gene specific primer that anneals further 5' in the known gene sequence). The products of this reaction can then be analysed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or carrying out a separate full-length PCR using the new sequence information for the design of the 5' primer.
Recombinant polypeptides of the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, in a further aspect, the present invention relates to expression systems which comprise a polynucleotide or polynucleotides of the present invention, to host cells which are genetically engineered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. For recombinant production, host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention. Introduction of polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al. , Basic Methods in Molecular Biology (1986) and Sambrook et al. , Molecular Cloning: A Laboratory Manual, 2nd Ed. , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989). Preferred such methods include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection.
Representative examples of appropriate hosts include bacterial cells, such as streptococci, staphylococci, E. coli, Streptomyces and Bacillus subtilis cells; fiingal cells, such as yeast cells and Aspergillus cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells; and plant cells.
A great variety of expression systems can be used, for instance, chromosomal, episomal and virus-derived systems, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids. The expression systems may contain control regions that regulate as well as engender expression. Generally, any system or vector which is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used. The appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook et al. , MOLECULAR CLONING, A LABORATORY MANUAL (supra). Appropriate secretion signals may be incorporated into the desired polypeptide to allow secretion of the translated protein into the lumen of the endoplasmic reticulum, the periplasmic space or the extracellular environment. These signals may be endogenous to the polypeptide or they may be heterologous signals. If a polypeptide of the present invention is to be expressed for use in screening assays, it is generally preferred that the polypeptide be produced at the surface of the cell. In this event, the cells may be harvested prior to use in the screening assay. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the polypeptide. If produced intracellularly, the cells must first be lysed before the polypeptide is recovered.
Polypeptides of the present invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
This invention also relates to the use of polynucleotides of the present invention as diagnostic reagents. Detection of a mutated form of the gene characterized by the polynucleotide of SEQ ID NO: 1 which is associated with a dysfunction 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 the gene. Individuals carrying mutations in the gene may be detected at the DNA level by a variety of techniques.
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 SAF-4 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 (ee, e.g., Myers et al. , Science (1985) 230: 1242). Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (see Cotton et al. , Proc NatlAcad Sci USA (1985) 85: 4397-4401). In another embodiment, an array of oligonucleotides probes comprising SAF-4 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 the Diseases through detection of mutation in the SAF-4 gene by the methods described. In addition, such diseases may be diagnosed by methods comprising determining from a sample derived from a subject an abnormally decreased or increased level of polypeptide or mRNA. Decreased or increased expression can be measured at the RNA level using any of the methods well known in the art for the quantitation of polynucleotides, such as, for example, nucleic acid amplification, for instance PCR, RT-PCR, RNase protection, Northern blotting and other hybridization methods. Assay techniques that can be used to determine levels of a protein, such as a polypeptide of the present 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 and flow cytometric analysis.
Thus in another aspect, the present invention relates to a diagonostic kit which comprises:
(a) a polynucleotide of the present invention, preferably the nucleotide sequence of SEQ ID NO: 1, or a fragment thereof ; (b) a nucleotide sequence complementary to that of (a);
(c) a polypeptide of the present invention, preferably the polypeptide of SEQ ID NO: 2 or a fragment thereof; or
(d) an antibody to a polypeptide of the present invention, preferably to the polypeptide of SEQ ID NO:2.
It will be appreciated that in any such kit, (a), (b), (c) or (d) may comprise a substantial component. Such a kit will be of use in diagnosing a disease or suspectability to a disease, particularly cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotrophic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reperfusion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormahties, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, amongst others.
The nucleotide sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome. The mapping of relevant sequences to chromosomes according to the present invention is an important first step in correlating those sequences with gene associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).
The differences in the cDNA or genomic sequence between affected and unaffected individuals can also be determined. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
The polypeptides of the invention or their fragments or analogs thereof, or cells expressing them, can also be used as immunogens to produce antibodies immunospecific for polypeptides of the present invention. The term "immunospecific" means that the antibodies have substantially greater affinity for the polypeptides of the invention than their affinity for other related polypeptides in the prior art.
Antibodies generated against polypeptides of the present invention may be obtained by administering the polypeptides or epitope-bearing fragments, analogs or cells to an animal, preferably a non-human animal, using routine protocols. For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature (1975) 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al. , Immunology Today (1983) 4:72) and the EBN-hybridoma technique (Cole et al. , MONOCLONAL ANTIBODIES AND CANCER THERAPY, pp. 77-96, Alan R. Liss, Inc. , 1985).
Techniques for the production of single chain antibodies, such as those described in U.S. Patent No. 4,946,778, can also be adapted to produce single chain antibodies to polypeptides of this invention. Also, transgenic mice, or other organisms, including other mammals, may be used to express humanized antibodies.
The above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or to purify the polypeptides by affinity chromatography.
Antibodies against polypeptides of the present invention may also be employed to treat the Diseases, amongst others. Antibodies against SAF-4 polypeptides may also be employed to subcharacterize cell populations during hematopoietic development, as a diagnostic marker to distinguish between different forms of cancer, to purge bone marrow ex vivo of cancer cells expressing SAF-4, as a tool to aid in the ex vivo expansion (proliferation and/or differentiation) of hematopoietic progenitor cells expressing SAF-4, as a stimulus in vivo for stem cell mobilization into the periphery, and as an in vivo chemoprotective agent. In a further aspect, the present invention relates to genetically engineered soluble fusion proteins comprising a polypeptide of the present invention, or a fragment thereof, and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclasses (IgG, IgM, IgA, IgE). Preferred as an immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgGl , where fusion takes place at the hinge region. In a particular embodiment, the Fc part can be removed simply by incorporation of a cleavage sequence which can be cleaved with blood clotting factor Xa. Furthermore, this invention relates to processes for the preparation of these fusion proteins by genetic engineering, and to the use thereof for drug screening, diagnosis and therapy. In another approach, soluble forms of SAF-4 polypeptides still capable of binding the ligand in competition with endogenous SAF-4 may be administered. Typical embodiments of such competitors comprise fragments of the SAF-4 polypeptide. One example is using the extracellular domain of SAF-4 fused to a human immunoglobulin Fc region which can then be employed to treat cancer, inflammation, autoimmunity and allergy, among others. SAF-4/Fc polypeptides may also be employed to purge bone marrow ex vivo of cancer cells expressing SAF-4 ligands, as a tool to aid in the ex vivo expansion (proliferation and/or differentiation) of hematopoietic progenitor cells expressing SAF-4 ligands, as a stimulus in vivo for stem cell mobilization into the periphery, and as an in vivo chemoprotective agent. A further aspect of the invention also relates to polynucleotides encoding such fusion proteins. Examples of fusion protein technology can be found in International Patent Application Nos. WO94/29458 and WO94/22914.
Another aspect of the invention relates to a method for inducing an immunological response in a mammal which comprises inoculating the mammal with a polypeptide of the present invention, adequate to produce antibody and/or T cell immune response to protect said animal from the Diseases hereinbefore mentioned, amongst others. Yet another aspect of the invention relates to a method of inducing immunological response in a mammal which comprises, delivering a polypeptide of the present invention via a vector directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce antibody to protect said animal from diseases. A further aspect of the invention relates to an immunological/vaccine formulation (composition) which, when introduced into a mammalian host, induces an immunological response in that mammal to a polypeptide of the present invention wherein the composition comprises a polypeptide or polynucleotide of the present invention. The vaccine formulation may further comprise a suitable carrier. Since a polypeptide may be broken down in the stomach, it is preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection). Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation instonic with the blood of the recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use. The vaccine formulation may also include adjuvant systems for enhancing the immunogenicity of the formulation, such as oil-in water systems and other systems known in the art. The dosage will depend on the specific activity of the vaccine and can be readily determined by routine experimentation.
Polypeptides of the present invention are responsible for many biological functions, including many disease states, in particular the Diseases hereinbefore mentioned. It is therefore desirous to devise screening methods to identify compounds which stimulate or which inhibit the function of the polypeptide. Accordingly, in a further aspect, the present invention provides for a method of screening compounds to identify those which stimulate or which inhibit the function of the polypeptide. In general, agonists or antagonists may be employed for therapeutic and prophylactic purposes for such Diseases as hereinbefore mentioned. Compounds may be identified from a variety of sources, for example, cells, cell- free preparations, chemical libraries, and natural product mixtures. Such agonists, antagonists or inhibitors so-identified may be natural or modified substrates, ligands, receptors, enzymes, etc. , as the case may be, of the polypeptide; or may be structural or functional mimetics thereof (see Coligan et al. , Current Protocols in Immunology l(2):Chapter 5 (1991)). The screening method may simply measure the binding of a candidate compound to the polypeptide, or to cells or membranes bearing the polypeptide, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound. Alternatively, the screening method may involve competition with a labeled competitor. Further, these screening methods may test whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells bearing the polypeptide. 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. Constitutively active polpypeptides may be employed in screening methods for inverse agomsts or inhibitors, in the absence of an agonist or inhibitor, by testing whether the candidate compound results in inhibition of activation of the polypeptide. Further, the screening methods may simply comprise the steps of mixing a candidate compound with a solution containing a polypeptide of the present invention, to form a mixture, measuring SAF-4 activity in the mixture, and comparing the SAF-4 activity of the mixture to a standard. Fusion proteins, such as those made from Fc portion and SAF-4 polypeptide, as hereinbefore described, can also be used for high-throughput screening assays to identify antagonists for the polypeptide of the present invention (see D. Bennett et al., J Mol Recognition, 8:52-58 (1995); and K. Johanson et al, J Biol Chem, 270(16):9459-9471 (1995)). The polynucleotides, polypeptides and antibodies to the polypeptide of the present invention may also be used to configure screening methods for detecting the effect of added compounds on the production of mRNA and polypeptide in cells. For example, an ELISA assay may be constructed for measuring secreted or cell associated levels of polypeptide using monoclonal and polyclonal antibodies by standard methods known in the art. This can be used to discover agents which may inhibit or enhance the production of polypeptide (also called antagonist or agonist, respectively) from suitably manipulated cells or tissues.
The polypeptide may be used to identify membrane bound or soluble receptors, if any, through standard receptor binding techniques known in the art. These include, but are not limited to, ligand binding and crosslinking assays in which the polypeptide is labeled with a radioactive isotope (for instance, ^I), chemically modified (for instance, biotinylated), or fused to a peptide sequence suitable for detection or purification, and incubated with a source of the putative receptor (cells, cell membranes, cell supernatants, tissue extracts, bodily fluids). Other methods include biophysical techniques such as surface plasmon resonance and spectroscopy. These screening methods may also be used to identify agonists and antagonists of the polypeptide which compete with the binding of the polypeptide to its receptors, if any. Standard methods for conducting such assays are well understood in the art.
Examples of potential polypeptide antagonists include antibodies or, in some cases, ohgonucleotides or proteins which are closely related to the ligands, substrates, receptors, enzymes, etc. , as the case may be, of the polypeptide, e.g. , a fragment of the ligands, substrates, receptors, enzymes, etc. ; or small molecules which bind to the polypetide of the present invention but do not elicit a response, so that the activity of the polypeptide is prevented. Thus, in another aspect, the present invention relates to a screening kit for identifying agonists, antagonists, Ugands, receptors, substrates, enzymes, etc. for polypeptides of the present invention; or compounds which decrease or enhance the production of such polypeptides, which comprises:
(a) a polypeptide of the present invention; (b) a recombinant cell expressing a polypeptide of the present invention;
(c) a cell membrane expressing a polypeptide of the present invention; or
(d) antibody to a polypeptide of the present invention; which polypeptide is preferably that of SEQ ID NO:2.
It will be appreciated that in any such kit, (a), (b), (c) or (d) may comprise a substantial component.
It will be readily appreciated by the skilled artisan that a polypeptide of the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by: (a) determining in the first instance the three-dimensional structure of the polypeptide;
(b) deducing the three-dimensional structure for the likely reactive or binding site(s) of an agonist, antagonist or inhibitor;
(c) synthesizing candidate compounds that are predicted to bind to or react with the deduced binding or reactive site; and
(d) testing whether the candidate compounds are indeed agomsts, antagonists or inhibitors.
It will be further appreciated that this will normally be an interactive process.
In a further aspect, the present invention provides methods of treating abnormal conditions such as, for instance, cancer, inflammation, autoimmunity, allergy, asthma, rheumatoid arthritis, CNS inflammation, cerebellar degeneration, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amylotrophic lateral sclerosis, head injury damage, and other neurological abnormalities, septic shock, sepsis, stroke, osteoporosis, osteoarthritis, ischemia reperfusion injury, cardiovascular disease, kidney disease, liver disease, ischemic injury, myocardial infarction, hypotension, hypertension, AIDS, myelodysplastic syndromes and other hematologic abnormahties, aplastic anemia, male pattern baldness, and bacterial, fungal, protozoan and viral infections, related to either an excess of, or an under-expression of, SAF-4 polypeptide activity.
If the activity of the polypeptide is in excess, several approaches are available. One approach comprises administering to a subject in need thereof an inhibitor compound (antagonist) as hereinabove described, optionally in combination with a pharmaceutically acceptable carrier, in an amount effective to inhibit the function of the polypeptide, such as, for example, by blocking the binding of ligands, substrates, receptors, enzymes, etc. , or by inhibiting a second signal, and thereby alleviating the abnormal condition. In another approach, soluble forms of the polypeptides still capable of binding the ligand, substrate, enzymes, receptors, etc. in competition with endogenous polypeptide may be administered. Typical examples of such competitors include fragments of the SAF-4 polypeptide. In still another approach, expression of the gene encoding endogenous SAF-4 polypeptide 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)). Alternatively, oligonucleotides which form triple helices with the gene can be supplied (see, for example, Lee et al , Nucleic Acids Res (1979) 6:3073; 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. For treating abnormal conditions related to an under-expression of SAF-4 and its activity, several approaches are also available. One approach comprises administering to a subject a therapeutically effective amount of a compound which activates a polypeptide of the present invention, i.e. , an agonist as described above, in combination with a pharmaceutically acceptable carrier, to thereby alleviate the abnormal condition. Alternatively, gene therapy may be employed to effect the endogenous production of SAF-4 by the relevant cells in the subject. For example, 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. For an overview of 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) . Another approach is to administer a therapeutic amount of a polypeptide of the present invention in combination with a suitable pharmaceutical carrier.
In a further aspect, the present invention provides for pharmaceutical compositions comprising a therapeutically effective amount of a polypeptide, such as the soluble form of a polypeptide of the present invention, agonist antagonist peptide or small molecule compound, in combination with a pharmaceutically acceptable carrier or excipient. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
The composition will be adapted to the route of administration, for instance by a systemic or an oral route. Preferred forms of systemic administration include injection, typically by intravenous injection. Other injection routes, such as subcutaneous, intramuscular, or intraperitoneal, can be used. Alternative means for systemic administration include transmucosal and transdermal administration using penetrants such as bile salts or fusidic acids or other detergents. In addition, if a polypeptide or other compounds of the present invention can be formulated in an enteric or an encapsulated formulation, oral administration may also be possible. Administration of these compounds may also be topical and or localized, in the form of salves, pastes, gels, and the like.
The dosage range required depends on the choice of peptide or other compounds of the present invention, the route of administration, the nature of the formulation, the nature of the subject's condition, and the judgment of the attending practitioner. Suitable dosages, however, are in the range of 0.1-100 μg/kg of subject. Wide variations in the needed dosage, however, are to be expected in view of the variety of compounds available and the differing efficiencies of various routes of administration. For example, oral administration would be expected to require higher dosages than administration by intravenous injection. Variations in these dosage levels can be adjusted using standard empirical routines for optimization, as is well understood in the art. Polypeptides used in treatment can also be generated endogenously in the subject, in treatment modalities often referred to as "gene therapy" as described above. Thus, for example, cells from a subject may be engineered with a polynucleotide, such as a DNA or RNA, to encode a polypeptide ex vivo, and for example, by the use of a retroviral plasmid vector. The cells are then introduced into the subject. Polynucleotide and polypeptide sequences form a valuable information resource with which to identify further sequences of similar homology. This is most easily facilitated by storing the sequence in a computer readable medium and then using the stored data to search a sequence database using well known searching tools, such as GCC. Accordingly, in a further aspect, the present invention provides for a computer readable medium having stored thereon a polynucleotide comprising the sequence of SEQ ID NO: 1 and/or a polypeptide sequence encoded thereby.
The following definitions are provided to facilitate understanding of certain terms used frequently hereinbefore. "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. For example, 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. In addition,
"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. A variety of modifications may be made to DNA and RNA; thus, "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 ohgonucleotides. "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 post-translational 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 may 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 to 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 post-translation 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 (see, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York, 1993; Wold, F., Post-translational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, 1983; Seifter et al. , "Analysis for protein modifications and nonprotein cofactors", Meth Enzymol (1990) 182:626-646 and Rattan et al. , "Protein Synthesis: Post-translational Modifications and Aging", Ann NY Acad Sci (1992) 663:48-62).
"Variant" refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, 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," as known in the art, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as the case may be, as determined by comparing the sequences. In the art, "identity" also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. "Identity" can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A.M., ed. , Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A.M. , and Griffin, H.G. , eds. , Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G. , Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D. , SIAM /. Applied Math. , 48: 1073 (1988). Methods to determine identity are designed to give the largest match between the sequences tested. Moreover, methods to determine identity are codified in pubhcly available computer programs. Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux, J. , et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S.F. et al., J. Molec. Biol 215: 403-410 (1990). The BLAST X program is pubhcly available from NCBI and other sources (BLAST Manual, Altschul, S. , et al , NCBI NLM NIH Bethesda, MD 20894; Altschul, S. , et al , J. Mol. Biol 215: 403- 410 (1990). The well known Smith Waterman algorithm may also be used to determine identity. Parameters for polypeptide sequence comparison include the following:
1) Algorithm: Needleman and Wunsch, J. Mol Biol. 48: 443-453 (1970)
Comparison matrix: BLOSSUM62 from Hentikoff and Hentikoff, Proc. Natl. Acad. Sci. USA. 89:10915-10919 (1992)
Gap Penalty: 12 Gap Length Penalty: 4
A program useful with these parameters is pubhcly available as the "gap" program from Genetics Computer Group, Madison WI. The aforementioned parameters are the default parameters for peptide comparisons (along with no penalty for end gaps).
Parameters for polynucleotide comparison include the following: 1) Algorithm: Needleman and Wunsch, J. Mol Biol. 48: 443-453 (1970)
Comparison matrix: matches = +10, mismatch = 0
Gap Penalty: 50 Gap Length Penalty: 3
Available as: The "gap" program from Genetics Computer Group, Madison WI. These are the default parameters for nucleic acid comparisons.
A preferred meaning for "identity" for polynucleotides and polypeptides, as the case may be, are provided in (1) and (2) below.
(1) Polynucleotide embodiments further include an isolated polynucleotide comprising a polynucleotide sequence having at least a 50, 60, 70, 80, 85, 90, 95, 97 or 100% identity to the reference sequence of SEQ ID NO:l, wherein said polynucleotide sequence may be identical to the reference sequence of SEQ ID NO: 1 or may include up to a certain integer number of nucleotide alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ ID NO: 1 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of nucleotides in SEQ ID NO:l, or: nn < xn - (xn • y),
wherein nn is the number of nucleotide alterations, xn is the total number of nucleotides in
SEQ ID NO:l, y is 0.50 for 50%, 0.60 for 60% , 0.70 for 70%, 0.80 for 80%, 0.85 for 85 %, 0.90 for 90%, 0.95 for 95 %, 0.97 for 97% or 1.00 for 100% , and • is the symbol for the multiplication operator, and wherein any non-integer product of xn and y is rounded down to the nearest integer prior to subtracting it from xn. Alterations of a polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 may create nonsense, missense or frameshift mutations in this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations. By way of example, a polynucleotide sequence of the present invention may be identical to the reference sequence of SEQ ID NO: 2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity. Such alterations are selected from the group consisting of at least one nucleic acid deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5 ' or 3 ' terminal positions of the reference polynucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleic acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of nucleic acid alterations for a given percent identity is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or: nn < xn - (xn • y),
wherein nn is the number of amino acid alterations, xn is the total number of amino acids in
SEQ ID NO:2, y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85 % etc., • is the symbol for the multiplication operator, and wherein any non-integer product of xn and y is rounded down to the nearest integer prior to subtracting it from xn.
(2) Polypeptide embodiments further include an isolated polypeptide comprising a polypeptide having at least a 50,60, 70, 80, 85, 90, 95, 97 or 100% identity to a polypeptide reference sequence of SEQ ID NO:2, wherein said polypeptide sequence may be identical to the reference sequence of SEQ ID NO: 2 or may include up to a certain integer number of amino acid alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence, and wherein said number of amino acid alterations is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO: 2, or: na < xa - (xa - y),
wherein na is the number of amino acid alterations, xa is the total number of amino acids in
SEQ ID NO:2, y is 0.50 for 50%, 0.60 for 60%, 0.70 for 70%, 0.80 for 80%, 0.85 for 85 % , 0.90 for 90% , 0.95 for 95 %, 0.97 for 97% or 1.00 for 100%, and • is the symbol for the multiplication operator, and wherein any non-integer product of xa and y is rounded down to the nearest integer prior to subtracting it from xa.
By way of example, a polypeptide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2, that is it may be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the percent identity is less than 100% identity. Such alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in SEQ ID NO: 2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of amino acids in SEQ ID NO:2, or: na < xa - (xa - y),
wherein na is the number of amino acid alterations, xa is the total number of amino acids in SEQ ID NO:2, y is, for instance 0.70 for 70 % , 0.80 for 80 % , 0.85 for 85 % etc. , and • is the symbol for the multiplication operator, and wherein any non-integer product of xa and y is rounded down to the nearest integer prior to subtracting it from xa. "Fusion protein" refers to a protein encoded by two, often unrelated, fused genes or fragments thereof. In one example, EP-A-0 464 discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, employing an immunoglobulin Fc region as a part of a fusion protein is advantageous for use in therapy and diagnosis resulting in, for example, improved pharmacokinetic properties [see, e.g., EP-A 0232 262]. On the other hand, for some uses it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected and purified.
All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual pubhcation were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
SEQUENCE INFORMATION SEQ ED NO:l
TCCGAGAGAACAGGCCTGTCTCAGGCAGGCCCTGCGCCTCCTATGCGGAGATGCTACTGCCACTGCTGCTGTCC TCGCTGCTGGGCGGGTCCCAGGCTATGGATGGGAGATTCTGGATACGAGTGCAGGAGTCAGTGATGGTGCCGGA GGGCCTGTGCATCTCTGTGCCCTGCTCTTTCTCCTACCCCCGACAAGACTGGACAGGGTCTACCCCAGCTTATG GCTACTGGTTCAAAGCΑGTGACTGAGAC^ICCΑAGGGTGCTCCTGTGGCCΑCAAACCACCAGAGTCGAGAGGTG GAAATGAGCACCCGGGGCCGATTCCAGCTCACTGGGGATCCCGCCAAGGGGAACTGCTCCTTGGTGATCAGAGA CGCGCAGATGCAGGATGAGTCACAGTACTTCTTTCGGGTGGAGAGAGGAAGCTATGTGAGATATAATTTCATGA ACGATGGGTTCTTTCTAAAAGTAACAGTGCTCAGCTTCACGCCCAGACCCCAGGACCACAACACCGACCTCACC TGCCATGTGGACTTCTCCAGAAAGGGTGTGAGCGCACAGAGGACCGTCCGACTCCGTGTGGCCTATGCCCCCAG AGACCTTGTTATCAGCATTTCACGTGACAACACGCCAGCCCTGGAGCCCCAGCCCCAGGGAAATGTCCCATACC TGGAAGCCCAAAAAGGCCAGTTCCTGCGGCTCCTCTGTGCTGCTGACAGCCAGCCCCCTGCCACACTGAGCTGG GTCCTGCAGAACAGAGTCCTCTCCTCGTCCCATCCCTGGGGCCCTAGACCCCTGGGGCTGGAGCTGCCCGGGGT GAAGGCTGGGGATTCAGGGCGCTACACCTGCCGAGCGGAGAACAGGCTTGGCTCCCAGCAGCGAGCCCTGGACC TCTCTGTGCAGTATCCTCCAGAGAACCTGAGAGTGATGGTTTCCCAAGCAAACAGGACAGTCCTGGAAAACCTT GGGAACGGCACGTCTCTCCCAGTACTGGAGGGCCAAAGCCTGTGCCTGGTCTGTGTCACACACAGCAGCCCCCC AGCCAGGCTGAGCTGGACCCAGAGGGGACAGGTTCTGAGCCCCTCCCAGCCCTCAGACCCCGGGGTCCTGGAGC TGCCTCGGGTTCAAGTGGAGCACGAAGGAGAGTTCACCTGCCACGCTCGGCACCCACTGGGCTCCCAGCACGTC TCTCTCAGCCTCTCCGTGCACTACTCCCCGAAGCTGCTGGGCCCCTCCTGCTCCTGGGAGGCTGAGGGTCTGCA CTGCAGCTGCTCCTCCCAGGCCAGCCCGGCCCCCTCTCTGCGCTGGTGGCTTGGGGAGGAGCTGCTGGAGGGGA ACAGCAGCCAGGACTCCTTCGAGGTCACCCCCAGCTCAGCCGGGCCCTGGGCCAACAGCTCCCTGAGCCTCCAT GGAGGGCTCAGCTCCGGCCTCAGGCTCCGCTGTGAGGCCTGGAACGTCCATGGGGCCCAGAGTGGATCCATCCT GCAGCTGCCAGATAAGAAGGGACTCATCTCAACGGCATTCTCCAACGGAGCGTTTCTGGGAATCGGCATCACGG CTCTTCTTTTCCTCTGCCTGGCCCTGATCATCATGAAGATTCTACCGAAGAGACGGACTCAGACAGAAACCCCG AGGCCCAGGTTCTCCCGGCACAGCACGATCCTGGATTACATCAATGTGGTCCCGACGGCTGGCCCCCTGGCTCA GAAGCGGAATCAGAAAGCCACACCAAACAGTCCTCGGACCCCTCTTCCACCAGGTGCTCCCTCCCCAGAATCAA AGAAGAACC1AGAAAAAGC-AGTATCAGTTGCCC-AGTTTCCCAGAACCCAAATCATCCACTCAAGCCCCAGAATCC CAGGAGAGCCAAGAGGAGCTCCATTATGCCACGCTCAACTTCCCAGGCGTCAGACCCAGGCCTGAGGCCCGGAT GCCCAAGGGCACCCAGGCGGATTATGCAGAAGTCAAGTTCCAATGAGGGTCTCTTAGGCTTTAGGACTGGGACT TCGGCTAGGGAGGAAGGTAGAGTAAGAGGTTGAAGATAACAGAGTGCAAAGTTTCCTTCTCTCCCTCTCTCTCT CTCTTTCTCTCTCTCTCTCTCTTTCTCTCTCTTTAAAAAACATCTGGCCAGGCAGCACAGTGGCTCACGCCTGT AATCCCAGCACTTTGGGAGGTTGAGGTGGGCAGATCGCCTGAGGTCGGGAGTTCGAGACCAGCCTGGCCAACTT GGTGAAACCCCGTCTCTACTAAAAATACAAAAATTAGCTGGGCATGGTGGCAGGCGCCTGTAATCCTACCTACT TGGGAAGCTGAGGC1AGGAGAATCΑCT GAACCTGGGAGACGGAGGTTGCAGTGAGCCAAGATCACACCATTGCA CGCCΑGCCTGGGC^^CΛAAGCGAGACTCC-ATCTC^AAAAAAAAATCCTCCTVAATGGGTTGGGTGTCTGTAATCC CAGCACTTTGGGAGGCTAAGGTGGGTGGATTGCTTGAGCCCAGGAGTTCGAGACCAGCCTGGGCAACATGGTGA AACCCCATCTCTACAAAAAATACAAAACATAGCTGGGCTTGGTGGTGTGTGCCTGTAAGTCCCCAGCTGTCAGA CATTTAAACCAGAGCAACTCCATCXTGGAATAGGAGGCTGAATAAAATGAGGCTGAGACCTXACTGGGCTGCAT TCTCCΆGACAGTGGAGGCATTCTAAGTCACAGGATGAGACAGGAGGTCCCGTACAAGATACΆGGTCATAAAGAC TTTGCTGATAAAACAGATTGCAGTAAΆGAAGCCAACCAAATCCCACCAAAACCAAGTTGGCCACGAGAGTGACC TCTGGTCGTCCTCACTGCTACIACTCCTGAI^GCΑCCATGAC-AGTTTACAAATGCCATGGCAACATCAGGAAGTT ACCCGATATGTCCCΑAAAGGGGGAGGAATGAATAATCCΑCCCCTTGTTTAGC-AAATAAGCAAGAAATAACCATA AAAGTGGGCAACCAGCAGCTCTAGGCGCTGCTCTTGTCCTATGGAGTAGCCTTCTTTTTTCCTTTAACTTTCTT GATAAACTTGCTTTCACCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA
SEQ ID NO:2
MLLPLLLSSLLGGSQAMDGRF IRVQESVMVPEGLCISVPCSFSYPRQD TGSTPAYGYWFKAVTETTKGAPVA TNHQSREVEMSTRGRFQLTGDPAKGNCSLVIRDAQMQDESQYFFRVERGSYVRYNFMNDGFFLKVTVLSFTPRP QDHNTDLTCHVDFSRKGVSAQRTVRLRVAYAPRDLVISISRDNTPA EPQPQGNVPYLEAQKGQFLRL CAADS QPPATLS VLQNRV SSSHP GPRPLGLELPGVKAGDSGRYTCRAENR GSQQRALDLSVQYPPENLRVMVSQA NRTVLENLGNGTSLPVLEGQSLCLVCVTHSSPPARLS TQRGQV SPSQPSDPGVLELPRVQVEHEGEFTCHAR HPLGSQHVSLSLSVHYSPK LGPSCS EAEGLHCSCSSQASPAPSLR LGEELLEGNSSQDSFEVTPSSAGPW ANSS SLHGGLSSGLRLRCEA VHGAQSGSILQ PDKKGLISTAFSNGAFLGIGITA LFLCIiA II KI PK RRTQTETPRPRFSRHSTILDYINWPTAGP AQKRNQKATPNSPRTPLPPGAPSPESKKNQKKQYQLPSFPEPK SSTQAPESQESQEELHYATLNFPGVRPRPEARMPKGTQADYAEVKFQ.
SEQ ID NO:3
TCCGAGAGAACAGGCCTGTCTCAGGCAGGCCCTGCGCCTCCTATGCGGAGATGCTACTGCCACTGCTGCTGTCC TCGCTGCTGGGCGGGTCCCAGGCTATGGATGGGAGATTCTGGATACGAGTGCAGGAGTCAGTGATGGTGCCGGA GGGCCTGTGCATCTCTGTGCCCTGCTCTTTCTCCTACCCCCGACAAGACTGGACAGGGTCTACCCCAGCTTATG GCTACTGGTTCAAAGCAGTGACTGAGACAACCAAGGGTGCTCCTGTGGCCACAAACCACCAGAGTCGAGAGGTG GAAATGAGCACCCGGGGCCGATTCCAGCTCACTGGGGATCCCGCCAAGGGGAACTGCTCCTTGGTGATCAGAGA CGCGCAGATGCAGGATGAGTCACAGTACTTCTTTCGGGTGGAGAGAGGAAGCTATGTGAGATATAATTTCATGA ACGATGGGTTCTTTCTAAAAGTAACAGTGCTCAGCTTCACGCCCAGACCCCAGGACCACAACACCGACCTCACC TGCCATGTGGACTTCTCCAGAAAGGGTGTGAGCGCACAGAGGACCGTCCGACTCCGTGTGGCCTATGCCCCCAG AGACCTTGTTATCAGCAT TCACGTGACAACACGCCAGCCCTGGAGCCCCAGCCCCAGGGAAATGTCCCATACC TGGAAGCCCAAAAAGGCCAGTTCCTGCGGCTCCTCTGTGCTGCTGACAGCCAGCCCCCTGCCACACTGAGCTGG GTCCTGCAGAACAGAGTCCTCTCCTCGTCCCATCCCTGGGGCCCTAGACCCCTGGGGCTGGAGCTGCCCGGGGT GAAGGCTGGGGATTCAGGGCGCTACACCTGCCGAGCGGAGAACAGGCTTGGCTCCCAGCAGCGAGCCCTGGACC TCTCTGTGCAGTATCCTCCAGAGAACCTGAGAGTGATGGTTTCCC^^GCAAACAGGACAGTCCTGGAAAACCTT GGGAACGGCACGTCTCTCCCAGTACTGGAGGGCCAAAGCCTGTGCCTGGTCTGTGTCACACACAGCAGCCCCCC AGCCAGGCTGAGCTGGACCCAGAGGGGACAGGTTCTGAGCCCCTCCCAGCCCTCAGACCCCGGGGTCCTGGAGC TGCCTCGGGTTCAAGTGGAGCACGAAGGAGAGTTCACCTGCCACGCTCGGCACCCACTGGGCTCCCAGCACGTC TCTCTCAGCCTCTCCGTGCACTACTCCCCGAAGCTGCTGGGCCCCTCCTGCTCCTGGGAGGCTGAGGGTCTGCA CTGCAGCTGCTCCTCCCAGGCCAGCCCGGCCCCCTCTCTGCGCTGGTGGCTTGGGGAGGAGCTGCTGGAGGGGA ACAGCAGCCAGGACTCCTTCGAGGTCACCCCCAGCTCAGCCGGGCCCTGGGCCAACAGCTCCCTGAGCCTCCAT GGAGGGCTCAGCTCCGGCCTCAGGCTCCGCTGTGAGGCCTGGAACGTCCATGGGGCCCAGAGTGGATCCATCCT GCAGCTGCCAGATAAGAAGGGACTCATCTCAACGGCATTCTCCAACGGAGCGTTTCTGGGAATCGGCATCACGG CTCTTCTTTTCCTCTGCCTGGCCCTGATCATCATGAAGATTCTACCGAAGAGACGGACTCAGACAGAAACCCCG AGGCCCAGGTTCTCCCGGCACAGCACGATCCTGGATTACATCAATGTGGTCCCGACGGCTGGCCCCCTGGCTCA GAAGCGGAATCAGAAAGCCACACCAAACAGTCCTCGGACCCCTCTTCCACCAGGTGCTCCCTCCCCAGAATCAA AGAAGAACCΆGAAAAAGCAGTATCΆGTTGCCCAGTTTCCC-AGAACCCAAATCATCCACTCAAGCCCCAGAATCC CAGGAGAGCCAAGAGGAGCTCCATTATGCCACGCTCAACTTCCCAGGCGTCAGACCCAGGCCTGAGGCCCGGAT GCCCAAGGGCACCCAGGCGGATTATGCAGAAGTCAAGTTCCAATGAGGGTCTCTTAGGCTTTAGGACTGGGACT TCGGCTAGGGAGGAAGGTAGAGTAAGAGGTTGAAGATAACAGAGTGCAAAGTTTCCTTCTCTCCCTCTCTCTCT CTCTTTCTCTCTCTCTCTCTCTTTCTCTCTCTTTAAAAAACATCTGGCCAGGCAGCACAGTGGCTCACGCCTGT AATCCCAGCACTTTGGGAGGTTGAGGTGGGCAGATCGCCTGAGGTCGGGAGTTCGAGACCAGCCTGGCCAACTT GGTGAAACCCCGTCTCTACTAAAAATACAAAAATTAGCTGGGCATGGTGGCAGGCGCCTGTAATCCTACCTACT TGGGAAGCTGAGGCAGGAGAATCACTTGAACCTGGGAGACGGAGGTTGCAGTGAGCCAAGATCACACCATTGCA CGCCAGCCTGGGCAACAAAGCGAGACTCCATCTCAAAAAAAAAATCCTCCAAATGGGTTGGGTGTCTGTAATCC CAGCACTTTGGGAGGCTAAGGTGGGTGGATTGCTTGAGCCCAGGAGTTCGAGACCAGCCTGGGCAACATGGTGA AACCCCATCTCTACAAAAAATACAAAACATAGCTGGGCTTGGTGGTGTGTGCCTGTAAGTCCCCAGCTGTCAGA CATTTAAACCAGAGCAACTCCATCXTGGAATAGGAGGCTGAATAAAATGAGGCTGAGACCTXACTGGGCTGCAT TCTCCAGACAGTGGAGGCATTCTAAGTCACAGGATGAGACAGGAGGTCCCGTACAAGATACAGGTCATAAAGAC TTTGCTGATAAAACAGATTGCAGTAAAGAAGCCAACCAAATCCCACCAAAACCAAGTTGGCCACGAGAGTGACC TCTGGTCGTCCTCIACTGCTACACTCCTGACAGC-ACCATGACAGTTTACAAATGCCATGGCAACATCAGGAAGTT ACCCGATATGTCCCAAAAGGGGGAGGAATGAATAATCCACCCCTTGTTTAGCAAATAAGCAAGAAATAACCATA AAAGTGGGC-AACCAGCAGCTCTAGGCGCTGCTCTTGTCCTATGGAGTAGCCTTCTTTTTTCCTTTAACTTTCTT GATAAACTTGCTTT(^CCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA SEQ ID NO:4 IMJLPLLLSSLLGGSQAMDGRF IRVQESVMVPEGLCISVPCSFSYPRQD TGSTPAYGYWFKAVTETTKGAPVA TISRAQSREVEMSTRGRFQLTGDPAKGNCSLVIRDAQMQDESQYFFRVERGSYVRYNFMNDGFFLKVTVLSFTPRP QDHNTDLTCHVDFSRKGVSAQRTVRLRVAYAPRDLVISISRDNTPALEPQPQGNVPYLEAQKGQFLRLLC-AADS QPPATLS V QNR\ΠL,SSSHP GPRPLGLELPGVKAGDSGRYTCRAENRLGSQQRADLSVQYPPENLRVMVSQA NRTVLENLGNGTSLPVLEGQSLCVCVTHSSPPARLS TQRGQVLSPSQPSDPGVLELPRVQVEHEGEFTCHAR HPLGSPARLSQPLRALPEAAGPLLLLGG* SEQUENCE LISTING
(1) GENERAL INFORMATION
(i) APPLICANT: SmithKline Beecham Corporation (ii) TITLE OF THE INVENTION: Sialoadhesin Family 4 cDNA (iii) NUMBER OF SEQUENCES: 4
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Ratner & Prestia (B) STREET: P.O. Box 980
(C) CITY: Valley Forge
(D) STATE: PA
(E) COUNTRY: USA
(F) ZIP: 19482
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Diskette
(B) COMPUTER: IBM Compatible
(C) OPERATING SYSTEM: DOS (D) SOFTWARE: FastSEQ for Windows Version 2
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: TO BE ASSIGNED
(B) FILING DATE: 26-MAY-1998 (C) CLASSIFICATION: UNKNOWN
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 60/047,572
(B) FILING DATE: 27 -MAY- 1997
(viii) ATTORNEY/AGENT INFORMATION: (A) NAME: Prestia, Paul F (B) REGISTRATION NUMBER: 23,031
(C) REFERENCE/DOCKET NUMBER: GH-7002G
(ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 610-407-0700 (B) TELEFAX: 610-407-0701
(C) TELEX:
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3099 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS : single (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
TCCGAGAGAA CAGGCCTGTC TCAGGCAGGC CCTGCGCCTC CTATGCGGAG ATGCTACTGC 60 CACTGCTGCT GTCCTCGCTG CTGGGCGGGT CCCAGGCTAT GGATGGGAGA TTCTGGATAC 120
GAGTGCAGGA GTCAGTGATG GTGCCGGAGG GCCTGTGCAT CTCTGTGCCC TGCTCTTTCT 180
CCTACCCCCG ACAAGACTGG ACAGGGTCTA CCCCAGCTTA TGGCTACTGG TTCAAAGCAG 240
TGACTGAGAC AACCAAGGGT GCTCCTGTGG CCACAAACCA CCAGAGTCGA GAGGTGGAAA 300
TGAGCACCCG GGGCCGATTC CAGCTCACTG GGGATCCCGC CAAGGGGAAC TGCTCCTTGG 360 TGATCAGAGA CGCGCAGATG CAGGATGAGT CACAGTACTT CTTTCGGGTG GAGAGAGGAA 420
GCTATGTGAG ATATAATTTC ATGAACGATG GGTTCTTTCT AAAAGTAACA GTGCTCAGCT 480
TCACGCCCAG ACCCCAGGAC CACAACACCG ACCTCACCTG CCATGTGGAC TTCTCCAGAA 540
AGGGTGTGAG CGCACAGAGG ACCGTCCGAC TCCGTGTGGC CTATGCCCCC AGAGACCTTG 600
TTATCAGCAT TTCACGTGAC AACACGCCAG CCCTGGAGCC CCAGCCCCAG GGAAATGTCC 660 CATACCTGGA AGCCCAAAAA GGCCAGTTCC TGCGGCTCCT CTGTGCTGCT GACAGCCAGC 720
CCCCTGCCAC ACTGAGCTGG GTCCTGCAGA ACAGAGTCCT CTCCTCGTCC CATCCCTGGG 780
GCCCTAGACC CCTGGGGCTG GAGCTGCCCG GGGTGAAGGC TGGGGATTCA GGGCGCTACA 840
CCTGCCGAGC GGAGAACAGG CTTGGCTCCC AGCAGCGAGC CCTGGACCTC TCTGTGCAGT 900
ATCCTCCAGA GAACCTGAGA GTGATGGTTT CCCAAGCAAA CAGGACAGTC CTGGAAAACC 960 TTGGGAACGG CACGTCTCTC CCAGTACTGG AGGGCCAAAG CCTGTGCCTG GTCTGTGTCA 1020
CACACAGCAG CCCCCCAGCC AGGCTGAGCT GGACCCAGAG GGGACAGGTT CTGAGCCCCT 1080
CCCAGCCCTC AGACCCCGGG GTCCTGGAGC TGCCTCGGGT TCAAGTGGAG CACGAAGGAG 1140
AGTTCACCTG CCACGCTCGG CACCCACTGG GCTCCCAGCA CGTCTCTCTC AGCCTCTCCG 1200
TGCACTACTC CCCGAAGCTG CTGGGCCCCT CCTGCTCCTG GGAGGCTGAG GGTCTGCACT 1260 GCAGCTGCTC CTCCCAGGCC AGCCCGGCCC CCTCTCTGCG CTGGTGGCTT GGGGAGGAGC 1320
TGCTGGAGGG GAACAGCAGC CAGGACTCCT TCGAGGTCAC CCCCAGCTCA GCCGGGCCCT 1380
GGGCCAACAG CTCCCTGAGC CTCCATGGAG GGCTCAGCTC CGGCCTCAGG CTCCGCTGTG 1440
AGGCCTGGAA CGTCCATGGG GCCCAGAGTG GATCCATCCT GCAGCTGCCA GATAAGAAGG 1500
GACTCATCTC AACGGCATTC TCCAACGGAG CGTTTCTGGG AATCGGCATC ACGGCTCTTC 1560 TTTTCCTCTG CCTGGCCCTG ATCATCATGA AGATTCTACC GAAGAGACGG ACTCAGACAG 1620
AAACCCCGAG GCCCAGGTTC TCCCGGCACA GCACGATCCT GGATTACATC AATGTGGTCC 1680
CGACGGCTGG CCCCCTGGCT CAGAAGCGGA ATCAGAAAGC CACACCAAAC AGTCCTCGGA 1740
CCCCTCTTCC ACCAGGTGCT CCCTCCCCAG AATCAAAGAA GAACCAGAAA AAGCAGTATC 1800
AGTTGCCCAG TTTCCCAGAA CCCAAATCAT CCACTCAAGC CCCAGAATCC CAGGAGAGCC 1860 AAGAGGAGCT CCATTATGCC ACGCTCAACT TCCCAGGCGT CAGACCCAGG CCTGAGGCCC 1920
GGATGCCCAA GGGCACCCAG GCGGATTATG CAGAAGTCAA GTTCCAATGA GGGTCTCTTA 1980
GGCTTTAGGA CTGGGACTTC GGCTAGGGAG GAAGGTAGAG TAAGAGGTTG AAGATAACAG 2040
AGTGCAAAGT TTCCTTCTCT CCCTCTCTCT CTCTCTTTCT CTCTCTCTCT CTCTTTCTCT 2100
CTCTTTAAAA AACATCTGGC CAGGCAGCAC AGTGGCTCAC GCCTGTAATC CCAGCACTTT 2160 GGGAGGTTGA GGTGGGCAGA TCGCCTGAGG TCGGGAGTTC GAGACCAGCC TGGCCAACTT 2220
GGTGAAACCC CGTCTCTACT AAAAATACAA AAATTAGCTG GGCATGGTGG CAGGCGCCTG 2280
TAATCCTACC TACTTGGGAA GCTGAGGCAG GAGAATCACT TGAACCTGGG AGACGGAGGT 2340
TGCAGTGAGC CAAGATCACA CCATTGCACG CCAGCCTGGG CAACAAAGCG AGACTCCATC 2400
TCAAAAAAAA AATCCTCCAA ATGGGTTGGG TGTCTGTAAT CCCAGCACTT TGGGAGGCTA 2460 AGGTGGGTGG ATTGCTTGAG CCCAGGAGTT CGAGACCAGC CTGGGCAACA TGGTGAAACC 2520
CCATCTCTAC AAAAAATACA AAACATAGCT GGGCTTGGTG GTGTGTGCCT GTAAGTCCCC 2580
AGCTGTCAGA CATTTAAACC AGAGCAACTC CATCXTGGAA TAGGAGGCTG AATAAAATGA 2640
GGCTGAGACC TXACTGGGCT GCATTCTCCA GACAGTGGAG GCATTCTAAG TCACAGGATG 2700
AGACAGGAGG TCCCGTACAA GATACAGGTC ATAAAGACTT TGCTGATAAA ACAGATTGCA 2760 GTAAAGAAGC CAACCAAATC CCACCAAAAC CAAGTTGGCC ACGAGAGTGA CCTCTGGTCG 2820
TCCTCACTGC TACACTCCTG ACAGCACCAT GACAGTTTAC AAATGCCATG GCAACATCAG 2880
GAAGTTACCC GATATGTCCC AAAAGGGGGA GGAATGAATA ATCCACCCCT TGTTTAGCAA 2940
ATAAGCAAGA AATAACCATA AAAGTGGGCA ACCAGCAGCT CTAGGCGCTG CTCTTGTCCT 3000
ATGGAGTAGC CTTCTTTTTT CCTTTAACTT TCTTGATAAA CTTGCTTTCA CCTAAAAAAA 3060 AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAA 3099 (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 639 amino acids (B) TYPE: amino acid
(C) STRANDEDNESS : single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:
Met Leu Leu Pro Leu Leu Leu Ser Ser Leu Leu Gly Gly Ser Gin Ala
1 5 10 15
Met Asp Gly Arg Phe Trp He Arg Val Gin Glu Ser Val Met Val Pro 20 25 30
Glu Gly Leu Cys He Ser Val Pro Cys Ser Phe Ser Tyr Pro Arg Gin
35 40 45
Asp Trp Thr Gly Ser Thr Pro Ala Tyr Gly Tyr Trp Phe Lys Ala Val 50 55 60 Thr Glu Thr Thr Lys Gly Ala Pro Val Ala Thr Asn His Gin Ser Arg 65 70 75 80
Glu Val Glu Met Ser Thr Arg Gly Arg Phe Gin Leu Thr Gly Asp Pro
85 90 95
Ala Lys Gly Asn Cys Ser Leu Val He Arg Asp Ala Gin Met Gin Asp 100 105 110
Glu Ser Gin Tyr Phe Phe Arg Val Glu Arg Gly Ser Tyr Val Arg Tyr
115 120 125
Asn Phe Met Asn Asp Gly Phe Phe Leu Lys Val Thr Val Leu Ser Phe
130 135 140 Thr Pro Arg Pro Gin Asp His Asn Thr Asp Leu Thr Cys His Val Asp
145 150 155 160
Phe Ser Arg Lys Gly Val Ser Ala Gin Arg Thr Val Arg Leu Arg Val
165 170 175
Ala Tyr Ala Pro Arg Asp Leu Val He Ser He Ser Arg Asp Asn Thr 180 185 190
Pro Ala Leu Glu Pro Gin Pro Gin Gly Asn Val Pro Tyr Leu Glu Ala
195 200 205
Gin Lys Gly Gin Phe Leu Arg Leu Leu Cys Ala Ala Asp Ser Gin Pro
210 215 220 Pro Ala Thr Leu Ser Trp Val Leu Gin Asn Arg Val Leu Ser Ser Ser
225 230 235 240
His Pro Trp Gly Pro Arg Pro Leu Gly Leu Glu Leu Pro Gly Val Lys
245 250 255
Ala Gly Asp Ser Gly Arg Tyr Thr Cys Arg Ala Glu Asn Arg Leu Gly 260 265 270
Ser Gin Gin Arg Ala Leu Asp Leu Ser Val Gin Tyr Pro Pro Glu Asn
275 280 285
Leu Arg Val Met Val Ser Gin Ala Asn Arg Thr Val Leu Glu Asn Leu
290 295 300 Gly Asn Gly Thr Ser Leu Pro Val Leu Glu Gly Gin Ser Leu Cys Leu
305 310 315 320
Val Cys Val Thr His Ser Ser Pro Pro Ala Arg Leu Ser Trp Thr Gin
325 330 335
Arg Gly Gin Val Leu Ser Pro Ser Gin Pro Ser Asp Pro Gly Val Leu 340 345 350
Glu Leu Pro Arg Val Gin Val Glu His Glu Gly Glu Phe Thr Cys His 355 360 365
Ala Arg His Pro Leu Gly Ser Gin His Val Ser Leu Ser Leu Ser Val
370 375 380
His Tyr Ser Pro Lys Leu Leu Gly Pro Ser Cys Ser Trp Glu Ala Glu 385 390 395 400
Gly Leu His Cys Ser Cys Ser Ser Gin Ala Ser Pro Ala Pro Ser Leu
405 410 415
Arg Trp Trp Leu Gly Glu Glu Leu Leu Glu Gly Asn Ser Ser Gin Asp 420 425 430 Ser Phe Glu Val Thr Pro Ser Ser Ala Gly Pro Trp Ala Asn Ser Ser 435 440 445
Leu Ser Leu His Gly Gly Leu Ser Ser Gly Leu Arg Leu Arg Cys Glu
450 455 460
Ala Trp Asn Val His Gly Ala Gin Ser Gly Ser He Leu Gin Leu Pro 465 470 475 480
Asp Lys Lys Gly Leu He Ser Thr Ala Phe Ser Asn Gly Ala Phe Leu
485 490 495
Gly He Gly He Thr Ala Leu Leu Phe Leu Cys Leu Ala Leu He He 500 505 510 Met Lys He Leu Pro Lys Arg Arg Thr Gin Thr Glu Thr Pro Arg Pro 515 520 525
Arg Phe Ser Arg His Ser Thr He Leu Asp Tyr He Asn Val Val Pro
530 535 540
Thr Ala Gly Pro Leu Ala Gin Lys Arg Asn Gin Lys Ala Thr Pro Asn 545 550 555 560
Ser Pro Arg Thr Pro Leu Pro Pro Gly Ala Pro Ser Pro Glu Ser Lys
565 570 575
Lys Asn Gin Lys Lys Gin Tyr Gin Leu Pro Ser Phe Pro Glu Pro Lys 580 585 590 Ser Ser Thr Gin Ala Pro Glu Ser Gin Glu Ser Gin Glu Glu Leu His 595 600 605
Tyr Ala Thr Leu Asn Phe Pro Gly Val Arg Pro Arg Pro Glu Ala Arg
610 615 620
Met Pro Lys Gly Thr Gin Ala Asp Tyr Ala Glu Val Lys Phe Gin 625 630 635
(2) INFORMATION FOR SEQ ID NO: 3:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 3099 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS : single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
TCCGAGAGAA CAGGCCTGTC TCAGGCAGGC CCTGCGCCTC CTATGCGGAG ATGCTACTGC 60
CACTGCTGCT GTCCTCGCTG CTGGGCGGGT CCCAGGCTAT GGATGGGAGA TTCTGGATAC 120 GAGTGCAGGA GTCAGTGATG GTGCCGGAGG GCCTGTGCAT CTCTGTGCCC TGCTCTTTCT 180
CCTACCCCCG ACAAGACTGG ACAGGGTCTA CCCCAGCTTA TGGCTACTGG TTCAAAGCAG 240
TGACTGAGAC AACCAAGGGT GCTCCTGTGG CCACAAACCA CCAGAGTCGA GAGGTGGAAA 300
TGAGCACCCG GGGCCGATTC CAGCTCACTG GGGATCCCGC CAAGGGGAAC TGCTCCTTGG 360
TGATCAGAGA CGCGCAGATG CAGGATGAGT CACAGTACTT CTTTCGGGTG GAGAGAGGAA 420 GCTATGTGAG ATATAATTTC ATGAACGATG GGTTCTTTCT AAAAGTAACA GTGCTCAGCT 480
TCACGCCCAG ACCCCAGGAC CACAACACCG ACCTCACCTG CCATGTGGAC TTCTCCAGAA 540 AGGGTGTGAG CGCACAGAGG ACCGTCCGAC TCCGTGTGGC CTATGCCCCC AGAGACCTTG 600
TTATCAGCAT TTCACGTGAC AACACGCCAG CCCTGGAGCC CCAGCCCCAG GGAAATGTCC 660
CATACCTGGA AGCCCAAAAA GGCCAGTTCC TGCGGCTCCT CTGTGCTGCT GACAGCCAGC 720
CCCCTGCCAC ACTGAGCTGG GTCCTGCAGA ACAGAGTCCT CTCCTCGTCC CATCCCTGGG 780 GCCCTAGACC CCTGGGGCTG GAGCTGCCCG GGGTGAAGGC TGGGGATTCA GGGCGCTACA 840
CCTGCCGAGC GGAGAACAGG CTTGGCTCCC AGCAGCGAGC CCTGGACCTC TCTGTGCAGT 900
ATCCTCCAGA GAACCTGAGA GTGATGGTTT CCCAAGCAAA CAGGACAGTC CTGGAAAACC 960
TTGGGAACGG CACGTCTCTC CCAGTACTGG AGGGCCAAAG CCTGTGCCTG GTCTGTGTCA 1020
CACACAGCAG CCCCCCAGCC AGGCTGAGCT GGACCCAGAG GGGACAGGTT CTGAGCCCCT 1080 CCCAGCCCTC AGACCCCGGG GTCCTGGAGC TGCCTCGGGT TCAAGTGGAG CACGAAGGAG 1140
AGTTCACCTG CCACGCTCGG CACCCACTGG GCTCCCAGCA CGTCTCTCTC AGCCTCTCCG 1200
TGCACTACTC CCCGAAGCTG CTGGGCCCCT CCTGCTCCTG GGAGGCTGAG GGTCTGCACT 1260
GCAGCTGCTC CTCCCAGGCC AGCCCGGCCC CCTCTCTGCG CTGGTGGCTT GGGGAGGAGC 1320
TGCTGGAGGG GAACAGCAGC CAGGACTCCT TCGAGGTCAC CCCCAGCTCA GCCGGGCCCT 1380 GGGCCAACAG CTCCCTGAGC CTCCATGGAG GGCTCAGCTC CGGCCTCAGG CTCCGCTGTG 1440
AGGCCTGGAA CGTCCATGGG GCCCAGAGTG GATCCATCCT GCAGCTGCCA GATAAGAAGG 1500
GACTCATCTC AACGGCATTC TCCAACGGAG CGTTTCTGGG AATCGGCATC ACGGCTCTTC 1560
TTTTCCTCTG CCTGGCCCTG ATCATCATGA AGATTCTACC GAAGAGACGG ACTCAGACAG 1620
AAACCCCGAG GCCCAGGTTC TCCCGGCACA GCACGATCCT GGATTACATC AATGTGGTCC 1680 CGACGGCTGG CCCCCTGGCT CAGAAGCGGA ATCAGAAAGC CACACCAAAC AGTCCTCGGA 1740
CCCCTCTTCC ACCAGGTGCT CCCTCCCCAG AATCAAAGAA GAACCAGAAA AAGCAGTATC 1800
AGTTGCCCAG TTTCCCAGAA CCCAAATCAT CCACTCAAGC CCCAGAATCC CAGGAGAGCC 1860
AAGAGGAGCT CCATTATGCC ACGCTCAACT TCCCAGGCGT CAGACCCAGG CCTGAGGCCC 1920
GGATGCCCAA GGGCACCCAG GCGGATTATG CAGAAGTCAA GTTCCAATGA GGGTCTCTTA 1980 GGCTTTAGGA CTGGGACTTC GGCTAGGGAG GAAGGTAGAG TAAGAGGTTG AAGATAACAG 2040
AGTGCAAAGT TTCCTTCTCT CCCTCTCTCT CTCTCTTTCT CTCTCTCTCT CTCTTTCTCT 2100
CTCTTTAAAA AACATCTGGC CAGGCAGCAC AGTGGCTCAC GCCTGTAATC CCAGCACTTT 2160
GGGAGGTTGA GGTGGGCAGA TCGCCTGAGG TCGGGAGTTC GAGACCAGCC TGGCCAACTT 2220
GGTGAAACCC CGTCTCTACT AAAAATACAA AAATTAGCTG GGCATGGTGG CAGGCGCCTG 2280 TAATCCTACC TACTTGGGAA GCTGAGGCAG GAGAATCACT TGAACCTGGG AGACGGAGGT 2340
TGCAGTGAGC CAAGATCACA CCATTGCACG CCAGCCTGGG CAACAAAGCG AGACTCCATC 2400
TCAAAAAAAA AATCCTCCAA ATGGGTTGGG TGTCTGTAAT CCCAGCACTT TGGGAGGCTA 2460
AGGTGGGTGG ATTGCTTGAG CCCAGGAGTT CGAGACCAGC CTGGGCAACA TGGTGAAACC 2520
CCATCTCTAC AAAAAATACA AAACATAGCT GGGCTTGGTG GTGTGTGCCT GTAAGTCCCC 2580 AGCTGTCAGA CATTTAAACC AGAGCAACTC CATCXTGGAA TAGGAGGCTG AATAAAATGA 2640
GGCTGAGACC TXACTGGGCT GCATTCTCCA GACAGTGGAG GCATTCTAAG TCACAGGATG 2700
AGACAGGAGG TCCCGTACAA GATACAGGTC ATAAAGACTT TGCTGATAAA ACAGATTGCA 2760
GTAAAGAAGC CAACCAAATC CCACCAAAAC CAAGTTGGCC ACGAGAGTGA CCTCTGGTCG 2820
TCCTCACTGC TACACTCCTG ACAGCACCAT GACAGTTTAC AAATGCCATG GCAACATCAG 2880 GAAGTTACCC GATATGTCCC AAAAGGGGGA GGAATGAATA ATCCACCCCT TGTTTAGCAA 2940
ATAAGCAAGA AATAACCATA AAAGTGGGCA ACCAGCAGCT CTAGGCGCTG CTCTTGTCCT 3000
ATGGAGTAGC CTTCTTTTTT CCTTTAACTT TCTTGATAAA CTTGCTTTCA CCTAAAAAAA 3060
AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAA 3099 (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 399 amino acids
(B) TYPE: amino acid (C) STRANDEDNESS : single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: Met Leu Leu Pro Leu Leu Leu Ser Ser Leu Leu Gly Gly Ser Gin Ala 1 5 10 15
Met Asp Gly Arg Phe Trp He Arg Val Gin Glu Ser Val Met Val Pro
20 25 30
Glu Gly Leu Cys He Ser Val Pro Cys Ser Phe Ser Tyr Pro Arg Gin 35 40 45
Asp Trp Thr Gly Ser Thr Pro Ala Tyr Gly Tyr Trp Phe Lys Ala Val
50 55 60
Thr Glu Thr Thr Lys Gly Ala Pro Val Ala Thr Asn His Gin Ser Arg 65 70 75 80 Glu Val Glu Met Ser Thr Arg Gly Arg Phe Gin Leu Thr Gly Asp Pro
85 90 95
Ala Lys Gly Asn Cys Ser Leu Val He Arg Asp Ala Gin Met Gin Asp
100 105 110
Glu Ser Gin Tyr Phe Phe Arg Val Glu Arg Gly Ser Tyr Val Arg Tyr 115 120 125
Asn Phe Met Asn Asp Gly Phe Phe Leu Lys Val Thr Val Leu Ser Phe
130 135 140
Thr Pro Arg Pro Gin Asp His Asn Thr Asp Leu Thr Cys His Val Asp 145 150 155 160 Phe Ser Arg Lys Gly Val Ser Ala Gin Arg Thr Val Arg Leu Arg Val
165 170 175
Ala Tyr Ala Pro Arg Asp Leu Val He Ser He Ser Arg Asp Asn Thr
180 185 190
Pro Ala Leu Glu Pro Gin Pro Gin Gly Asn Val Pro Tyr Leu Glu Ala 195 200 205
Gin Lys Gly Gin Phe Leu Arg Leu Leu Cys Ala Ala Asp Ser Gin Pro
210 215 220
Pro Ala Thr Leu Ser Trp Val Leu Gin Asn Arg Val Leu Ser Ser Ser 225 230 235 240 His Pro Trp Gly Pro Arg Pro Leu Gly Leu Glu Leu Pro Gly Val Lys
245 250 255
Ala Gly Asp Ser Gly Arg Tyr Thr Cys Arg Ala Glu Asn Arg Leu Gly
260 265 270
Ser Gin Gin Arg Ala Leu Asp Leu Ser Val Gin Tyr Pro Pro Glu Asn 275 280 285
Leu Arg Val Met Val Ser Gin Ala Asn Arg Thr Val Leu Glu Asn Leu
290 295 300
Gly Asn Gly Thr Ser Leu Pro Val Leu Glu Gly Gin Ser Leu Cys Leu 305 310 315 320 Val Cys Val Thr His Ser Ser Pro Pro Ala Arg Leu Ser Trp Thr Gin
325 330 335
Arg Gly Gin Val Leu Ser Pro Ser Gin Pro Ser Asp Pro Gly Val Leu
340 345 350
Glu Leu Pro Arg Val Gin Val Glu His Glu Gly Glu Phe Thr Cys His 355 360 365
Ala Arg His Pro Leu Gly Ser Pro Ala Arg Leu Ser Gin Pro Leu Arg
370 375 380
Ala Leu Leu Pro Glu Ala Ala Gly Pro Leu Leu Leu Leu Gly Gly 385 390 395

Claims

What is claimed is:
1. An isolated polypeptide selected from the group consisting of:
(i) an isolated polypeptide comprising an amino acid sequence selected from the group having at least:
(a) 70% identity;
(b) 80% identity;
(c) 90% identity; or
(d) 95% identity to the amino acid sequence of SEQ ID NO: 2 over the entire length of SEQ ID NO:2;
(ii) an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 2 or
(iii) an isolated polypeptide which is the amino acid sequence of SEQ ID NO:2.
2. An isolated polynucleotide selected from the group consisting of:
(i) an isolated polynucleotide comprising a nucleotide sequence encoding a polypeptide that has at least
(a) 70% identity;
(b) 80% identity;
(c) 90% identity; or
(d) 95% identity; to the amino acid sequence of SEQ ID NO:2, over the entire length of SEQ ID NO:2;
(ii) an isolated polynucleotide comprising a nucleotide sequence that has at least:
(a) 70% identity (b) 80% identity;
(c) 90% identity; or
(d) 95 % identity; over its entire length to a nucleotide sequence encoding the polypeptide of SEQ ID NO:2;
(iii) an isolated polynucleotide comprising a nucleotide sequence which has at least:
(a) 70% identity;
(b) 80% identity;
(c) 90% identity; or
(d) 95% identity; to that of SEQ ID NO: 1 over the entire length of SEQ ID NO:l;
(iv) an isolated polynucleotide comprising a nucleotide sequence encoding the polypeptide of SEQ ID NO:2;
(vi) an isolated polynucleotide which is the polynucleotide of SEQ ID NO: 1; or
(vi) an isolated polynucleotide obtainable by screening an appropriate hbrary under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: 1 or a fragment thereof.; or a nucleotide sequence complementary to said isolated polynucleotide.
3. An antibody immunospecific for the polypeptide of claim 1.
4. A method for the treatment of a subject:
(i) in need of enhanced activity or expression of the polypeptide of claim 1 comprising:
(a) administering to the subject a therapeutically effective amount of an agonist to said polypeptide; and/or (b) providing to the subject an isolated polynucleotide comprising a nucleotide sequence encoding said polypeptide in a form so as to effect production of said polypeptide activity in vivo. ; or
(ii) having need to inhibit activity or expression of the polypeptide of claim 1 comprising:
(a) administering to the subject a therapeutically effective amount of an antagonist to said polypeptide; and/or
(b) administering to the subject a nucleic acid molecule that inhibits the expression of a nucleotide sequence encoding said polypeptide; and/or
(c) administering to the subject a therapeutically effective amount of a polypeptide that competes with said polypeptide for its ligand, substrate , or receptor.
5. A process for diagnosing a disease or a susceptibility to a disease in a subject related to expression or activity of the polypeptide of claim 1 in a subject comprising:
(a) determining the presence or absence of a mutation in the nucleotide sequence encoding said polypeptide in the genome of said subject; and/or
(b) analyzing for the presence or amount of said polypeptide expression in a sample derived from said subject.
6. A method for screening to identify compounds which stimulate or which inhibit the function of the polypeptide of claim 1 which comprises a method selected from the group consisting of:
(a) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound; (b) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof in the presence of a labeled competitor;
(c) testing whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells or cell membranes bearing the polypeptide;
(d) mixing a candidate compound with a solution containing a polypeptide of claim 1, to form a mixture, measuring activity of the polypeptide in the mixture, and comparing the activity of the mixture to a standard; or
(e) detecting the effect of a candidate compound on the production of mRNA encoding said polypeptide and said polypeptide in cells, using for instance, an ELJSA assay.
7. An agonist or an antagonist of the polypeptide of claim 1.
8. An expression system comprising a polynucleotide capable of producing a polypeptide of claim 1 when said expression system is present in a compatible host cell.
9. A process for producing a recombinant host cell comprising transforming or transfecting a cell with the expression system of claim 8 such that the host cell, under appropriate culture conditions, produces a polypeptide comprising an amino acid sequence having at least 70% identity to the amino acid sequence of SEQ ID NO: 2 over the entire length of SEQ ID NO:2.
10. A recombinant host cell produced by the process of claim 9.
11. A membrane of a recombinant host cell of claim 10 expressing a polypeptide comprising an amino acid sequence having at least 70% identity to the amino acid sequence of SEQ ID NO:2 over the entire length of SEQ ID NO:2.
12. A process for producing a polypeptide comprising culturing a host cell of claim 10 under conditions sufficient for the production of said polypeptide and recovering the polypeptide from the culture.
13. An isolated polynucleotide selected form the group consisting of:
(a) an isolated polynucleotide comprising a nucleotide sequence which has at least 70%, 80%, 90%, 95 %, 97% identity to SEQ ID NO: 3 over the entire length of SEQ ID NO: 3;
(b) an isolated polynucleotide comprising the polynucleotide of SEQ ID NO: 3;
(c) the polynucleotide of SEQ ID NO: 3; or
(d) an isolated polynucleotide comprising a nucleotide sequence encoding a polypeptide which has at least 70 % , 80 % , 90 % , 95 % , 97-99 % identity to the amino acid sequence of SEQ ID NO:4, over the entire length of SEQ ID NO:4.
14. A polypeptide selected from the group consisting of:
(a) a polypeptide which comprises an amino acid sequence which has at least 70%, 80% , 90% , 95 % , 97-99 % identity to that of SEQ ID NO:4 over the entire length of SEQ ID NO:4;
(b) a polypeptide which has an amino acid sequence which is at least 70%, 80%, 90%, 95 %, 97-99% identity to the amino acid sequence of SEQ ID NO: 4 over the entire length of SEQ ID NO:4;
(c) a polypeptide which comprises the amino acid of SEQ ID NO: 4;
(d) a polypeptide which is the polypeptide of SEQ ID NO:4; (e) a polypeptide which is encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 3.
EP98923812A 1997-05-27 1998-05-27 SIALOADHESIN FAMILY 4 cDNA Withdrawn EP0983078A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US4757297P 1997-05-27 1997-05-27
US47572P 1997-05-27
PCT/US1998/010791 WO1998053840A1 (en) 1997-05-27 1998-05-27 SIALOADHESIN FAMILY 4 cDNA

Publications (2)

Publication Number Publication Date
EP0983078A1 true EP0983078A1 (en) 2000-03-08
EP0983078A4 EP0983078A4 (en) 2003-03-26

Family

ID=21949754

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98923812A Withdrawn EP0983078A4 (en) 1997-05-27 1998-05-27 SIALOADHESIN FAMILY 4 cDNA

Country Status (4)

Country Link
EP (1) EP0983078A4 (en)
JP (2) JP2001502359A (en)
AU (1) AU7601698A (en)
WO (1) WO1998053840A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6590088B1 (en) 1996-07-19 2003-07-08 Human Genome Sciences, Inc. CD33-like protein
CA2365040A1 (en) * 1999-04-02 2000-10-12 Eli Lilly And Company Hob-bp2h compositions, methods and uses thereof
JP2004516012A (en) * 2000-07-21 2004-06-03 ブリストル−マイヤーズ スクイブ カンパニー New siglecs and their uses
ZA200804162B (en) * 2005-11-21 2009-12-30 Genentech Inc Novel gene disruptions, compositions and methods relating thereto

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998006733A1 (en) * 1996-08-09 1998-02-19 Human Genome Sciences, Inc. Nucleotide sequence encoding the cd33-like protein

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998006733A1 (en) * 1996-08-09 1998-02-19 Human Genome Sciences, Inc. Nucleotide sequence encoding the cd33-like protein

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
ADAMS M D ET AL: "COMPEMENTARY DNA SEQUENCING: EXPRESSED SEQUENCE TAGS AND HUMAN GENOME PROJECT" SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE,, US, vol. 252, no. 5013, 21 June 1991 (1991-06-21), pages 1651-1656, XP000645049 ISSN: 0036-8075 *
DATABASE EMBL [Online] 8 January 1996 (1996-01-08) HILLIER L ET AL.: "The WashU-Merk EST Project" retrieved from EBI Database accession no. N20948 XP002228250 *
DATABASE EMBL [Online] HILLIER L. ET AL.: "The WashU-Merck EST Project" retrieved from EBI Database accession no. N94950 XP002228251 *
KELM S ET AL: "THE SIALOADHESINS - A FAMILY OF SIALIC ACID-DEPENDENT CELLULAR RECOGNITION MOLECULES WITHIN THE IMMUNOGLOBULIN SUPERFAMILY" GLYCOCONJUGATE JOURNAL, CHAPMAN & HALL, GB, vol. 13, no. 6, 1996, pages 913-926, XP002066896 ISSN: 0282-0080 *
See also references of WO9853840A1 *
TAKEI Y ET AL: "MOLECULAR CLONING OF A NOVEL GENE SIMILAR TO MYELOID ANTIGEN CD33 AND ITS SPECIFIC EXPRESSION IN PLACENTA" CYTOGENETICS AND CELL GENETICS, BASEL, CH, vol. 78, 1997, pages 295-300, XP002066897 ISSN: 0301-0171 *

Also Published As

Publication number Publication date
JP2001502359A (en) 2001-02-20
JP2004041214A (en) 2004-02-12
AU7601698A (en) 1998-12-30
EP0983078A4 (en) 2003-03-26
WO1998053840A1 (en) 1998-12-03

Similar Documents

Publication Publication Date Title
WO1999062951A1 (en) A human zinc finger protein gene (bmzf3)
WO1999062952A1 (en) A human zinc finger protein gene (bmzf2)
US7214772B2 (en) Sialoadhesin family member-2 (SAF-2)
US20010011077A1 (en) Secreted cysteine rich protein-6 (SCRP-6); polynucleotides and processes for producing polypeptides
EP0869178A1 (en) SAF-3, Sialoadhesin family member-3
EP1124851A1 (en) Hnovilr
WO1999047658A1 (en) The cytokine family member ef-7
EP0983078A1 (en) SIALOADHESIN FAMILY 4 cDNA
EP0986400A1 (en) Integrin ligand, human mindin
EP0911391A2 (en) A clone HWHHJ20
US6090582A (en) Sialoadhesin Family Member-3
EP1064364A1 (en) The cytokine family member 2-21
EP1107788A1 (en) Scrp-5: secreted cysteine rich protein-5
WO1999056778A1 (en) The cytokine family member, 2-19
WO1999011788A1 (en) Kringle1
WO2000017349A1 (en) A HUMAN Hsg III GENE
US20020055476A1 (en) RAMP2a: receptor activity modifying protein-2a
WO2000034470A1 (en) Npcbac06: human transitional endoplasmic reticulum atpase gene
WO2000058353A1 (en) Npcahh01: human transmembrane protein e3-16 gene
US20020065238A1 (en) EPO primary response gene, EPRG3pt
EP1009765A1 (en) EPO PRIMARY RESPONSE GENE, EPRG3Spt
WO2000021992A1 (en) A human fk506 binding protein (fkbp)
WO2000034328A1 (en) Tpaaoh04: human g protein sara gene
EP0897986A2 (en) Aspartic proteinase 5 (ASP5)
WO2000022116A1 (en) Human mdg1 gene (cbfawb10)

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: 19991227

AK Designated contracting states

Kind code of ref document: A1

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

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 12Q 1/68 B

Ipc: 7G 01N 33/50 B

Ipc: 7A 61K 38/17 B

Ipc: 7C 12N 5/10 B

Ipc: 7C 12N 15/63 B

Ipc: 7C 12N 15/12 B

Ipc: 7C 12N 15/01 B

Ipc: 7C 07K 16/28 B

Ipc: 7C 07K 16/18 B

Ipc: 7C 07K 16/00 B

Ipc: 7C 07K 14/705 B

Ipc: 7C 07K 14/435 B

Ipc: 7C 07H 21/04 B

Ipc: 7A 61K 48/00 B

Ipc: 7A 61K 39/395 B

Ipc: 7A 61K 38/00 A

A4 Supplementary search report drawn up and despatched

Effective date: 20030206

17Q First examination report despatched

Effective date: 20030707

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: 20040118