CA2429265A1 - Mammalian alpha-helical protein-53 - Google Patents

Mammalian alpha-helical protein-53 Download PDF

Info

Publication number
CA2429265A1
CA2429265A1 CA002429265A CA2429265A CA2429265A1 CA 2429265 A1 CA2429265 A1 CA 2429265A1 CA 002429265 A CA002429265 A CA 002429265A CA 2429265 A CA2429265 A CA 2429265A CA 2429265 A1 CA2429265 A1 CA 2429265A1
Authority
CA
Canada
Prior art keywords
glu
zalpha53
polypeptide
lys
leu
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.)
Abandoned
Application number
CA002429265A
Other languages
French (fr)
Inventor
Zeren Gao
Kimberly E. Shoemaker
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.)
Zymogenetics Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2429265A1 publication Critical patent/CA2429265A1/en
Abandoned 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Abstract

The present invention relates to polynucleotide and polypeptide molecules for mammalian alpha helix-53 (Zalpha53). The polypeptides, and polynucleotides encoding them, are hormonal and may be used to regulate the functioning of the immune system. The present invention also includes antibodies to the Zalpha53 polypeptides. Antagonists to Zalpha53 can be used to treat inflammation and inflammation-related disease.

Description

BACKGROUND OF THE INVENTION
Inflammation normally is a localized, protective response to trauma or microbial invasion that destroys, dilutes, or walls-off the injurious agent and the injured tissue. It is characterized in the acute form by the classic signs of pain, neat, redness, swelling, and loss of function. Microscopically it involves a complex series of events, including dilation of arterioles, capillaries, and venules, with increased permeability and blood flow, exudation of fluids, including plasma proteins, and leukocyte migration into l5 the area of inflammation.
Diseases characterized by inflammation are significant causes of morbidity and mortality in humans. Commonly, inflammation occurs as a defensive response to invasion of the host by foreign, particularly microbial, material.
Responses to mechanical trauma, toxins, and neoplasia also may results in inflammatory reactions.
2 o The accumulation and subsequent activation of leukocytes are central events in the pathogenesis of most forms of inflammation. Deficiencies of inflammation compromise the host. Excessive inflammation caused by abnormal recognition of host tissue as foreign or prolongation of the inflammatory process may lead to inflammatory diseases as diverse as diabetes, arteriosclerosis, cataracts, reperfusion injury, and cancer, to post-2 5 infectious syndromes such as in infectious meningitis, rheumatic fever, and to rheumatic diseases such as systemic lupus erythematosus and rheumatoid arthritis. The centrality of the inflammatory response in these varied disease processes makes its regulation a major element in the prevention control or cure of human disease.
Thus, there is a need to discover cytokines, which contribute to inflammation and 3 0 inflammatory related diseases so that antagonists such as antibodies can be administered to down-regulate the cytokine so as to ameliorate the inflammation.

DESCRIPTION OF THE INVENTION
Introduction The present invention addresses this need by providing novel polypeptides and related compositions and methods and their antagonists.
Within one aspect, the present invention provides an isolated polynucleotide encoding a mammalian cytokine termed Zalpha53. The data show that the cytokine is involved in the inflammation response. Thus, antagonists of Zalpha53 can be used to lessen inflammation especially inflammation associated with coronary heart disease, rheumatoid arthritis, arteriosclerosis, Crohn's disease and inflammatory bowel disease.
Two variants have been discovered. The first variant is SEQ ID NOs: 1 and 2. The polypeptide has a signal sequence extending from amino acid residue 1 to amino acid residue 25. The mature polypeptide is SEQ ID NO: 3. The Zalpha53 polypeptide has four alpha helices, A, B, C and D. Helix A extends from amino acid residue 28 to amino acid residue 43, and is also represented by SEQ ID NO: 4.
Helix B
extends from amino acid residue 74 to amino acid residue 88, and is also represented by SEQ ID NO: S.Helix C of SEQ ID NO: 2 extends from amino acid residue extends from amino acid residue 91 to amino acid residue 106, and is also represented by SEQ
1D NO: 6. Helix D extends from amino acid residue 147 to amino acid residue 162, and is also represented by SEQ ID NO: 7. ' 2 o The second variant is SEQ ID NOs: 10 and 20. The polypeptide of SEQ
ID NO: 20 has a signal sequence extending from amino acid residue 1 to amino acid residue 25. The mature polypeptide is SEQ ID NO: 21. The polypeptide also has four alpha helices, A, B, C and D. Helix A extends from amino acid residue 28 to amino acid residue 43 of SEQ 1D NO: 20, and is also represented by SEQ ID NO: 28.
Helix B
2 5 extends from amino acid residue 61 to amino acid residue 76 of SEQ ID NO:
20, and is also represented by SEQ ID NO: 29. Helix C extends from.amino acid residue 79 to amino acid residue 94 of SEQ DJ NO: 20, and is also represented by SEQ 117 NO:
30.
Helix D extends from amino acid residue 135 to amino acid residue 150 of SEQ
ID
NO: 20, and is also represented by SEQ ID NO: 31.
3 0 Within a second aspect of the invention there is provided an expression vector comprising (a) a transcription promoter; (b) a DNA segment encoding Zalpha53 polypeptide, and (c) a transcription terminator, wherein the promoter, DNA
segment, and terminator are operably linked.
Within a third aspect of the invention there is provided a cultured eukaryotic or prokaryotic cell into which has been introduced an expression vector as disclosed above, wherein said cell expresses a protein polypeptide encoded by the DNA
segment.
Within a further aspect of the invention there is provided a chimeric polypeptide consisting essentially of a first portion and a second portion joined by a peptide bond. The first portion of the chimeric polypeptide consists essentially of (a) a 1 o Zalpha53 polypeptide (b) allelic variants of Zalpha53; and (c) protein polypeptides that are at Least 90°Io identical to (a) or (b). The second portion of the chimeric polypeptide consists essentially of another polypeptide such as an affinity tag. Within one embodiment the affinity tag is an immunoglobulin Fc polypeptide. The invention also provides expression vectors encoding the chimeric polypeptides and host cells transfected to produce the chimeric polypeptides.
Within an additional aspect of the invention there is provided an antibody that specifically binds to a Zalpha53 polypeptide as disclosed above, and also an anti-idiotypic antibody that neutralizes the antibody to a Zalpha53 polypeptide.
An additional embodiment of the present invention relates to a peptide 2 0 or polypeptide that has the amino acid sequence of an epitope-bearing portion of a Zalpha53 polypeptide having an amino acid sequence described above. Peptides or polypeptides having the amino acid sequence of an epitope-bearing portion of a Zalpha53 polypeptide of the present invention include portions of such polypeptides with at Least nine, preferably at Least 15 and more preferably at Least 30 to 50 amino 2 5 acids, although epitope-bearing polypeptides of any length up to and including the entire amino acid sequence of a polypeptide of the present invention described above are also included in the present invention. Examples of such epitope binding regions are SEQ ID NOs: 2-18, and 20-37. Also claimed are any of these polypeptides that are fused to another polypeptide or carrier molecule.
3 0 The teachings of all the references cited herein are incorporated in their entirety herein by reference.
Prior to setting forth the invention in detail, it may be helpful to the understanding thereof to define the following terms:
The term "affinity tag" is used herein to denote a polypeptide segment that can be attached to a second polypeptide to provide for purification or detection of the second polypeptide or provide sites for attachment of the second polypeptide to a substrate. In principal, any peptide or protein for which an antibody or other specific binding agent is available can be used as an affinity tag. Affinity tags include a poly-histidine tract, protein A, Nilsson et al., EMBO J. 4:1075 (1985); Nilsson et al., Methods Eyzzy~i.ol. 198:3 (1991), glutathione S transferase, Smith and Johnson, Gefae I o X7:31 (1988), Glu-Glu affinity tag, Grussenmeyer et al., PYOG. Natl. Acad.
Sci. USA
82:7952-4 (1985), substance P, FlagTM peptide, Hopp et al., Biotechfaology 6:1204-1210 (1988), streptavidin binding peptide, or other antigenic epitope or binding domain. See, in general, Ford et al., Protein Expressiofz afi.d Purificatiofa 2: 95-107 (1991). DNAs encoding affinity tags are available from commercial suppliers (e.g., Pharmacia Biotech, Piscataway, NJ).
The term "allelic variant" is used herein to denote any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in phenotypic polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or 2 0 may encode polypeptides having altered amino acid sequence. The term allelic variant is also used herein to denote a protein encoded by an allelic variant of a gene.
The terms "amino-terminal" and "carboxyl-terminal" are used herein to denote positions within polypeptides. Where the context allows, these terms are used with reference to a particular sequence or portion of a polypeptide to denote proximity 2 5 or relative position. For example, a certain sequence positioned carboxyl-terminal to a reference sequence within a polypeptide is located proximal to the carboxyl terminus of the reference sequence, but is not necessarily at the carboxyl terminus of the complete polypeptide.
The term "complementlanti-complement pair" denotes non-identical 3 0 moieties that form a non-covalently associated, stable pair under appropriate conditions.
For instance, biotin and avidin (or streptavidin) are prototypical members of a complement/anti-complement pair. Other exemplary complement/anti-complement pairs include receptor/ligand pairs, antibody/antigen (or hapten or epitope) pairs, sense/antisense polynucleotide pairs, and the like. Where subsequent dissociation of the eomplement/anti-complement pair is desirable, the complement/anti-complement 5 pair preferably has a binding affinity of <109 M-1.
The term "complements of a polynucleotide molecule" is a polynucleotide molecule having a complementary base sequence and reverse orientation as compared to a reference sequence. For example, the sequence 5' ATGCACGGG 3' is complementary to 5' CCCGTGCAT 3'.
1 o The term "contig" denotes a polynucleotide that has a contiguous stretch of identical or complementary sequence to another polynucleotide. Contiguous sequences are said to "overlap" a given stretch of polynucleotide sequence either in their entirety or along a partial stretch of the polynucleotide. For example, representative contigs to the polynucleotide sequence 5'-ATGGCTTAGCTT-3' are 5'-TAGCTTgagtct-3' and 3'-gtcgacTACCGA-5'.
The term "degenerate nucleotide sequence" denotes a sequence of nucleotides that includes one or more degenerate codons (as compared to a reference polynucleotide molecule that encodes a polypeptide). Degenerate codons contain different triplets of nucleotides, but encode the same amino acid residue (i.e., GAU and 2 o GAC triplets each encode Asp).
The term "expression vector" is used to denote a DNA molecule, linear or circular, that comprises a segment encoding a polypeptide of interest operably linked to additional segments that provide for its transcription. Such additional segments include promoter and terminator sequences, and may also include one or more origins 2 5 of replication, one or more selectable markers, an enhancer, a polyadenylation signal, etc. Expression vectors are generally derived from plasmid or viral DNA, or may contain elements of both.
The term "isolated", when applied to a polynucleotide, denotes that the polynucleotide has been removed from its natural genetic milieu and is thus free of 3 0 other extraneous or unwanted coding sequences, and is in a form suitable for use within genetically engineered protein production systems. Such isolated molecules are those that are separated from their natural environment and include cDNA and genomic clones. Isolated DNA molecules of the present invention are free of other genes with which they are ordinarily associated, but may include naturally occurring 5' and 3' untranslated regions such as promoters and terminators. The identification of associated regions will be evident to one of ordinary skill in the art (see for example, Dynan and Tijan, Nature 316:774-7~ (1985).
An "isolated" polypeptide or protein is a polypeptide or protein that is found in a condition other than its native environment, such as apart from blood and animal tissue. In a preferred form, the isolated polypeptide is substantially free of other 1 o polypeptides, particularly other polypeptides of animal origin. It is preferred to provide the polypeptides in a highly purified form, i.e. greater than 95% pure, more preferably greater than 99% pure. When used in this context, the term "isolated" does not exclude the presence of the same polypeptide in alternative physical forms, such as dimers or alternatively glycosylated or derivatized forms.
The term "operably linked", when referring to DNA segments, indicates that the segments are arranged so that they function in concert for their intended purposes, e.g., transcription initiates in the promoter and proceeds through the coding segment to the terminator.
The term "ortholog" denotes a polypeptide or protein obtained from one 2 0 species that is the functional counterpart of a polypeptide or protein from a different species. Sequence differences among orthologs are the result of speciation.
"Paralogs" are distinct but structurally related proteins made by an organism. Paralogs are believed to arise through gene duplication. For example, a-globin, b-globin, and myoglobin are paralogs of each other.
2 5 A "polynucleotide" is a single- or double-stranded polymer of deoxyribonucleotide ox ribonucleotide bases read from the 5' to the 3' end.
Polynucleotides include RNA and DNA, and may be isolated from natural sources, synthesized in vitro, or prepared from a combination of natural and synthetic molecules.
Sizes of polynucleotides are expressed as base pairs (abbreviated "bp"), nucleotides 3 0 ("nt"), or kilobases ("kb"). Where the context allows, the latter two terms may describe polynucleotides that are single-stranded or double-stranded. When the texm is applied to double-stranded molecules it is used to denote overall length and will be understood to be equivalent to the term "base pairs". It will be recognized by those skilled in the art that the two strands of a double-stranded polynucleotide may differ slightly in length and that the ends thereof may be staggered as a result of enzymatic cleavage;
thus all nucleotides within a double-stranded polynucleotide molecule may not be paired. Such unpaired ends will in general not exceed 20 nt in length.
A "polypeptide" is a polymer of amino acid residues joined by peptide bonds, whether produced naturally or synthetically. Polypeptides of less than about 10 amino acid residues are commonly referred to as "peptides".
1 o The term "promoter" is used herein for its art-recognized meaning to denote a portion of a gene containing DNA sequences that provide for the binding of RNA polymerase and initiation of transcription. Promoter sequences are commonly, but not always, found in the 5' non-coding regions of genes.
A "protein" is a macromolecule comprising one or more polypeptide chains. A protein may also comprise non-peptidic components, such as carbohydrate groups. Carbohydrates and other non-peptidic substituents may be added to a protein by the cell in which the protein is produced, and will vary with the type of cell.
Proteins are defined herein in terms of their amino acid backbone structures;
substituents such as carbohydrate groups are generally not specified, but may be present 2 0 nonetheless.
The term "receptor" denotes a cell-associated protein that binds to a bioactive molecule (i.e., a ligand) and mediates the effect of the ligand on the cell.
Membrane-bound receptors are characterized by a multi-domain structure comprising an extracellular ligand-binding domain and an intracellular effector domain that is 2 5 typically involved in signal transduction. Binding of ligand to receptor results in a conformational change in the receptor that causes an interaction between the effector domain and other molecules) in the cell. This interaction in turn leads to an alteration in the metabolism of the cell. Metabolic events that are linked to receptor-.ligand interactions include gene transcription, phosphorylation, dephosphorylation, increases 3 0 in cyclic AMP production, mobilization of cellular calcium, mobilization of membrane lipids, cell adhesion, hydrolysis of inositol lipids and hydrolysis of phospholipids. In general, receptors can be membrane bound, cytosolic or nuclear; monomeric (e.g., thyroid stimulating hormone receptor, beta-adrenergic receptor) or multimeric (e.g., PDGF receptor, growth hormone receptor, IL-3 receptor, GM-CSF receptor, G-CSF
receptor, erythropoietin receptor and IL-6 receptor).
The term "secretory signal sequence" denotes a DNA sequence that encodes a polypeptide (a "secretory peptide") that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized. The larger polypeptide is commonly cleaved to remove the secretory peptide during transit through the secretory pathway.
The term "splice variant" is used herein to denote alternative forms of RNA transcribed from a gene. Splice variation arises naturally through use of alternative splicing sites within a transcribed RNA molecule, or less commonly between separately transcribed RNA molecules, and may result in several mRNAs transcribed from the same gene. Splice variants may encode polypeptides having altered amino acid sequence. The term splice variant is also used herein to denote a protein encoded by a splice variant of an mRNA transcribed from a gene.
Molecular weights and lengths of polymers determined by imprecise analytical methods (e.g., gel electrophoresis) will be understood to be approximate values. When such a value is expressed as "about" X or "approximately" X, the stated 2 o value of X will be understood to be accurate to t10%.
Expressi~n of Zalpha53 Expression of the Zalp7~a53 gene is seen only in the testis. This pattern of expression suggests that Zalpha53 may play a general role in development and exert 2 5 important regulatory control of testicular differentiation, of the hypothalamic-pituitary-gonadal axis, and of gonadal steroidogenesis and spermatogenesis.
Development of testicular hormone production can be divided into early and late steps, with the latter dependent on the activation of functionally determined Leydig cell precursors by luteinizing hormone (LH). However, the factors that control 3 0 the early steps in this process remain unknown, Huhtaniemi , Reprod.
Fertil. Dev. 7:

1025-1035 (1995) suggesting that Zalpha53 might be responsible for activation of a non-steroidogenic, non-LH responsive precursor cell.
Once Leydig cell differentiation has occurred, production of steroid hormones in the testis is dependent on the secretion of the gonadotrophins, LH
and follicle-stimulating hormone (FSH), by the pituitary. LH stimulates production of testosterone by the Leydig cells, whereas spermatogenesis depends on both FSH
and high intratesticular testosterone concentrations. LH and FSH secretion is in turn under control of gonadotrophin releasing hormone (GnRH) produced in the hypothalamus, I~aufman, The neuro endocrine regulation of male reproduction. in: Male Infertility.
Clinical Investigation, Cause Evaluation and Treatment., FH Comhaire, ed., pp 54(Chapman and Hall, London, 1996). Since testicular products have been shown to control LH and FSH production, this suggests that Zalpha53 might regulate hormone production by the hypothalamus.
It is well known that steroidogenesis and spermatogenesis take place within two different cellular compartments of the testes, with Leydig and Sertoli cells responsible for the former and latter, respectively, Saez, Endocrin. Rev. I5:

(1994). The activity of each of these cell types appears to be regulated by the secretory products of the other. Sertoli cell derived tumor necrosis factor-a, fibroblast growth factor, interleukin-1, transforming growth factor-B, epidermal growth 2 0 factorltransforming growth factor-a, activin, inhibin, insulin-like growth factor-1, platelet derived growth factor, endothelin, and ariginine-vasopressin have all been shown to regulate Leydig cell function, Saez , Endocrin. Rev. 15: 574-626 (1994).
Thus, Zalpha53 might control or modulate the activities of one or more of these genes.
In men, aging is associated with a progressive decline in testicular 2 5 function. These changes are manifest clinically by decreased virility, vigor, and libido that point towards a relative testicular deficiency, Vermeulen, Anna. Med.
25:531-534 (1993); Pugeat et al., Horyn. Res. 43: 104-110 (1995). Hormone replacement therapy in elderly men is not currently recommended which suggests that a new therapy for the male climacterium would be very valuable.

POLYNUCLEOTIDES
The present invention also provides polynucleotide molecules, including DNA and RNA molecules that encode the Zalpha53 polypeptides disclosed herein.
Those skilled in the art will readily recognize that, in view of the degeneracy of the 5 genetic code, considerable sequence variation is possible among these polynucleotide molecules.
Polynucleotides, generally a cDNA sequence, of the present invention encode the described polypeptides herein. A cDNA sequence that encodes a polypeptide of the present invention is comprised of a series of colons, each amino acid 10 residue of the polypeptide being encoded by a colon and each colon being comprised of three nucleotides. The amino acid residues are encoded by their respective colons as follows.
Alanine (Ale) is encoded by GCA, GCC, GCG or GCT;
Cysteine (Cys) is encoded by TGC or TGT;
Aspartic acid (Asp) is encoded by GAC or GAT;
Glutamic acid (Glu) is encoded by GAA or GAG;
Phenylalanine (Phe) is encoded by TTC or TTT;
Glycine (Gly) is encoded by GGA, GGC; GGG or GGT;
2 0 Histidine (His) is encoded by CAC or CAT;
Isoleucine (Ile) is encoded by ATA, ATC or ATT;
Lysine (Lys) is encoded by AAA, or AAG;
Leucine (Leu) is encoded by TTA, TTG, CTA, CTC, CTG or CTT;
Methionine (Met) is encoded by ATG;
2 5 Asparagine (Asn) is encoded by AAC or AAT;
Proline (Pro) is encoded by CCA, CCC, CCG or CCT;
Glutamine (Gln) is encoded by CAA or CAG;
Arginine (Arg) is encoded by AGA, AGG, CGA, CGC, CGG or CGT;
Serine (Ser) is encoded by AGC, AGT, TCA, TCC, TCG or TCT;
3 0 Threonine (Thr) is encoded by ACA, ACC, ACG or ACT;
Valine (Val) is encoded by GTA, GTC, GTG or GTT;

Tryptophan (Trp) is encoded by TGG; and Tyrosine (Tyr) is encoded by TAC or TAT.
It is to be recognized that according to the present invention, when a polynucleotide is claimed as described herein, it is understood that what is claimed are both the sense strand, the anti-sense strand, and the DNA as double-stranded having both the sense and anti-sense strand annealed together by their respective hydrogen bonds. Also claimed is the messenger RNA (mRNA) that encodes the polypeptides of the president invention, and which mRNA is encoded by the cDNA described herein.
1 o Messenger RNA (mRNA) will encode a polypeptide using the same colons as those defined herein, with the exception that each thymine nucleotide (T) is replaced by a uracil nucleotide (U).
One of ordinary skill in the art will also appreciate that different species can exhibit "preferential colon usage." In general, see, Grantham, et czl., Nuc. Acids Res. 8:1893-1912 (1980); Haas, et al. Curr. Biol. 6:315-324 (1996); Wain-Hobson, et al., Gef2e 13:355-364 (1981); Grosjean and Fiers, Gene 18:199-209 (1982);
Holm, Nuc.
Acids Res. 14:3075-3087 (1986); Ikemura, J. Mol. Biol. 158:573-597 (1982). As used herein, the term "preferential colon usage" or "preferential colons" is a term of art referring to protein translation colons that are most frequently used in cells of a certain 2 0 species, thus favoring one or a few representatives of the possible colons encoding each amino acid (See Table 2). For example, the amino acid Threonine (Thr) may be encoded by ACA, ACC, ACG, or ACT, but in mammalian cells ACC is the most commonly used colon; in other species, for example, insect cells, yeast, viruses or bacteria, different Thr colons may be preferential. Preferential colons for a particular 2 5 species can be introduced into the polynucleotides of the present invention by a variety of methods known in the art. Introduction of preferential colon sequences into recombinant DNA can, for example, enhance production of the protein by making protein translation more efficient within a particular cell type or species.
Sequences containing preferential colons can be tested and optimized for expression in various 3 0 species, and tested for functionality as disclosed herein.

Within preferred embodiments of the invention the isolated polynucleotides will hybridize to similar sized regions of SEQ m NOs:l, or 19 or a sequence complementary thereto, under stringent conditions. In general, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50°10 of the target sequence hybridizes to a perfectly matched probe. Typical stringent conditions are those in which the salt concentration is up to about 0.03 M at pH 7 and the temperature is at least about 60°C.
As previously noted, the isolated polynucleotides of the present invention include DNA and RNA. Methods for preparing DNA and RNA are well known in the art. In general, RNA is isolated from a tissue or cell that produces large amounts of Zalpha53 RNA. Such tissues and cells are identified by Northern blotting, Thomas, Proc. Natl. Acad. Sci. USA 77:5201 (1980) and are discussed below.
Total RNA can be prepared using guanidine HCl extraction followed by isolation by centrifugation in a CsCI gradient, Chirgwin et al., Biochemistry 18:52-94 (1979). Poly (A)+ RNA is prepared from total RNA using the method of Aviv and Leder, Proc.
Natl.
Acad. Sci. USA 69:1408-1412 (1972). Complementary DNA (cDNA) is prepared from poly(A)+ RNA using known methods. In the alternative, genomic DNA can be isolated. Polynucleotides encoding Zalpha53 polypeptides are then identified and 2 0 isolated by, for example, hybridization or PCR.
A full-length clone encoding Zalpha53 cari be obtained by conventional cloning procedures. Complementary DNA (cDNA) clones are preferred, although for some applications (e.g., expression in transgenic animals) it may be preferable to use a genomic clone, onto modify a cDNA clone to include at least one genomic intron.
2 5 Methods for preparing cDNA and genomic clones are well known and within the level of ordinary skill in the art, and include the use of the sequence disclosed herein, or parts thereof, for probing or priming a library. Expression libraries can be probed with antibodies to Zalpha53, receptor fragments, or other specific binding partners.
The polynucleotides of the present invention can also be synthesized 3 0 using DNA synthesizers. Currently the method of choice is the phosphoramidite method. If chemically synthesized double stranded DNA is required for an application such as the synthesis of a gene or a gene fragment, then each complementary strand is made separately. The production of short genes (60 to 80 bp) is technically straightforward and can be accomplished by synthesizing the complementary strands and then annealing them. For the production of longer genes (>300 bp), however, special strategies must be invoked, because the coupling efficiency of each cycle during chemical DNA synthesis is seldom 100/0. To overcome this problem, synthetic genes (double-stranded) are assembled in modular form from single-stranded fragments that are from 20 to 100 nucleotides in length.
See Glick and Pasternak, Molecular Biotechnology, PriyZCiples &
Applicati.orzs of Reconabiyzant DNA, (ASM Press, Washington, D.C. 1994);
Itakura et al., Anfau. Rev. Bioclzem. 53: 323-356 (1984) and Climie et al., Proc. Natl.
Acad. Sci.
USA 87:633-637 (1990).
The present invention further provides counterpart polypeptides and polynucleotides from other species (orthologs). These species include, but are not limited to mammalian, avian, amphibian, reptile, fish, insect and other vertebrate and invertebrate species. Of particular interest are Zalpha53 polypeptides from other mammalian species, including murine, porcine, ovine, bovine, canine, feline, equine, and other primate polypeptides. Orthologs of human Zalpha53 can be cloned using information and compositions provided by the present invention in combination with 2 o conventional cloning techniques. For example, a cDNA can be cloned using mRNA
obtained from a tissue or cell type that expresses Zalpha53 as disclosed herein. Suitable sources of mRNA can be identified by probing Northern blots with probes designed from the sequences disclosed herein. A library is then prepared from mRNA of a positive tissue or cell line. A Zalpha53-encoding cDNA can then be isolated by a variety of methods, such as by probing with a complete or partial human cDNA
or with one or more sets of degenerate probes based on the disclosed sequences. A cDNA
can also be cloned using the polymerase chain reaction, or PCR (Mullis, U.S.
Patent No.
4,683,202), using primers designed from the representative human Zalpha53 sequence disclosed herein. Within an additional method, the cDNA library can be used to 3 0 transform or transfect host cells, and expression of the cDNA of interest can be detected with an antibody to Zalpha53 polypeptide. Similar techniques can also be applied to the isolation of genomic clones.
Those skilled in the art will recognize that the sequence disclosed in SEQ ~ NOs: 1, or 19 represent specific alleles of human Zalpha53 and that allelic variation and alternative splicing are expected to occur. Allelic variants of this sequence can be cloned by probing cDNA or genomic libraries from different individuals according to standard procedures. Allelic variants of the DNA
sequence shown in SEQ m NO:l, including those containing silent mutations and those in which mutations result in amino acid sequence changes, are within the scope of the present invention, as are proteins which are allelic variants of SEQ ID NOs:2, 3, 20 or 21.
cDNAs generated from alternatively spliced mRNAs, which retain the properties of the Zalpha53 polypeptide are included within the scope of the present invention, as are polypeptides encoded by such cDNAs and mRNAs. Allelic variants and splice variants of these sequences can be cloned by probing cDNA or genomic libraries from different individuals or tissues according to standard procedures known in the art.
The present invention also provides isolated Zalpha53 polypeptides that are substantially identical to the polypeptides of SEQ m NOs: 2, 3, 20, or 21 and their orthologs. The term "substantially identical" is used herein to denote polypeptides having 50%, 60%, 70%, 80% and most preferably at least 90%, 95% or 99%
sequence 2 0 identity to the sequences shown in SEQ ID NOs: 2, 3, 20, or 21 or their orthologs.
Percent sequence identity is determined by conventional methods. See, for example, Altschul et al., Bull. MatJy. Bio. 48: 603-616 (1986) and Henikoff and Henikoff, Proc.
Natl. Acad. Sci. USA 89:10915-10919 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1, and the "BLOSUM 62" scoring matrix of Henikoff and Henikoff (ibid.) as shown in Table 1 (amino acids are indicated by the standard one-letter codes).
The percent identity is then calculated as:
Total number of identical matches x 100 3 0 [length of the longer sequence plus the number of gaps introduced into the longer sequence in order to align the two sequences]

E~ ,-~
N
M
i Cl~ V~ N N
O

d' M CV
,~ N
d' ---,M N

d- N M , N i r V7 ' ,--i O i N on -, N
-1 in ~ M O N , , . N
N .' ' ~N OM N~ i N '-a .--~
i M r--t M O M N M
d' N .-a N N
M ' M

,-, ,.-~ N
~ M N ~
~ N d' ~ N i i N
cn M
N

W O M
N O M M ,_, N M ,-, ~ N
,-r N

i O
a V~ N (V O M N ~ O M O N ;' ,~ ,-~ N

U ~ M .d- M M ~ ~ M ~ N r, i M r--~d' ' M ' ' ,-~ , M O i M
~O M O N ..~ ~ M d ,-i ,., M
O ~ M cn O N ~ M
~

'-' O p M N .-i N
~ ' tn O N M ~ p N O ; ,-, M N
f~ N N M N ,-, M
.
. M ~

, . ~
"''~, Ut ~ N N O .~ ,-~ O '~ ~ M (V
N ~ ,--~ ,-, N O O
~

~xzr~~aw~x~a~ ~w~, ~~' ~~~

Those skilled in the art appreciate that there are many established algorithms to align two amino acid sequences. The "FASTA" similarity search algorithm of Pearson and Lipman is a suitable protein alignment method for examining the level of identity shared by an amino acid sequence and the amino acid sequence of a putative variant. The FASTA algorithm is described by Pearson and Lipman, Proc.
Nat'l Acad. Sci. USA 85:2444 (1988), and by Pearson, Metla. Erazyrnol. 183:63 (1990).
Briefly, FASTA first characterizes sequence similarity by identifying regions shared by the query sequence (e.g., SEQ m NOs: 2, 3, 20, or 21) and a test sequence that have either the highest density of identities (if the letup variable is 1 ) or pairs of identities (if letup=2), without considering conservative amino acid substitutions, insertions or deletions. The ten regions with the highest density of identities are then re-scored' by comparing the similarity of all paired amino acids using an amino acid substitution matrix, and the ends of the regions are "trimmed" to include only those residues that contribute to the highest score. If there are several regions with scores greater than the "cutoff' value (calculated by a predetermined formula based upon the length of the sequence and the letup value), then the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps. Finally, the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman-Wunsch-Sellers algorithm (Needleman and 2 0 Wunsch, J. Mol. Biol. 48:444 (1970); Sellers, SIAM J. Appl. Math. 26:787 (1974), which allows for amino acid insertions and deletions. Illustrative parameters for FASTA analysis are: letup=l, gap opening penalty=10, gap extension penalty=1, and substitution matrix=BLOSUM62. These parameters can be introduced into a FASTA
program by modifying the scoring matrix file ("SMATRIX"), as explained in Appendix 2 of Pearson, Meth. Enzyj~zol. 183:63 (1990).
FASTA can also be used to determine the sequence identity of nucleic acid molecules using a ratio as disclosed above. For nucleotide sequence comparisons, the letup value can range between one to six, preferably from four to six.
The present invention includes nucleic acid molecules that encode a 3 0 polypeptide having one or more conservative amino acid changes, compared with the amino acid sequence of SEQ ID NOs: 2, 3, 20, or 21. The BLOSUM62 table is an amino acid substitution matrix derived from about 2,000 local multiple alignments of protein sequence segments, representing highly conserved regions of more than groups of related proteins [Henikoff and Henikoff, Proc. Nat'l Acad. Sci. USA
89:10915 (1992)]. Accordingly, the BLOSUM62 substitution frequencies can be used to define conservative amino acid substitutions that may be introduced into the amino acid sequences of the present invention. As used herein, the language "conservative amino acid substitution" refers to a substitution represented by a BLOSUM62 value of greater than -1. For example, an amino acid substitution is conservative if the substitution is characterized by a BLOSUM62 value of 0,1,2, or 3. Preferred conservative annino acid substitutions are characterized by a BLOSUM62 value of at least 1 (e.g., 1,2 or 3), while more preferred conservative substitutions are characterized by a BLOSUM62 value of at least 2 (e.g., 2 or 3).
Sequence identity of polynucleotide molecules is determined by similar methods using a ratio as disclosed above.
Variant Zalpha53 polypeptides or substantially homologous Zalpha53 polypeptides are characterized as having one or more amino acid substitutions, deletions or additions. These changes are preferably of a minor nature, that is conservative amino acid substitutions (see Table 2) and other substitutions that do not significantly affect the folding or activity of the polypeptide; small deletions, typically 2 0 of one to about 30 amino acids; and small amino- or carboxyl-terminal extensions, such as an amino-terminal methionine residue, a small linker peptide of up to about residues, or an affinity tag. The present invention thus includes polypeptides of from 20 to 30 amino acid residues that comprise a sequence that is at least 90%, preferably at least 95%, and more preferably 99% or more identical to the corresponding region of 2 5 SEQ m NOs: 2, 3, 20, or 21. Polypeptides comprising affinity tags can further comprise a proteolytic cleavage site between the Zalpha53 polypeptide and the affinity tag. Preferred such sites include thrombin cleavage sites and factor Xa cleavage sites.

Table 2 Conservative amino acid substitutions Basic: arginine lysine histidine Acidic: glutamic acid aspartic acid Polar: glutamine o asparagine Hydrophobic: leucine isoleucine valine Aromatic: phenylalanine tryptophan tyrosine Small: glycine 2 0 alanine serine threonine methionine 2 5 The present invention further provides a variety of other polypeptide fusions [and related multimeric proteins comprising one or more polypeptide fusions].
For example, a Zalpha53 polypeptide can be prepared as a fusion to a dimerizing protein as disclosed in U.S. Patents Nos. 5,155,027 and 5,567,584. Preferred dimerizing proteins in this regard include immunoglobulin constant region domains.
3 0 Immunoglobulin-Zalpha53 polypeptide fusions can be expressed in genetically engineered cells [to produce a variety of multimeric Zalpha53 analogs].
Auxiliary domains can be fused to Zalpha53 polypeptides to target them to specific cells, tissues, or macromolecules (e.g., collagen). For example, a Zalpha53 polypeptide or protein could be targeted to a predetermined cell type by fusing a Zalpha53 polypeptide to a ligand that specifically binds to a receptor on the surface of the target cell. Tn this way, polypeptides and proteins can be targeted for therapeutic or diagnostic purposes. A
Zalpha53 polypeptide can be fused to two or more moieties, such as an affinity tag for purification and a targeting domain. Polypeptide fusions can also comprise one or more cleavage sites, particularly between domains. See, Tuan et al., Cofzrzective Tissue Research 34:1-9 (1996).
The proteins of the present invention can also comprise non-naturally occurring amino acid residues. Non-naturally occurring amino acids include, without limitation, trarzs-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trazzs-4-hydroxyproline, N methylglycine; allo-threonine, methylthreonine, hydroXyethylcysteine, hydroxyethylhomocysteine, nitroglutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, ter-t-leucine, norvaline, 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine. Several methods are known in the art for incorporating non-naturally occurring amino acid residues into proteins. For example, an ijz vitro system can be employed wherein nonsense mutations are suppressed using 2 0 chemically aminoacylated suppressor tRNAs.
Methods for synthesizing amino acids and aminoacylating tRNA are known in the art. Transcription and translation of plasmids containing nonsense mutations is carried out in a cell-free system comprising an E. coli S30 extract and commercially available enzymes and other reagents. Proteins are purified by 2 5 chromatography. See, for example, Robertson et al., J. Arzz. Clzenz. Soc.
113:2722 (1991); Ellman et al., Methods Efzzymol. 202:301 (1991; Chung et al., Sciezzce 259:806-809 (1993); and Chung et al., Proc. Natl. Acad. Sci. USA 90:10145-1019 (1993).
Tn a second method, translation is carried out in Xerzopus oocytes by microinjection of mutated mRNA and chemically aminoacylated suppressor tRNAs, Turcatti et al., J.
3 0 Biol. Chem. 271:19991-19998 (1996). Within a third method, E. coli cells are cultured in the absence of a natural amino acid that is to be replaced (e.g., phenylalanine) and in the presence of the desired non-naturally occurring amino acids) (e.g., 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, or 4-fluorophenylalanine).
The non-naturally occurring amino acid is incorporated into the protein in place of its natural counterpart. See, Koide et al., Bioclaem. 33:7470-7476 (1994).
Naturally 5 occurring amino acid residues can be converted to non-naturally occurring species by in vitro chemical modification. Chemical modification can be combined with site-directed mutagenesis to further expand the range of substitutions, Wynn and Richards, Proteifa Sci. 2:395-403 (1993).
A limited number of non-conservative amino acids, amino acids that are 10 not encoded by the genetic code, non-naturally occurring amino acids, and unnatural amino acids may be substituted for Zalpha53 amino acid residues.
Essential amino acids in the polypeptides of the present invention can be identified according to procedures known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis, Cunningham and Wells, Science 244: 1081-1085 15 (1989); Bass et al., Pr-oc. Natl. Acad. Sci. USA.88:4498-502 (1991). In the latter technique, single alanine mutations are introduced at every residue in the molecule, and the resultant mutant molecules are tested for biological activity as disclosed below to identify amino acid residues that are critical to the activity of the molecule. See also, Hilton et al., J. Biol. Chem. 271:4699-708, 1996. Sites of ligand-receptor interaction 2 0 can also be determined by physical analysis of structure, as determined by such techniques as nuclear magnetic resonance, crystallography, electron diffraction or photoaffinity labeling, in conjunction with mutation of putative contact site amino acids. See, for example, de Vos et al., Science 255:306-312 (1992); Smith et al., J.
Mol. Biol. 224:899-904 (1992); Wlodaver et al., FEBS Lett. 309:59-64 (1992).
2 5 Multiple amino acid substitutions can be made and tested using known methods of mutagenesis and screening, such as those disclosed by Reidhaar-Olson and Sauer, Science 241:53-57 (1988) or Bowie and Sauer, Proc. Natl. Acad. Sci. USA
86:2152-2156 (1989). Briefly, these authors disclose methods for simultaneously randomizing two or more positions in a polypeptide, selecting for functional 3 0 polypeptide, and then sequencing the mutagenized polypeptides to determine the spectrum of allowable substitutions at each position. Other methods that can be used include phage display, e.g., Lowman et al., Biocherf2. 30:10832-10837 (1991);
Ladner et al., U.S. Patent No. 5,223,409; Huse, WIPO Publication WO 92/06204) and region-directed mutagenesis, Derbyshire et al., Gene 46:145 (1986); Ner et al., DNA
7:127 ( 1988).
Variants of the disclosed Zalpha53 DNA and polypeptide sequences can be generated through DNA shuffling as disclosed by Stemmer, Nature 370:389-391, (1994), Stemmer, Proc. Natl. Acad. Sci. USA 91:10747-10751 (1994) and W1P0 Publication WO 97/20078. Briefly, variant DNAs are generated by iyz vitro homologous recombination by random fragmentation of a parent DNA followed by 1 o reassembly using PCR, resulting in randomly introduced point mutations.
This technique can be modified by using a family of parent DNAs, such as allelic variants or DNAs from different species, to introduce additional variability into the process.
Selection or screening for the desired activity, followed by additional iterations of mutagenesis and assay provides for rapid "evolution" of sequences by selecting for desirable mutations while simultaneously selecting against detrimental changes.
Mutagenesis methods as disclosed herein can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides in host cells. Mutagenized DNA molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using modern 2 0 equipment. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide of interest, and can be applied to polypeptides of unknown structure.
Using the methods discussed herein, one of ordinary skill in the art can identify and/or prepare a variety of polypeptide fragments or variants of SEQ
ID NOs:
2 5 2, 3, 20, or 21 or that retain the properties of the wild-type Zalpha53 protein.
For any Zalpha53 polypeptide, including variants and fusion proteins, one of ordinary skill in the art can readily generate a fully degenerate polynucleotide sequence encoding that variant using the information set forth in Tables 1 and 2 above.

PROTEIN PRODUCTION
The Zalpha53 polypeptides of the present invention, including full-length polypeptides, biologically active fragments, and fusion polypeptides, can be produced in genetically engineered host cells according to conventional techniques.
Suitable host cells are those cell types that can be transformed or transfected with exogenous DNA and grown in culture, and include bacteria, fungal cells, and cultured higher eukaryotic cells. Eukaryotic cells, particularly cultured cells of multicellular organisms, are preferred. Techniques for manipulating cloned DNA molecules and introducing exogenous DNA into a variety of host cells are disclosed by Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd ed., (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), and Ausubel et al., eds., Current Protocols in Molecular Biology (John Wiley and Sons, Inc., NY, 1987). .
In general, a DNA sequence encoding a Zalpha53 polypeptide is operably linked to other genetic elements required for its expression, generally including a transcription promoter and terminator, within an expression vector. The vector will also commonly contain one or more selectable markers and one or more origins of replication, although those skilled in the art will recognize that within certain systems selectable markers may be provided on separate vectors, and replication of the exogenous DNA may be provided by integration into the host cell genome.
Selection 2 0 of promoters, terminators, selectable markers, vectors and other elements is a matter of routine design within the level of ordinary skill in the art. Many such elements are described in the literature and are available through commercial suppliers.
To direct a Zalpha53 polypeptide into the secretory pathway of a host cell, a secretory signal sequence (also known as a leader sequence, prepro sequence or 2 5 pre sequence) is provided in the expression vector. The secretory signal sequence may be that of Zalpha53, or may be derived from another secreted protein (e.g., t-PA) or synthesized de novo. The secretory signal sequence is operably linked to the Zalpha53 DNA sequence, i.e., the two sequences are joined in the correct reading frame and positioned to direct the newly synthesized polypeptide into the secretory pathway of the 3 0 host cell. Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the polypeptide of interest, although certain secretory signal sequences may be positioned elsewhere in the DNA sequence of interest (see, e.g., Welch et al., U.S.
Patent No. 5,037,743; Holland et al., U.S. Patent No. 5,143,830).
Alternatively, the secretory signal sequence contained in the polypeptides of the present invention is used to direct other polypeptides into the secretory pathway. The present invention provides for such fusion polypeptides. The secretory signal sequence contained in the fusion polypeptides of the present invention is preferably fused amino-terminally to an additional peptide to direct the additional peptide into the secretory pathway. Such constructs have numerous applications known in the art. For example, these novel secretory signal sequence fusion constructs can direct the secretion of an active component of a normally non-secreted protein, such as a receptor. Such fusions may be used irz vivo or in vitro to direct peptides through the secretory pathway.
Cultured mammalian cells are suitable hosts within the present invention. Methods for introducing exogenous DNA into mammalian host cells include calcium phosphate-mediated transfection, Wigler et al., Cell 14:725 (1978);
Corsaro and Pearson, Somatic Cell Genetics 7:603 ( 1981 ); Graham and Van der Eb, Virology 52:456 (1973), electroporation, Neumann et al., EMBO J. 1:841-845 (1982), DEAE-dextran mediated transfection (Ausubel et al., ibid., and liposome-mediated transfection, Hawley-Nelson et al., Focus 15:73 (1993); Ciccarone et al., Focus 15:80 (1993), and viral vectors, Miller and Rosman, BioTeclzniques 7:980(1989); Wang and Finer, Nature Med. 2:714 (1996). The production of recombinant polypeptides in cultured mammalian cells is disclosed, for example, by Levinson et al., U.S.
Patent No.
4,713,339; Hagen et al., U.S. Patent No. 4,784,950; Palmiter et al., U.S.
Patent No.
4,579,821; and Ringold, U.S. Patent No. 4,656,134. Suitable cultured mammalian cells include the COS-1 (ATCC No. CRL 1650), COS-7 (ATCC No. CRL 1651), BHK
(ATCC No. CRL 1632), BHK 570 (ATCC No. CRL 10314), 293 (ATCC No. CRL
1573; Graham et al., J. Gefz. Virol. 36:59 (1977) and Chinese hamster ovary (e.g. CHO-Kl; ATCC No. CCL 61) cell lines. Additional suitable cell lines are known in the art and available from public depositories such as the American Type Culture Collection, 3 0 Rockville, Maryland. In general, strong transcription promoters are preferred, such as promoters from SV-40 or cytomegalovirus. See, e.g., U.S. Patent No. 4,956,288.
Other suitable promoters include those from metallothionein genes (U.S. Patent Nos.
4,579,821 and 4,601,978) and the adenovirus major late promoter.
Drug selection is generally used to select for cultured mammalian cells into which foreign DNA has been inserted. Such cells are commonly referred to as "transfectants". Cells that have been cultured in the presence of the selective agent and are able to pass the gene of interest to their progeny are referred to as "stable transfectants." A preferred selectable marker is a gene encoding resistance to the antibiotic neomycin. Selection is carried out in the presence of a neomycin-type drug, such as G-418 or the like. Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as "amplification."
Amplification is carried out by culturing transfectants in the presence of a low level of the selective agent and then increasing the amount of selective agent to select for cells that produce high levels of the products of the introduced genes. A preferred amplifiable selectable marker is dihydrofolate reductase, which confers resistance to methotrexate.
Other drug resistance genes (e.g. hygromycin resistance, multi-drug resistance, puromycin acetyltransferase) can also be used. Alternative markers that introduce an altered phenotype, such as green fluorescent protein, or cell surface proteins such as CD4, CDB, Class I MHC, placental alkaline phosphatase may be used to sort transfected cells from untransfected cells by such means as FAGS sorting or magnetic bead separation 2 0 technology.
Other higher eukaryotic cells can also be used as hosts, including plant cells, insect cells and avian cells. The use of Agrobacteriurn rhizogenes as a vector for expressing genes in plant cells has been reviewed by Sinkar et al., J. Biosci.
(Bangalore) 11:47 (1987). Transformation of insect cells and production of foreign polypeptides therein is disclosed by Guarino et al., U.S. Patent No. 5,162,222 and WIPO publication WO 94/06463. Insect cells can be infected with recombinant baculovirus, commonly derived from Autographa californica nuclear polyhedrosis virus (AcNPV). DNA encoding the Zalpha53 polypeptide is inserted into the baculoviral genome in place of the AcNPV polyhedrin gene coding sequence by one of two 3 o methods. The first is the traditional method of homologous DNA
recombination between wild-type AcNPV and a transfer vector containing the Zalpha53 flanked by AcNPV sequences. Suitable insect cells, e.g. SF9 cells, are infected with wild-type AcNPV and transfected with a transfer vector comprising a Zalpha53 polynucleotide operably linked to an AcNPV polyhedrin gene promoter, terminator, and flanking sequences. See, King, L.A. and Possee, R.D., The Baculovirus Expression Systerrz: A
5 Laboratory Guide, (Chapman & Hall, London); O'Reilly, D.R. et al., Baculovirus Expression Vectors: A Laboratory Mafzual (Oxford University Press, New York, New York, 1994); and, Richardson, C. D., Ed., Baculovirus Expression Protocols.
Methods irz Molecular Biology, (Humana Press, Totowa, NJ 1995). Natural recombination within an insect cell will result in a recombinant baculovirus that contains Zalpha53 10 driven by the polyhedrin promoter. Recombinant viral stocks are made by methods commonly used in the art.
The second method of making recombinant baculovirus utilizes a transposon-based system described by Luckow, V.A, et al., J Virol 67:4566 (1993).
This system is sold in the Bac-to-Bac kit (Life Technologies, Rockville, MD).
This 15 system utilizes a transfer vector, pFastBaclT"' (Life Technologies) containing a Tn7 transposon to move the DNA encoding the Zalpha53 polypeptide into a baculovirus genome maintained in E. coli as a large plasmid called a "bacmid." The pFastBaclT""
transfer vector utilizes the AcNPV polyhedrin promoter to drive the expression of the gene of interest, in this case Zalpha53. However, pFastBaclT"" can be modified to a 2 o considerable degree. The polyhedrin promoter can be removed and substituted with the baculovirus basic protein promoter (also known as Pcor, p6.9 or MP promoter), which is expressed earlier in the baculovirus infection, and has been shown to be advantageous for expressing secreted proteins. See, Hill-Perkins, M.S. and Possee, R.D., J Gen Virol 71:971 (1990); Bonning, B.C. et al., J Gefz Virol 75:1551 (1994);
25 and, Chazenbalk, G.D., and Rapoport, B., JBiol Chem 270:1543 (1995). In such transfer vector constructs, a short or long version of the basic protein promoter can be used. Moreover, transfer vectors can be constructed that replace the native Zalpha53 secretory signal sequences with secretory signal sequences derived from insect proteins.
For example, a secretory signal sequence from Ecdysteroid Glucosyltransferase (EGT), 3 0 honey bee Melittin (Invitrogen, Carlsbad, CA), or baculovirus gp67 (PharMingen, San Diego, CA) can be used in constructs to replace the native Zalpha53 secretory signal sequence. In addition, transfer vectors can include an in-frame fusion with DNA
encoding an epitope tag at the C- or N-terminus of the expressed Zalpha53 polypeptide, for example, a Glu-Glu epitope tag, Grussenmeyer, T. et al., Proc Natl Acad Sci.
82:7952 (1985). Using a technique known in the art, a transfer vector containing Zalpha53 is transformed into E. coli, and screened for bacmids which contain an interrupted lacZ gene indicative of recombinant baculovirus. The bacmid DNA
containing the recombinant baculovirus genome is isolated, using common techniques, and used to transfect Spodoptera frzzgiperda cells, e.g. Sf9 cells.
Recombinant virus that expresses Zalpha53 is subsequently produced. Recombinant viral stocks are made by methods commonly used the art.
The recombinant virus is used to infect host cells, typically a cell line derived from the fall army worm, Spodoptera frugiperda. See, in general, Glick and Pasternak, Molecular Biotechnology: PriYZCiples azzd Applicatiozzs of Recoznbizzant DNA (ASM Press, Washington, D.C., 1994). Another suitable cell line is,the High FiveOrM cell line (Invitrogen) derived from Trichoplusia ni (U.S. Patent #5,300,435).
Commercially available serum-free media are used to grow and maintain the cells.
Suitable media are Sf900 IIT"" (Life Technologies) or ESF 921rM (Expression Systems) for the Sf9 cells; and Ex-ce110405T"" (JRH Biosciences, Lenexa, KS) or Express FiveOT~" (Life Technologies) for the T. ni cells. The cells are grown up from an 2 o inoculation density of approximately 2-5 x 105 cells to a density of 1-2 x 106 cells at which time a recombinant viral stock is added at a multiplicity of infection (MOB of 0.1 to 10, more typically near 3. The recombinant virus-infected cells typically produce the recombinant Zalpha53 polypeptide at 12-72 hours post-infection and secrete it with varying efficiency into the medium. The culture is usually harvested 48 hours post-2 5 infection. Centrifugation is used to separate the cells from the medium (supernatant).
The supernatant containing the Zalpha53 polypeptide is filtered through micropore filters, usually 0.45 ~,m pore size. Procedures used are generally described in available laboratory manuals (King, L. A. and Possee, R.D., ibid.; O'Reilly, D.R. et al., ibid.;
Richardson, C. D., ibid.). Subsequent purification of the Zalpha53 polypeptide from 3 0 the supernatant can be achieved using methods described herein.

Fungal cells, including yeast cells, can also be used within the present invention. Yeast species of particular interest in this regard include Saccharorrzyces cerevisiae, Pichia pastoris, and Piclzia rrzethanolica. Methods for transforming S.
cerevisiae cells with exogenous DNA and producing recombinant polypeptides therefrom are disclosed by, for example, Kawasaki, U.S. Patent No. 4,599,31 l;
Kawasaki et al., U.S. Patent No. 4,931,373; Brake, U.S. Patent No. 4,870,008;
Welch et al., U.S. Patent No. 5,037,743; and Murray et al., U.S. Patent No. 4,845,075.
Transformed cells are selected by phenotype determined by the selectable marker, commonly drug resistance or the ability to grow in the absence of a particular nutrient (e.g., leucine). A preferred vector system for use in Saccharomyces cerevisiae is the POTI vector system disclosed by Kawasaki et al. (U.S. Patent No. 4,931,373), which allows transformed cells to be selected by growth in glucose-containing media.
Suitable promoters and terminators for use in yeast include those from glycolytic enzyme genes (see, e.g., Kawasaki, U.S. Patent No. 4,599,311; Kingsman et al., U.S.
Patent No. 4,615,974; and Bitter, U.S. Patent No. 4,977,092) and alcohol dehydrogenase genes. See also U.S. Patents Nos. 4,990,446; 5,063,154;
5,139,936 and 4,661,454. Transformation systems for other yeasts, including Flarzserzula polymorplaa, Schizosaccharonzyces pombe, Kluyveronzyces lactic, Kluyveromyces fragilis, Ustilago maydis, Pichia pastoris, Piclzia metl2arzolica, Pich.ia guillermondii and Carcdida 2 0 maltosa are known in the art. See, for example, Gleeson et al., J. Gerz.
Microbiol.
132:3459 (1986) and Cregg, U.S. Patent No. 4,882,279. Aspergillus cells may be utilized according to the methods of McKnight et al., U.S. Patent No.
4,935,349.
Methods for transforming Acrefnoniunz chfysogerzum are disclosed by Sumino et al., U.S. Patent No. 5,162,228. Methods for transforming Neurospof°a are disclosed by 2 5 Lambowitz, U.S. Patent No. 4,486,533.
The use of Pichia methanolica as host for the production of recombinant proteins is disclosed in WIPO Publications WO 97/17450, WO 97/17451, WO
98/02536, and WO 98/02565. DNA molecules for use in transforming P.
nzetha~zolica will commonly be prepared as double-stranded, circular plasmids, which are preferably 3 0 linearized prior to transformation. For polypeptide production in P.
methanolica, it is preferred that the promoter and terminator in the plasmid be that of a P.
rraethafzolica gene, such as a P. metlzatzolica alcohol utilization gene (AUGI orAUG2). Other useful promoters include those of the dihydroxyacetone synthase (DHAS), formate dehydrogenase (FMD), and catalase (CAT) genes. To facilitate integration of the DNA
into the host chromosome, it is preferred to have the entire expression segment of the plasmid flanked at both ends by host DNA sequences. A preferred selectable marker for use in Pichia methaf2olica is a P. metlzayaolica ADE2 gene, which encodes phosphoribosyl-5-aminoimidazole carboxylase (AIRC; EC 4.1.1.21), which allows ade2 host cells to grow in the absence of adenine. For large-scale, industrial processes where it is desirable to minimize the use of methanol, it is preferred to use host cells in 1 o which both methanol utilization genes (AUGl and AUG2) are deleted. For production of secreted proteins, host cells deficient in vacuolar protease genes (PEP4 and PRBI ) are preferred. Electroporation is used to facilitate the introduction of a plasmid containing DNA encoding a polypeptide of interest into P. metharaolica cells.
It is preferred to transform P. fraetlaafaolica cells by electroporation using an exponentially l5 decaying, pulsed electric field having a field strength of from 2.5 to 4.5 kV/cm, preferably about 3.75 kV/cm, and a time constant (t) of from 1. to 40 milliseconds, most preferably about 20 milliseconds.
Prokaryotic host cells, including strains of the bacteria Eschericlaia coli, Bacillus and other genera are also useful host cells within the present invention.
2 0 Techniques for transforming these hosts and expressing foreign DNA
sequences cloned therein are well known in the art, see, e.g., Sambrook et al., ibid.). When expressing a Zalpha53 polypeptide in bacteria such as E. coli, the polypeptide may be retained in the cytoplasm, typically as insoluble granules, or may be directed to the periplasmic space by a bacterial secretion sequence. In the former case, the cells are lysed, and the 2 5 granules are recovered and denatured using, for example, guanidine isothiocyanate or urea. The denatured polypeptide can then be refolded and dimerized by diluting the denaturant, such as by dialysis against a solution of urea and a combination of reduced and oxidized glutathione, followed by dialysis against a buffered saline solution. In the latter case, the polypeptide can be recovered from the periplasmic space in a soluble 3 o and functional form by disrupting the cells (by, for example, sonication or osmotic shock) to release the contents of the periplasmic space and recovering the protein, thereby obviating the need for denaturation and refolding.
Transformed or transfected host cells are cultured according to conventional procedures in a culture medium containing nutrients and other components required for the growth of the chosen host cells. A variety of suitable media, including defined media and complex media, are known in the art and generally include a carbon source, a nitrogen source, essential amino acids, vitamins and minerals. Media may also contain such components as growth factors or serum, as required. The growth medium will generally select for cells containing the exogenously added DNA by, for example, drug selection or deficiency in an essential nutrient, which is complemented by the selectable marker carried on the expression vector or co-transfected into the host cell. P. rrzethanolica cells are cultured in a medium comprising adequate sources of carbon, nitrogen and trace nutrients at a temperature of about 25°G
to 35°C. Liquid cultures are provided with sufficient aeration by conventional means, such as shaking of small flasks or sparging of fermentors. A preferred culture medium for P. rnethafzolica is YEPD (2% D-glucose, 2% BactoTM Peptone (Difco Laboratories, Detroit, Mn, 1% BactoTM yeast extract (Difco Laboratories), 0.004% adenine and 0.006% L-leucine).
Another embodiment of the present invention provides for a peptide or 2 0 polypeptide comprising an epitope-bearing portion of a Zalpha53 polypeptide of the invention. The epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide of the invention. A region of a protein to which an antibody can bind is defined as an "antigenic epitope". See for instance, Geysen, H.M.
et al., Proc. Natl. Acad Sci. USA 81: 3998-4002 (1984).
2 5 As to the selection of peptides or polypeptides bearing an antigenic epitope (i.e., that contain a region of a protein molecule to which an antibody can bind), it is well known in the art that relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See Sutcliffe, J.G. et al. Sciezzce 219: 660-666 (1983).
3 0 Peptides capable of eliciting protein-reactive sera are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins (i.e., immunogenic epitopes) nor to the amino or carboxyl terminals. Peptides that are extremely hydrophobic and those of six or fewer residues generally are ineffective at inducing antibodies that bind to the mimicked protein; longer soluble peptides, 5 especially those containing proline residues, usually are effective.
Antigenic epitope-bearing peptides and polypeptides of the invention are therefore useful to raise antibodies, including monoclonal antibodies, which bind specifically to a polypeptide of the invention. Antigenic epitope-bearing peptides and polypeptides of the present invention contain a sequence of at least nine, preferably 10 between 15 to about 30 amino acids contained within the amino acid sequence of a polypeptide of the invention. However, peptides or polypeptides comprising a larger portion of an amino acid sequence of the invention, containing from 30 to 50 amino acids, or any length up to and including the entire amino acid sequence of a polypeptide of the invention, also are useful for inducing antibodies that react with the protein.
15 Preferably, the amino acid sequence of the epitope-bearing peptide is selected to provide substantial solubility in aqueous solvents (i.e., the sequence includes relatively hydrophilic residues and hydrophobic residues are preferably avoided); and sequences containing proline residues are particularly preferred. All of the polypeptides shown in the sequence listing contain antigenic epitopes to be used according to the present 2 0 invention. The present invention also provides polypeptide fragments or peptides comprising an epitope-bearing portion of a Zalpha53 polypeptide described herein.
Such fragments or peptides may comprise an "immunogenic epitope," which is a part of a protein that elicits an antibody response when the entire protein is used as an immunogen. Immunogenic epitope-bearing peptides can be identified using standard 2 5 methods [see, for example, Geysen et al., supra. See also U.S. Patent No.
4,708,781 (1987) further describes how to identify a peptide bearing an immunogenic epitope of a desired protein.

3l Protein Isolation It is preferred to purify the polypeptides of the present invention to >80% purity, more preferably to >_90% purity, even more preferably >_95°lo purity, and particularly preferred is a pharmaceutically pure state, that is greater than 99.9% pure with respect to contaminating macromolecules, particularly other proteins and nucleic acids, and free of infectious and pyrogenic agents. Preferably, a purified polypeptide is substantially free of other polypeptides, particularly other polypeptides of animal origin.
Expressed recombinant Zalpha53 polypeptides (or chimeric Zalpha53 polypeptides) can be purified using fractionation and/or conventional purification methods and media. Ammonium sulfate precipitation and acid or chaotrope extraction may be used for fractionation of samples. Exemplary purification steps may include hydroxyapatite, size exclusion, FPLC and reverse-phase high performance liquid chromatography. Suitable chromatographic media include derivatized dextrans, agarose, cellulose, polyacrylamide, specialty silicas, and the like. PEI, DEAE, QAE
and Q derivatives are preferred. Exemplary chromatographic media include those media derivatized with phenyl, butyl, or octyl groups, such as Phenyl-Sepharose FF
(Pharmacia), Toyopearl butyl 650 (Toso Haas, Montgomeryville, PA), Octyl-Sepharose (Pharmacia) and the like; or polyacrylic resins, such as Amberchrom CG 71 (Toso Haas) and the like. Suitable solid supports include glass beads, silica-based resins, 2 0 cellulosic resins, agarose beads, cross-linked agarose beads, polystyrene beads, cross-linked polyacrylamide resins and the like that are insoluble under the conditions in which they are to be used. These supports may be modified with reactive groups that allow attachment of proteins by amino groups, carboxyl groups, sulfhydryl groups, hydroxyl groups and/or carbohydrate moieties. Examples of coupling chemistries 2 5 include cyanogen bromide activation, N-hydroxysuccinimide activation, epoxide activation, sulfhydryl activation, hydrazide activation, and carboxyl and amino derivatives for carbodiimide coupling chemistries. These and other solid media are well known and widely used in the art, and are available from commercial suppliers.
Methods for binding receptor polypeptides to support media are well known in the art.
3 0 Selection of a particular method is a matter of routine design and is determined in part by the properties of the chosen support. See, for example, Affinity Chrornatogr-aphy:
Principles & Methods (Pharmacia LIMB Biotechnology, Uppsala, Sweden, 1988).
The polypeptides of the present invention can be isolated by exploitation of their properties. For example, immobilized metal ion adsorption (IMAC) chromatography can be used to purify histidine-rich proteins, including those comprising polyhistidine tags. Briefly, a gel is first charged with divalent metal ions to form a chelate, Sulkowski, Trends irz Biocheni. 3:1 (1985). Histidine-rich proteins will be adsorbed to this matrix with differing affinities, depending upon the metal ion used, and will be eluted by competitive elution, lowering the pH, or use of strong chelating agents. Other methods of purification include purification of glycosylated proteins by lectin affinity chromatography and ion exchange chromatography. Methods in Enzyrnol., Vol. 182, "Guide to Protein Purification", M. Deutscher, (ed.),page (Acad. Press, San Diego, 1990). Within additional embodiments of the invention, a fusion of the polypeptide of interest and an affinity tag (e.g., maltose-binding protein, an immunoglobulin domain) may be constructed to facilitate purification.
Moreover, using methods described in the art, polypeptide fusions, or hybrid Zalpha53 proteins, are constructed using regions or domains of the inventive Zalpha53, Sambrook et al., ibid., Altschul et al., ibid., Picard, Cur. Opin.
Biology, 5:511 (1994). These methods allow the determination of the biological importance of 2 0 larger domains or regions in a polypeptide of interest. Such hybrids may alter reaction kinetics, binding, constrict or expand the substrate specificity, or alter tissue and cellular localization of a polypeptide, and can be applied to polypeptides of unknown structure.
Fusion proteins can be prepared by methods known to those skilled in 2 5 the art by preparing each component of the fusion protein and chemically conjugating them. Alternatively, a polynucleotide encoding both components of the fusion protein in the proper reading frame can be generated using known techniques and expressed by the methods described herein. For example, part or all of a domains) conferring a biological function may be swapped between Zalpha53 of the present invention with 3 0 the functionally equivalent domains) from another family member. Such domains include, but are not limited to, the secretory signal sequence, conserved, and significant domains or regions in this family. Such fusion proteins would be expected to have a biological functional profile that is the same or similar to polypeptides of the present invention or other known family proteins, depending on the fusion constructed.
Moreover, such fusion proteins may exhibit other properties as disclosed herein.
Zalpha53 polypeptides or fragments thereof may also be prepared through chemical synthesis. Zalpha53 polypeptides may be monomers or multimers;
glycosylated or non-glycosylated; pegylated or non-pegylated; and may or may not include an initial methionine amino acid residue.
1 o Chemical Synthesis of Polypeptides Polypeptides, especially polypeptides of the present invention can also be synthesized by exclusive solid phase synthesis, partial solid phase methods, fragment .
condensation or classical solution synthesis. The polypeptides are preferably prepared by solid phase peptide synthesis, for example as described by Merrifield, J.
Arn. Chena.
Soc. 85:2149 (1963).
ASSAYS
The activity of.molecules of the present invention can be measured using a variety of assays. Of particular interest are changes in steroidogenesis, 2 o spermatogenesis, in the testis, LH and FSH production and GnRH in the hypothalamus.
Such assays are well known in the art.
Proteins of the present invention are useful for increasing sperm production. Zalpha53 can be measured in vitro using cultured cells or in vivo by administering molecules of the claimed invention to the appropriate animal model. For 2 5 instance, Zalpha53 transfected (or co-transfected) expression host cells may be embedded in an alginate environment and injected (implanted) into recipient animals.
Alginate-poly-L-lysine microencapsulation, permselective membrane encapsulation and diffusion chambers have been described as a means to entrap transfected mammalian cells or primary mammalian cells. These types of non-immunogenic "encapsulations"
3 0 or microenvironments permit the transfer of nutrients into the microenvironment, and also permit the diffusion of proteins and other macromolecules secreted or released by the captured cells across the environmental barrier to the recipient animal.
Most importantly, the capsules or microenvironments mask and shield the foreign, embedded cells from the recipient animal's immune response. Such microenvironments can extend the life of the injected cells from a few hours or days (naked cells) to several weeks (embedded cells).
Alginate threads provide a simple and quick means for generating embedded cells. The materials needed to generate the alginate threads are readily available and relatively inexpensive. Once made, the alginate threads are relatively strong and durable, both iya vitro and, based on data obtained using the threads, in vivo.
The alginate threads~are easily manipulable and the methodology is scalable for preparation of numerous threads. In an exemplary procedure, 3% alginate is prepared in sterile H20, and sterile filtered. Just prior to preparation of alginate threads, the alginate solution is again filtered. An approximately 50% cell suspension (containing about 5 x 105 to about 5 x 107 cells/ml) is mixed with the 3% alginate solution. One ml of the alginate/cell suspension is extruded into a 100 nnM sterile filtered CaCl2 solution over a time period of ~15 min, forming a "thread". The extruded thread is then transferred into a solution of 50 mM CaCI~, and then into a solution of 25 mM
CaCI~.
The thread is then rinsed with deionized water before coating the thread by incubating in a 0.01% solution of poly-L-lysine. Finally, the thread is rinsed with Lactated 2 o Ringer's Solution and drawn from solution into a syringe barrel (without needle attached). A large bore needle is then attached to the syringe, and the thread is intraperitoneally injected into a recipient in a minimal volume of the Lactated Ringer's Solution.
An alternative ijz vivo approach for assaying proteins of the present 2 5 invention involves viral delivery systems. Exemplary viruses for this purpose include adenovirus, herpesvirus, vaccinia virus and adeno-associated virus (AAV).
Adenovirus, a double-stranded DNA virus, is currently the best studied gene transfer vector for delivery of heterologous nucleic acid [for a review, see T.C.
Becker et al., Metla. Cell Biol. 43:161 (1994); and J.T. Douglas and D.T. Curiel, Science &
Medicine 3 0 4:44 (1997)]. The adenovirus system offers several advantages: adenovirus can (i) accommodate relatively large DNA inserts; (ii) be grown to high-titer; (iii) infect a broad range of mammalian cell types; and (iv) be used with a large number of available vectors containing different promoters. Also, because adenoviruses are stable in the bloodstream, they can be administered by intravenous injection.
By deleting portions of the adenovirus genome, larger inserts (up to 7 5 kb) of heterologous DNA can be accommodated. These inserts can be incorporated into the viral DNA by direct ligation or by homologous recombination with a co transfected plasmid. In an exemplary system, the essential E 1 gene has been deleted from the viral vector, and the virus will not replicate unless the E1 gene is provided by the host cell (the human 293 cell line is exemplary). When intravenously administered 1 o to intact animals, adenovirus primarily targets the liver. If the adenoviral delivery system has an E1 gene deletion, the virus cannot replicate in the host cells.
However, the host's tissue (e.g., liver) will express and process (and, if a secretory signal sequence is present, secrete) the heterologous protein. Secreted proteins will enter the circulation in the highly vascularized liver, and effects on the infected animal can be 15 determined.
The adenovirus system can also be used for protein production i~ vitro.
By culturing adenovirus-infected non-293 cells under conditions where the cells are not rapidly dividing, the cells can produce proteins for extended periods of time.
For instance, BHI~ cells are grown to confluence in cell factories then exposed to the 2 0 adenoviral vector encoding the secreted protein of interest. The cells are then grown under serum-free conditions, which allows infected cells to survive for several weeks without significant cell division. Alternatively, adenovirus vector infected 2935 cells can be grown in suspension culture at relatively high cell density to produce significant amounts. of protein (see Gamier et al., Cytotechnol. 15:145 (1994). With either 2 5 protocol, an expressed, secreted heterologous protein can be repeatedly isolated from the cell culture supernatant. Within the infected 2935 cell production protocol, non-secreted, proteins may also be effectively obtained.
Agonists and Antagonists 3 o In view of the tissue distribution observed for Zalpha53, agonists (including the natural ligand/ substrate/ cofactor/ etc.) and antagonists have enormous potential in both in vitro and r.'fa vivo applications. Compounds identified as Zalpha53 agonists are useful for stimulating the immune system or spermatogenesis. For example, Zalpha53 and agonist compounds are useful as components of defined cell culture media, and may be used alone or in combination with other cytokines and hormones to replace serum that is commonly used in cell culture.
Antagonists Antagonists are also useful as research reagents for characterizing sites of ligand-receptor interaction. Antagonists of Zalpha53 can also be used to down-regulate inflammation as discussed in more further detail below. Inhibitors of Zalpha53 activity (Zalpha53 antagonists) include anti-Zalpha53 antibodies and soluble Zalpha53 receptors, as well as other peptidic and non-peptidic agents (including ribozymes).
Zalpha53 can also be used to identify inhibitors (antagonists) of its activity. Test compounds are added to the assays disclosed herein to identify compounds that inhibit the activity of Zalpha53. In addition to those assays disclosed herein, samples can be tested for inhibition of Zalpha53 activity within a variety of assays designed to measure receptor binding or the stimulation/inhibition of Zalpha53-dependent cellular responses.
For example, Zalpha53-responsive cell lines can be transfected with a reporter gene construct that is responsive to a Zalpha53-stimulated cellular pathway.
Reporter gene 2 0 constructs of this type are known in the art, and will generally comprise a Zalpha53-DNA response element operably linked to a gene encoding a protein that can be assayed, such as luciferase. DNA response elements can include, but are not limited to, cyclic AMP response elements (CRE), hormone response elements (HRE) insulin response element (IRE), Nasrin et al., Proc. Natl. Acad. Sci. USA $7:5273 (1990) and 2 5 serum response elements (SRE) (Shaw et al. Cell 56: 563 (1989). Cyclic AMP
response elements are reviewed in Roestler et al., J. Biol. Chenz. 263 ( 19):9063 (1988) and Habener, Molec. Endocrif~ol. 4 (8):1087 (1990). Hormone response elements are reviewed in Beato, Cell 56:335 (1989). Candidate compounds, solutions, mixtures or extracts are tested for the ability to inhibit the activity of Zalpha53 on the target cells as 3 0 evidenced by a decrease in Zalpha53 stimulation of reporter gene expression. Assays of this type will detect compounds that directly block Zalpha53 binding to cell-surface receptors, as well as compounds that block processes in the cellular pathway subsequent to receptor-ligand binding. In the alternative, compounds or other samples can be tested for direct blocking of Zalpha53 binding to receptor using Zalpha53 tagged with a detectable Iabel (e.g., lash biotin, horseradish peroxidase, FTTC, and the like). Within assays of this type, the ability of a test sample to inhibit the binding of labeled Zalpha53 to the receptor is indicative of inhibitory activity, which can be confirmed through secondary assays. Receptors used within binding assays may be cellular receptors or isolated, immobilized receptors.
A Zalpha53 polypeptide can be expressed as a fusion with an immunoglobulin heavy chain constant region, typically an Fc fragment, which contains two constant region domains and lacks the variable region. Methods for preparing such fusions are disclosed in U.S. Patents Nos. 5,155,027 and 5,567,584.. Such fusions are typically secreted as multimeric molecules wherein the Fc portions are disulfide bonded to each other and two non-Ig polypeptides are arrayed in closed proximity to each other.
Fusions of this type can be used to affinity purify the ligand. For use in assays, the chimeras are bound to a support via the Fc region and used in an ELISA format.
, A Zalpha53 ligand-binding polypeptide can also be used for purification of ligand. The polypeptide is immobilized on a solid support, such as beads of agarose, cross-linked agarose, glass, cellulosic resins, silica-based resins, polystyrene, cross-2 0 linked polyacrylamide, or like materials that are stable under the conditions of use.
Methods for linking polypeptides to solid supports are known in the art, and include amine chemistry, cyanogen bromide activation, N-hydroxysuccinimide activation, epoxide activation, sulfhydryl activation, and hydrazide activation. The resulting medium will generally be configured in the form of a column, and fluids containing 2 5 ligand are passed through the column one or more times to allow ligand to bind to the receptor polypeptide. The ligand is then eluted using changes in salt concentration, chaotropic agents (guanidine HCl), or pH to disrupt ligand-receptor binding.
An assay system that uses a ligand-binding receptor (or an antibody, one member of a complement/ anti-complement pair) or a binding fragment thereof, and a 3 0 commercially available biosensor instrument (BIAcore, Pharmacia Biosensor, Piscataway, NJ) may be advantageously employed. Such receptor, antibody, member of a complement/anti-complement pair or fragment is immobilized onto the surface of a receptor chip. Use of this instrument is disclosed by Karlsson, J. Izzzmurzol.
Methods 145:229 (1991) and Cunningham and Wells, J. Mol. Biol. 234:554. (I993). A
receptor, antibody, member or fragment is covalently attached, using amine or sulfhydryl chemistry, to dextran fibers that are attached to gold film within the flow cell. A test sample is passed through the cell. If a ligand, epitope, or opposite member of the complementlanti-complement pair is present in the sample, it will bind to the immobilized receptor, antibody or member, respectively, causing a change in the refractive index of the medium, which is detected as a change in surface plasmon resonance of the gold film. This system allows the determination of on- and off rates, from which binding affinity can be calculated, and assessment of stoichiometry of binding.
Ligand-binding receptor polypeptides can also be used within other assay systems known in the art. Such systems include Scatchard analysis for determination of binding affinity, Scatchard, Anzz. NYAcad. Sci. S1: 660 (1949) and calorimetric assays, Cunningham et al., Science 253:545 (1991); Cunningham et al., Sciezzce 245:821 (1991).
Zalpha53 polypeptides can also be used to prepare antibodies that specifically bind to Zalpha53 epitopes, peptides or polypeptides. The Zalpha53 2 0 polypeptide or a fragment thereof serves as an antigen (immunogen) to inoculate an animal and elicit an immune response. Suitable antigens would be the Zalpha53 polypeptides encoded by SEQ 1D NOs: 2-18 and 20-37. Antibodies generated from this immune response can be isolated and purified as described herein. Methods for preparing and isolating polyclonal and monoclonal antibodies are well known in the art.
2 5 See, for example, Current Protocols in Izriznuzzology, Cooligan, et al.
(eds.), National Institutes of Health, (John Wiley and Sons, Inc., 1995); Sambrook et al., Molecular Clozzifzg: A Laboratory Mayzual, Second Edition (Cold Spring Harbor, NY, 1989); and Hurrell, J. G. R., Ed., Mofaoclofzal Hybridoma Antibodies: Techfziques and Applications (CRC Press, Inc., Boca Raton, FL, 1982).
3 0 As would be evident to one of ordinary skill in the art, polyclonal antibodies can be generated from inoculating a variety of warm-blooded animals such as horses, cows, goats, sheep, dogs, chickens, rabbits, mice, and rats with a Zalpha53 polypeptide or a fragment thereof. The immunogenicity of a Zalpha53 polypeptide may be increased through the use of an adjuvant, such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant. Polypeptides useful for immunization also include fusion polypeptides, such as fusions of Zalpha53 or a portion thereof with an immunoglobulin polypeptide or with maltose binding protein. The polypeptide immunogen may be a full-length molecule or a portion thereof. If the polypeptide portion is "hapten-like", such portion may be advantageously joined or linked to a macromolecular carrier (such as keyhole limpet hemocyanin (I~LH), bovine serum albumin (BSA) or tetanus toxoid) for immunization.
As used herein, the term "antibodies" includes polyclonal antibodies, affinity-purified polyclonal antibodies, monoclonal antibodies, and antigen-binding fragments, such as F(ab')2, Fab proteolytic fragments, Genetically engineered intact antibodies or fragments, such as chimeric antibodies, Fv fragments, single chain antibodies and the like, as well as synthetic antigen-binding peptides and polypeptides.
Non-human antibodies may be humanized by grafting non-human CDRs onto human framework and constant regions, or by incorporating the entire non-human variable domains (optionally "cloaking" them with a human-like surface by replacement of exposed residues, wherein the result is a "veneered" antibody). In some instances, 2 0 humanized antibodies may retain non-human residues within the human variable region framework domains to enhance proper binding characteristics. Through humanizing antibodies, biological half life may be increased, and the potential for adverse immune reactions upon administration to humans is reduced.
Alternative techniques for generating or selecting antibodies useful 2 5 herein include ira vitro exposure of lymphocytes to Zalpha53 protein or peptide, and selection of antibody display libraries in phage or similar vectors (for instance, through use of immobilized or labeled Zalpha53 protein or peptide). Genes encoding polypeptides having potential Zalpha53 polypeptide-binding domains can be obtained by screening random peptide libraries displayed on phage (phage display) or on 3 0 bacteria, such as E. coli. Nucleotide sequences encoding the polypeptides can be obtained in a number of ways, such as through random mutagenesis and random polynucleotide synthesis. These random peptide display libraries can be used to screen for peptides that interact with a known target, which can be a protein or polypeptide, such as a ligand or receptor, a biological or synthetic macromolecule, or organic or inorganic substances. Techniques for creating and screening such random peptide 5 display libraries are known in the art (Ladner et al., US Patent NO.
5,223,409; Ladner et al., US Patent NO. 4,946,778; Ladner et al., US Patent NO. 5,403,484 and Ladner et al., US Patent NO. 5,571,698) and random peptide display libraries and kits for screening such libraries are available commercially, for instance from Clontech (Palo Alto, CA), Invitrogen Inc. (San Diego, CA), New England Biolabs, Inc.
(Beverly, MA) 10 and Pharmacia LIMB Biotechnology Inc. (Piscataway, NJ). Random peptide display libraries can be screened using the Zalpha53 sequences disclosed herein to identify proteins that bind to Zalpha53. These "binding proteins" which interact with Zalpha53 polypeptides can be used for tagging cells; for isolating homolog polypeptides by affinity purification; they can be directly or indirectly conjugated to drugs, toxins, 15 radionuclides and the like. These binding proteins can also be used in analytical methods such as for screening expression libraries and neutralizing activity.
The binding proteins can also be used for diagnostic assays for determining circulating levels of polypeptides; for detecting or quantitating soluble polypeptides as marker of underlying pathology or disease. These binding proteins can also act as Zalpha53 2 0 "antagonists" to block Zalpha53 binding and signal transduction iyz vitro and irz vivo.
Antibodies are determined to be specifically binding if: (1) they exhibit a threshold level of binding activity, and (2) they do not significantly cross-react with related polypeptide molecules. First, antibodies herein specifically bind if they bind to a Zalpha53 polypeptide, peptide or epitope with a binding affinity (Ka) of 106 M 1 or 2 5 greater, preferably 107 M 1 or greater, more preferably 108 M 1 or greater, and most preferably 109 M 1 or greater. The binding affinity of an antibody can be readily determined by one of ordinary skill in the art, for example, by Scatchard analysis.
Second, antibodies are determined to specifically bind if they do not significantly cross-react with related polypeptides. Antibodies do not significantly 3 0 cross-react with related polypeptide molecules, for example, if they detect Zalpha53 but not known related polypeptides using a standard Western blot analysis (Ausubel et al., ibid.). Examples of known related polypeptides are orthologs, proteins from the same species that are members of a protein family (e.g. IL-16), Zalpha53 polypeptides, and non-human Zalpha53. Moreover, antibodies may be "screened against" known related polypeptides to isolate a population that specifically binds to the inventive polypeptides. For example, antibodies raised to Zalpha53 are adsorbed to related polypeptides adhered to insoluble matrix; antibodies specific to Zalpha53 will flow through the matrix under the proper buffer conditions. Such screening allows isolation of polyclonal and monoclonal antibodies non-crossreactive to closely related 1 o polypeptides, Antibodies: A Laboratory Manual, Harlow and Lane (eds.) (Cold Spring Harbor Laboratory~Press, 1988); Current Protocols in Iminujaology, Cooligan, et al.
(eds.), National Institutes of Health (John Wiley and Sons, Inc., 1995).
Screening and isolation of specific antibodies is well known in the art. See, Fundamental Irratnunology, Paul (eds.) (Raven Press, 1993); Getzoff et al., Adv. iya Iframunol. 43: 1-98 (1988); Monoclonal Antibodies: Principles and Practice, Goding, J.W. (eds.), (Academic Press Ltd., 1996); Benjamin et al., Ann. Rev. Inamunol. 2: 67-101 (1984).
A variety of assays known to those skilled in the art can be utilized to detect antibodies that specifically bind to Zalpha53 proteins or peptides.
Exemplary assays are described in detail in Afatibodies: A Laboratory Manual, Harlow and Lane 2 0 (Eds.) (Cold Spring Harbor Laboratory Press, 1988). Representative examples of such assays include: concurrent immunoelectrophoresis, radioimmunoassay, radioimmuno-precipitation, enzyme-linked immunosorbent assay (ELISA), dot blot or Western blot assay, inhibition or competition assay, and sandwich assay. In addition, antibodies can be screened for binding to wild type versus mutant Zalpha53 protein or polypeptide.
2 5 Antibodies to Zalpha53 may be used for tagging cells that express Zalpha53; for isolating Zalpha53 by affinity purification; for diagnostic assays for determining circulating levels of Zalpha53 polypeptides; for detecting or quantitating soluble Zalpha53 as marker of underlying pathology or disease; in analytical methods employing FAGS; for screening expression libraries; for generating anti-idiotypic 3 0 antibodies; and as neutralizing antibodies or as antagonists to block Zalpha53 in vitro and in vivo. Suitable direct tags or labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent markers, chemiluminescent markers, magnetic particles and the like; indirect tags or labels may feature use of biotin-avidin or other complement/anti-complement pairs as intermediates. Antibodies herein may also be directly or indirectly conjugated to drugs, toxins, radionuclides and the like, and these conjugates used for ih vivo diagnostic or therapeutic applications. Moreover, antibodies to Zalpha53 or fragments thereof may be used in vitro to detect denatured Zalpha53 or fragments thereof in assays, for example, Western Blots or other assays known in the art.
1o BIOACTIVE CONJUGATES:
Antibodies or polypeptides herein can also be directly or indirectly conjugated to drugs, toxins, radionuclides and the like, and these conjugates used for 132 vivo diagnostic or therapeutic applications. For instance, polypeptides or antibodies of the present invention can be used to identify or treat tissues or organs that express a corresponding anti-complementary molecule (receptor or antigen, respectively, for instance). More specifically, Zalpha53 polypeptides or anti-Zalpha53 antibodies, or bioactive fragments or portions thereof, can be coupled to detectable or cytotoxic molecules and delivered to a mammal having cells, tissues or organs that express the anti-complementary molecule.
2 o Suitable detectable molecules may be directly or indirectly attached to the polypeptide or antibody, and include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent markers, chemiluminescent markers, magnetic particles and the like. Suitable cytotoxic molecules may be directly or indirectly attached to the polypeptide or antibody, and include bacterial or plant toxins (for instance, diphtheria 2 5 toxin, Pseudosnonas exotoxin, ricin, abrin and the like), as well as therapeutic radionuclides, such as iodine-131, rhenium-188 or yttrium-90 (either directly attached to the polypeptide or antibody, or indirectly attached through means of a chelating moiety, for instance). Polypeptides or antibodies may also be conjugated to cytotoxic drugs, such as adriamycin. For indirect attachment of a detectable or cytotoxic 3 0 molecule, the detectable or cytotoxic molecule can be conjugated with a member of a complementary/ anticomplementary pair, where the other member is bound to the polypeptide or antibody portion. For these purposes, biotin/streptavidin is an exemplary complementary/ anticomplementary pair.
In another embodiment, polypeptide-toxin fusion proteins or antibody-toxin fusion proteins can be used for targeted cell or tissue inhibition or ablation (for instance, to treat cancer cells or tissues). Alternatively, if the polypeptide has multiple functional domains (i.e., an activation domain or a ligand binding domain, plus a targeting domain), a fusion protein including only the targeting domain may be suitable for directing a detectable molecule, a cytotoxic molecule or a complementary molecule to a cell or tissue type of interest. In instances where the domain only fusion protein includes a complementary molecule, the anti-complementary molecule can be conjugated to a detectable or cytotoxic molecule. Such domain-complementary molecule fusion proteins thus represent a generic targeting vehicle for cell/tissue-specific delivery of generic anti-complementary-detectable/ cytotoxic molecule conjugates. .
In another embodiment, Zalpha53-cytokine fusion proteins or antibody-cytokine fusion proteins can be used for enhancing i~ vivo killing of target tissues (for example, blood and bone marrow cancers), if the Zalpha53 polypeptide or anti-Zalpha53 antibody targets the hyperproliferative blood or bone marrow cell.
See, generally, Hornick et al., Blood 89:4437 (1997). The described fusion proteins enable 2 0 targeting of a cytokine to a desired site of action, thereby providing an elevated local concentration of cytokine. Suitable Zalpha53 polypeptides or anti-Zalpha53 antibodies target an undesirable cell or tissue (i.e., a tumor or a leukemia), and the fused cytokine mediated improved target cell lysis by effector cells. Suitable cytokines for this purpose include interleukin 2 and granulocyte-macrophage colony-stimulating factor (GM-2 5 CSF), for instance.
In yet another embodiment, if the Zalpha53 polypeptide or anti-Zalpha53 antibody targets vascular cells or tissues, such polypeptide or antibody may be conjugated with a radionuclide, and particularly with a beta-emitting radionuclide, to reduce restenosis. Such therapeutic approach poses less danger to clinicians who 3 0 administer the radioactive therapy. For instance, iridium-192 impregnated ribbons placed into scented vessels of patients until the required radiation dose was delivered showed decreased tissue growth in the vessel and greater luminal diameter than the control group, which received placebo ribbons. Further, revascularisation and stmt thrombosis were significantly lower in the treatment group. Similar results are predicted with targeting of a bioactive conjugate containing a radionuclide, as described herein.
The bioactive polypeptide or antibody conjugates described herein can be delivered intravenously, intraarterially or intraductally, or may be introduced locally at the intended site of action.
USES OF POLYNUCLEOTIDElPOLYPEPTIDE:
Molecules of the present invention can be used to identify and isolate receptors involved in spermatogenesis, steroidogenesis, testicular differentiation and regulatory control of the hypothalamic-pituitary-gonadal axis or receptors of the immune system. For example, proteins and peptides of the present invention can be immobilized on a column and membrane preparations run over the column, Izzzzzzobilized Affinity Ligazzd Tec7zniques, Hermanson et al., eds., pp.195-202 (Academic Press, San Diego, CA, 1992,). Proteins and peptides can also be radiolabeled, Methods izz Ezzzyznol., vol. 182, "Guide to Protein Purification", M. Deutscher, ed., pp 721-737 (Acad. Press, San Diego, 1990) or photoaffinity labeled, Brunner et al., Anfz.
Rev.
2 0 Biochem. 62:483-514 (1993) and Fedan et al., Biochezzz. Pharnzacol.
33:1167 (1984) and specific cell-surface proteins can be identified.
The molecules of the present invention will be useful for testing disorders of the reproductive system and immunological systems.
2 5 GENE THERAPY:
Polynucleotides encoding Zalpha53 polypeptides are useful within gene therapy applications where it is desired to increase or inhibit Zalpha53 activity. If a mammal has a mutated or absent Zalp7ia53 gene, the Zalpha53 gene can be introduced into the cells of the mammal. In one embodiment, a gene encoding a Zalpha53 3 0 polypeptide is introduced izz vivo in a viral vector. Such vectors include an attenuated or defective DNA virus, such as, but not linnited to, herpes simplex virus (HSV), papillomavinis, Epstein Barr virus (EBV), adenovirus, adeno-associated virus (AAV), and the like. Defective viruses, which entirely or almost entirely lack viral genes, are preferred. A defective virus is not infective after introduction into a cell.
Use of defective viral vectors allows for administration to cells in a specific, localized area, 5 without concern that the vector can infect other cells. Examples of particular vectors include, but are not limited to, a defective herpes simplex virus 1 (HSV1) vector, Kaplitt et al., Molec. Cell. Neurosci. 2:320 (1991); an attenuated adenovirus vector, such as the vector described by Stratford-Perricaudet et al., J. Clizz.
Itzvest. 90:626 (1992); and a defective adeno-associated virus vector, Samulski et al., J.
Virol. 61:3096 l0 (1987); Samulski et al., J. Virol. 63:3822 (1989).
In another embodiment, a Zalpha53 gene can be introduced in a retroviral vector, e.g., as described in Anderson et al., U.S. Patent No.
5,399,346; Mann et al. Cell 33:153, 1983; Temin et al., U.S. Patent No. 4,650,764; Temin et al:, U.S..
Patent No. 4,980,289; Markowitz et al., J. Virol. 62:1120 (1988); Temin et al., U.S.
15 Patent No. 5,124,263; International Patent Publication No. WO 95/07358, published March 16, 1995 by Dougherty et al.; and I~uo et al., Blood 82:84.5 (1993).
Alternatively, the vector can be introduced by lipofection izz vivo using liposomes.
Synthetic cationic lipids can be used to prepare liposomes for izz vivo transfection of a gene encoding a marker, Felgner et al., Proc. Natl. Acad. Sci. USA 84:7413 (1987);
2 0 Mackey et al., Proc. Natl. Acad. Sci. USA 85:8027 ( 1988). The use of lipofection to introduce exogenous genes into specific organs in vivo has certain practical advantages.
Molecular targeting of liposomes to specific cells represents one area of benefit. More particularly, directing transfection to particular cells represents one area of benefit. For instance, directing transfection to particular cell types would be particularly 2 5 advantageous in a tissue with cellular heterogeneity, such as the pancreas, liver, kidney, and brain. Lipids may be chemically coupled to other molecules for the purpose of targeting. Targeted peptides (e.g., hormones or neurotransmitters), proteins such as antibodies, or non-peptide molecules can be coupled to Iiposomes chemically.
It is possible to remove the target cells from the body; to introduce the 3 0 vector as a naked DNA plasmid; and then to re-implant the transformed cells into the body. Naked DNA vectors for gene therapy can be introduced into the desired host cells by methods known in the art, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAF dextran, calcium phosphate precipitation, use of a gene gun or use of a DNA vector transporter. See, for example, Wu et al., J. Biol.
Cherra.
267:963 (1992); Wu et al., J. Biol. Chefn. 263:14621-4, (1958).
Antisense methodology can be used to inhibit Zalpha53 gene transcription, such as to inhibit cell proliferation in vivo. Polynucleotides that are complementary to a segment of a Zalpha53 polynucleotide (e.g., a polynucleotide as set froth in SEQ m NO: 1 or 19) are designed to bind to Zalpha53-encoding mRNA and to inhibit translation of such mRNA. Such antisense polynucleotides are used to inhibit expression of Zalpha53 polypeptide-encoding genes in cell culture or in a subject.
The present invention also provides reagents that will find use in diagnostic applications. For example, the Zalpha53 gene, a probe comprising Zalplaa53 DNA or RNA or a'subsequence thereof can be used to determine if the Zalpha53 gene is present on chromosome 10 or if a mutation has occurred. Detectable chromosomal aberrations at the Zalpha53 gene locus include, but are not limited to, aneuploidy, gene copy number changes, insertions, deletions, restriction site changes and rearrangements.
Such aberrations can be detected using polynucleotides of the present invention by employing molecular genetic techniques, such as restriction fragment length polymorphism (RFLP) analysis, short tandem repeat (STR) analysis employing PCR
2 0 techniques, and other genetic linkage analysis techniques known in the art (Sambrook et al., ibid.; Ausubel et. aL, ibid.; Marian, Chest 108:255 (1995).
Transgenic mice, engineered to express the Zalpha53 gene, and mice that exhibit a complete absence of Zalpl~a53 gene function, referred to as "knockout mice", Snouwaert et al., Science 257:1053 (1992), may also be generated, Lowell et al., Nature 366:740-42 (1993). These mice may be employed to study the Zalplza53 gene and the protein encoded thereby in an if2 vivo system.
CHROMOSOMAL LOCALIZATION:
Zalplza53 has been mapped to chromosome 10. Radiation hybrid 3 0 mapping is a somatic cell genetic technique developed for constructing high-resolution, contiguous maps of mammalian chromosomes (Cox et al., Scieyace 25D:245 (1990).

Partial or full knowledge of a gene's sequence allows one to design PCR
primers suitable for use with chromosomal radiation hybrid mapping panels. Radiation hybrid mapping panels are commercially available which cover the entire human genome, such as the Stanford G3 RH Panel and the GeneBridge 4 RH Panel (Research Genetics, Inc., Huntsville, AL). These panels enable rapid, PCR-based chromosomal localizations and ordering of genes, sequence-tagged sites (STSs), and other nonpolymorphic and polymorphic markers within a region of interest. This includes establishing directly proportional physical distances between newly discovered genes of interest and previously mapped markers. The precise knowledge of a gene's position can be useful for a number of purposes, including: 1) determining if a sequence is part of an existing contig and obtaining additional surrounding genetic sequences in various forms, such as YACs, BACs or cDNA clones; 2)'providing a possible candidate gene for an inheritable disease which shows linkage to the same chromosomal region; and 3) cross-referencing model organisms, such as mouse, which may aid in determining what function a particular gene might have.
Sequence tagged sites (STSs) can also be used independently for chromosomal localization. An STS is a DNA sequence that is unique in the human genome and can be used as a reference point for a particular chromosome or region of a chromosome. An STS is defined by a pair of oligonucleotide primers that are used in a 2 0 polymerase chain reaction to specifically detect this site in the presence of all other genomic sequences. Since STSs are based solely on DNA sequence they can be completely described within an electronic database, for example, Database of Sequence Tagged Sites (dbSTS), GenBank, (National Center for Biological Information, National Institutes of Health, Bethesda, MD http:llwww.ncbi.nlm.nih.gov), and can be searched 2 5 with a gene sequence of interest for the mapping data contained within these short genomic landmark STS sequences.
For pharmaceutical use, the proteins of the present invention are formulated fox parenteral, particularly intravenous or subcutaneous, delivery according to conventional methods. Intravenous administration will be by bolus injection or 3 0 infusion over a typical period of one to several hours. In general, pharmaceutical formulations will include a Zalpha53 protein in combination with a pharmaceutically acceptable vehicle, such as saline, buffered saline, 5% dextrose in water or the like.
Formulations may further include one or more excipients, preservatives, solubilizers, buffering agents, albumin to prevent protein loss on vial surfaces, etc.
Methods of formulation are well known in the art and are disclosed, for example, in Remington:
The Science afad Pf actice of Phafmacy, Gennaro, ed.,(Mack Publishing Co., Easton, PA, 19th ed,, 1995). Therapeutic doses will generally be in the range of 0.1 to 100 p.g/kg of patient weight per day, preferably 0.5-20 mg/kg per day, with the exact dose determined by the clinician according to accepted standards, taking into account the nature and severity of the condition to be treated, patient traits, etc.
Determination of 1 o dose is within the level of ordinary skill in the art. The proteins may be administered fox acute treatment, over one week or less, often over a period of one to three days or may be used in chronic treatment, over several months or years. Administration of the protein can be subcutaneous, intraperitoneal or rectal depending on the disease to be treated.
Tissue Expression and Use Zalpha53 represents a novel polypeptide with a putative signal peptide leader sequence and alpha helical structure. This gene encodes a secreted polypeptide with secondary structure indicating it is a member of the four-helix bundle cytokine 2 o family. Alternatively, this polypeptide may have other activities associated with other biological functions including: enzymatic activity, association with the cell membrane, or function as a carrier protein.
Use of Zalpha53 2 5 Zalpha53 can be administered to an immunocompromised mammal, preferably a human, such as cancer patients who have undergone chemotherapy, AmS
patients and the elderly. This will stimulate their immune systems. Zalpha53 can also be used as a vaccine adjuvant to be administered before, With or after the administration of a vaccine. Zalpha53 may also be administered to stimulate the immune system to attack 3 0 tumors.

Use of Antagonists of Zalplza53 An antagonist to Zalpha53, such as an antibody, soluble receptor or small molecule antagonist can be administered to a mammal, preferably a human, to alleviate an inflammatory response. Antagonists, such as antibodies, to Zalpha53 can be used to treat patients having inflammatory related diseases such arteriosclerotic heart disease [see Paulsson,G. et al., Arterioscler- Throfnb. Vasc. Biol., 20:10-17 (2000)], inflammatory bowel disease, Crohn's disease, rheumatoid arthritis and pancreatitis.
EDUCATIONAL KTT UTILITY OF ZALPHA53 POLYPEPTIDES, l0 POLYNUCLEOTIDES AND ANTIBODIES
Polynucleotides and polypeptides of the present invention will additionally find use as educational tools as a laboratory practicum kits for courses related to genetics and molecular biology, protein chemistry and antibody production and analysis. Due to its unique polynucleotide and polypeptide sequence molecules of Zalpha53 can be used as standards or as "unknowns" for testing purposes. For example, Zalpha53 polynucleotides can be used as an aid, such as, for example, to teach a student how to prepare expression constructs for bacterial, viral, and/or mammalian expression, including fusion constructs, wherein Zalpha53 is the gene to be expressed;
2 0 for determining the restriction endonuclease cleavage sites of the polynucleotides;
determining mRNA and DNA localization of Zalpha53 polynucleotides in tissues (i.e., by Northern and Southern blotting as well as polymerase chain reaction); and for identifying related polynucleotides and polypeptides by nucleic acid hybridization.
Zalpha53 polypeptides can be used educationally as an aid to teach 2 5 preparation of antibodies; identifying proteins by Western blotting;
protein purification;
determining the weight of expressed Zalpha53 polypeptides as a ratio to total protein expressed; identifying peptide cleavage sites; coupling amino and carboxyl terminal tags; amino acid sequence analysis, as well as, but not limited to monitoring biological activities of both the native and tagged protein (i.e., receptor binding, signal 3 0 transduction, proliferation, and differentiation) in vitro and in vivo.
Zalpha53 polypeptides can also be used to teach analytical skills such as mass spectrometry, circular dichroism to determine conformation, in particular the locations of the disulfide bonds, x-ray crystallography to determine the three-dimensional structure in atomic detail, nuclear magnetic resonance spectroscopy to reveal the structure of proteins in solution. For example, a kit containing the Zalpha53 polypeptide can be given to the 5 student to analyze. Since the amino acid sequence would be known by the professor the protein can be given to the student as a test to determine the skills or develop the skills of the student, the teacher would then know whether or not the student has correctly analyzed the polypeptide. Since every polypeptide is unique, the educational utility of Zalpha53 would be unique unto itself.
10 The antibodies which bind specifically to Zalpha53 can be used as a teaching aid to instruct students how to prepare affinity chromatography columns to purify Zalpha53, cloning and sequencing the polynucleotide that encodes an antibody and thus as a practicum for teaching a student how to design humanized antibodies. The Zalpha53 gene, polypeptide or antibody would then be packaged by reagent companies 15 and sold to universities so that the students gain skill in art of molecular biology.
Because each gene and protein is unique, each gene and protein creates unique challenges and learning experiences for students in a lab practicum. Such educational kits containing the Zalpha53 gene, polypeptide, or antibody, are considered within the scope of the present invention.
2 0 The invention is further illustrated by the following non-limiting examples.
Example 1 Cloning of Zalpha53 2 5 The Zalpha53 cDNA was discovered in a testis cDNA library. The marathon cDNA was made using the marathon-ReadyTM kit (Clontech, Palo Alto, CA) and QC tested with clathrin primers ZC21195 GAGGAGACCATAACCCCCGACAG
(SEQ ID NO: 38) and ZC21196 CATAGCTCCCACCACACGATTTT (SEQ 1l? NO:
39) and then diluted based on the intensity of the clathrin band. To assure quality of the 3 0 panel samples, three tests for quality control (QC) were run: (1) To assess the RNA
quality used for the libraries, the in-house cDNAs were tested for average insert size by PCR with vector oligos that were specific for the vector sequences for an individual cDNA library; (2) Standardization of the concentration of the cDNA in panel samples was achieved using standard PCR methods to amplify full length alpha tubulin or G3PDH cDNA using a 5' vector oligo ZC14,063 CACCAGACATAATAGCTGACAGACT (SEQ ID NO: 40) and 3' alpha tubulin specific oligo primer ZC17,574 GGTRTTGCTCAGCATGCACAC (SEQ ID NO: 41) or 3' G3PDH specific oligo primer ZC17,600 CATGTAGGCCATGAGGTCCACCAC1 (SEQ ID N0:.42); and (3) a sample was sent to sequencing to check for possible ribosomal or mitochondrial DNA
contamination.
1 o The panel was set up in a 96-well format that included a human genomic DNA
(Clontech, Palo Alto, CA) positive control sample. Each well contained approximately 0.2-100 pg/~.l of cDNA. The PCR reactions were set up using oligos ZC37195 GTCCCTGTTTCAGCACATCATC (SEQ D~ NO: 43) and ZC37194 GTCTTCCTCCTCTATCATTTGTTTC (SEQ m NO: 44)., TaKaRa Ex TaqTM
(TAKARA Shuzo Co LTD, Biomedicals Group, Japan), and Rediload dye (Research Genetics, Inc., Huntsville, AL). The amplification was carried out as follows:
1 cycle at 94°C for 2 minutes, 35 cycles of 94°C for 30 seconds, 57.0°C for 30 seconds and 72°C
for 30 seconds, followed by 1 cycle at 72°C for 5 minutes. About 10 ~,l of the PCR
reaction product was subjected to standard Agarose gel electrophoresis using a 4%
2 0 agarose gel. The correctly predicted DNA fragment size of ~272bp was observed in four testis samples.
The DNA fragment for testis cDNA and testis 1K library pool were excised and purified using a Gel Extraction Kit (Qiagen, Chatsworth, CA) according to manufacturer's instructions. The fragment for the testis 1 K library pool was confirmed 2 5 by sequencing to show that it was indeed Zalpha53 but had an insertion of 36bp.
From the foregoing, it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

SEQUENCE LISTING
<110> Gao, Zeren Shoemaker, Kimberly E.
<120> Mammalian Alpha-Helical Protein-53 <130> 00-106PC
<150> 60124-9,686 <151> 2000-11-17 <160> 44 <170> FastSEQ fior Windows Version 3.0 <210>1 <211>774 <212>DNA

<213>Homo Sapiens <220>
<221> CDS
<222> (247)...(753) <400>

ggcccgacctgaagcactggctccagccttagggaaggttttggccgtgggttttgttgg 60 cacgtaccattttgctgaaagacagagcaccttgaggacgttatccctaaaatgagagag 120 gactgggattgaaaggctgactacagaaatggctgctgcccagacgccctcaaaagccaa 180 ggatcctcagrgttggttataaaatatttaagggcgagaaaaggatcgcaggagccaggc 240 cctgag agc ttg tcc ctg atc atc acc gag 288 atg gag ttt cag ttc cac Met Ser Leu Ser Leu e Gln Ile Ile Thr Glu Glu Ph His Phe cat cag gcg gag gag agt cgc cgt ttg atg cga gaa gta agg tcg gaa 336 His Gln Ala Glu Glu Ser Arg Arg Leu Met Arg Glu Val Arg Ser Glu ata acc aga tgt cgt gaa aaa att aag aaa gca acg gag gag ctg aat 384 Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn gaa gag aaa atc aag ctg gaa tct aag gtt caa cag ttt ttt gaa aaa 432 Glu Glu Lys Tle Lys Leu Glu Ser Lys Ual Gln Gln Phe Phe Glu Lys tcc ttc ttc tta cag ctt ttg aaa get cat gaa aat get tta gaa aaa 480 Ser Phe Phe Leu Gln Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys cag tac agt gaa att aca aac cat agg aat atg ctt ctt caa acc ttt 528 Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe gag get ata aag aaa caa atg ata gag gag gaa gac aaa ttt att aag 576 Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp Lys Phe Ile Lys gaa att aca gac ttt aat aat gat tat gaa ata aca aag aaa aga gag 624 Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu ctt ttg atg aaa gaa aat gtc aag att gaa ata tct gac tta gaa aac 672 Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn caa gca aac atg ttg aaa agt ggt atg aat aaa tat cac ctc att tgt 720 Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys ctt gca tta atg aaa ata act tat ttt gaa tga atgaattttc caaaaattta 773 Leu Ala Leu Met Lys Ile Thr Tyr Phe Glu a 774 <210>2 <211>168 <212>PRT

<213>Homo Sapiens <400> 2 Met Ser Leu Glu Ser Leu Phe Gln His Ile Ile Phe Thr Glu His Gln Ala Glu Glu Ser Arg Arg Leu Met Arg Glu Ual Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu Ser Lys Ual Gln Gln Phe Phe Glu Lys Ser Phe Phe Leu Gln Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glw Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asri Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met Lys Ile Thr Tyr Phe Glu <210>3 <211>143 <212>PRT

<213>Homo sapiens <400> 3 Glu Ual Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu Ser Lys Ual Gln Gln Phe Phe Glu Lys Ser Phe Phe Leu Gln Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met Lys Ile Thr Tyr Phe Glu <210>4 <211>16 <212>PRT

<213>Homo sapiens <400> 4 Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu <210>5 <211>15 <212>PRT

<213>Homo sapiens <400> 5 Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn <210>6 <211>15 <212>PRT

<213>Homo Sapiens <400> 6 Gln Thr Phe Glu Ala Ile Lys Lys GIn Met Ile Glu Glu Glu Asp <210>7 <211>16 <212>PRT

<213>Homo Sapiens <400> 7 Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met <210> 8 <211> 28 <212> PRT
<213> Homo sapiens <400> 8 Glu Ual Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Tle Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu <210>9 <211>43 <212>PRT

<213>Homo sapiens <400> 9 GluGlu AsnGlu Glu Lys Lys Leu Glu Ser Lys Val Leu Ile Gln Gln PhePhe LysSer Phe Phe Gln Leu Leu Lys Ala His Glu Leu Glu Asn AlaLeu LysGln Tyr Ser Ile Thr Asn Glu Glu <210>10 <211>42 <212>PRT

<213>Homo Sapiens <400> 10 GluLys TyrSer Ile AsnHis Arg Asn Met Leu Gln Glu Thr Leu Gln ThrPhe AlaIle Lys MetIle Glu Glu Glu Asp Glu Lys Gln Lys Phe IleLys IleThr Phe AsnAsp Glu Asp Asn <210>11 <211>41 <212>PRT

<213>Homo sapiens <400> 11 Glu Glu Glu Asp Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln <210>12 <211>38 <212>PRT

<213>Homo Sapiens <400> 12 Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu 1 5 . 10 15 Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys <210>13 <211>61 <212>PRT

<213>Homo Sapiens <400> 13 ArgSerGlu IleThrArg CysArgGluLys IleLys LysAlaThr Glu GluLeuAsn GluGluLys IleLysLeuGlu SerLys UalGlnGln Phe PheGluLys SerPhePhe LeuGlnLeuLeu LysAla HisGluAsn Ala LeuGluLys GlnTyrSer GluIleThrAsn HisArg Asn <210>14 <211>79 <212>PRT

<213>Homo sapiens <400> 14 Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Giu Lys Ile Lys Leu Glu Ser Lys Ual Gln Gln Phe PheGluLys SerPhe LeuGlnLeuLeu AlaHis Glu Asn Phe Lys Ala LeuGluLys GlnTyr GluIleThrAsn ArgAsn Met Leu Ser His Leu GlnThrPhe GluAla LysLysGlnMet GluGlu Glu Asp Ile Ile <210>15 <211>135 <212>PRT

<213>Homo sapiens <400> 15 ArgSerGlu IleThrArg CysArgGluLys IleLys LysAlaThr Glu GluLeuAsn GluGluLys IleLysLeuGlu SerLys UalGlnGln Phe 20 25 ~ 30 PheGluLys SerPhePhe LeuGlnLeuLeu LysAla HisGluAsn Ala LeuGluLys GlnTyrSer GluIleThrAsn HisArg AsnMetLeu Leu GlnThrPhe GluAlaIle LysLysGlnMet IleGlu GluGluAsp Lys PheIleLys GluIleThr AspPheAsnAsn AspTyr GluIleThr Lys LysArgGlu LeuLeuMet LysGluAsnUal LysIle GluIleSer Asp LeuGluAsn GlnAlaAsn MetLeuLysSer GlyMet AsnLysTyr His LeuIleCys LeuAlaLeu Met <210>16 <211>33 <212>PRT

<213>Homo sapiens <400> 16 Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu 20 25 ' 30 Asp <210>17 <211>89 <212>PRT

<213>Homo sapiens <400> 17 AsnAlaLeu GluLysGln TyrSerGluIle ThrAsn HisArgAsn Met LeuLeuGln ThrPheGlu AlaIleLysLys GlnMet IleGluGlu Glu AspLysPhe IleLysGlu IleThrAspPhe AsnAsn AspTyrGlu Ile ThrLysLys ArgGluLeu LeuMetLysGlu AsnUal LysTleGlu Ile SerAspLeu GluAsnGln AlaAsnMetLeu LysSer GlyMetAsn Lys TyrHisLeu IleCysLeu AlaLeuMet <210>18 <211>72 <212>PRT

<213>Homo sapiens <400> 18 LeuGlnThr PheGluAla IleLysLysGln MetIle GluGluGlu Asp LysPheIle LysGluIle ThrAspPheAsn AsnAsp TyrGluIle Thr LysLysArg GluLeuLeu MetLysGluAsn UalLys IleGluIle Ser AspLeuGlu AsnGlnAla AsnMetLeuLys SerGly MetAsnLys Tyr HisLeuIle CysLeuAla LeuMet <210>19 <211>738 <212>DNA

<213>Homo sapiens <220>
<221> CDS
<222> (247)...(717) <400>

ggcccgacctgaagcactggctccagccttagggaaggttttggccgtgggttttgttgg 60 cacgtaccattttgctgaaagacagagcaccttgaggacgttatccctaaaatgagagag 120 gactgggattgaaaggctgactacagaaatggctgctgcccagacgccctcaaaagccaa 180 ggatcctcagggttggttataaaatatttaagggcgagaaaaggatcgcaggagccaggc 240 cctgag agc ttg tcc ctg atc atc acc gag 288 atg gag ttt cag ttc cac Met Ser Leu Ser Leu e Gln Ile Tle Thr Glu Glu Ph His Phe cat cag gcg gag gag agt cgc cgt ttg atg cga gaa gta agg tcg gaa 336 His Gln Ala Glu Glu Ser Arg Arg Leu Met Arg Glu Ual Arg Ser Glu ata acc aga tgt cgt gaa aaa att aag aaa gca acg gag gag ctg aat - 384 Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn gaa gag aaa atc aag ctg gaa tct aag ctt ttg aaa get cat gaa aat 432 Glu Glu Lys Ile Lys Leu Glu Ser Lys Leu Leu Lys Ala His Glu Asn get tta gaa aaa cag tac agt gaa att aca aac cat agg aat atg ctt 480 Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu ctt caa acc ttt gag get ata aag aaa caa atg ata gag gag gaa gac 528 Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp aaa ttt att aag gaa att aca gac ttt aat aat gat tat gaa ata aca 576 Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr aag aaa aga gag ctt ttg atg aaa gaa aat gtc aag att gaa ata tct 624 Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser gac tta gaa aac caa gca aac atg ttg aaa agt ggt atg aat aaa tat 672 Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr cac ctc att tgt ctt gca tta atg aaa ata act tat ttt gaa tga 717 His Leu Ile Cys Leu Ala Leu Met Lys Ile Thr Tyr Phe Glu atgaattttc caaaaattta a 738 <210>20 <211>156 <212>PRT

<213>Homo sapiens <400> 20 MetSerLeu GluSer LeuPheGlnHis IleIlePhe ThrGlu HisGln AlaGluGlu SerArg ArgLeuMetArg GluUalArg SerGlu IleThr ArgCysArg GluLys IleLysLysAla ThrGluGlu LeuAsn GluGlu LysIleLys LeuGlu SerLysLeuLeu LysAlaHis GluAsn AlaLeu GluLysGln TyrSer GluTleThrAsn HisArgAsn MetLeu LeuGln ThrPheGlu AlaIle LysLysGlnMet IleGluGlu GluAsp LysPhe IleLysGlu IleThr AspPheAsnAsn AspTyrGlu IleThr LysLys ArgGluLeu LeuMet LysGluAsnUal LysIleGlu IleSer AspLeu GluAsnGln AlaAsn MetLeuLysSer GlyMetAsn LysTyr HisLeu IleCysLeu AlaLeu MetLysIleThr TyrPheGlu <210> 21 <211> 131 <212> PRT
<213> Homo sapiens <400> 21 Glu Ual Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu Ser Lys Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met Lys Ile Thr Tyr Phe Glu <210>22 <211>45 <212>PRT

<213>Homo Sapiens <400> 22 GluUal SerGlu Ile Thr Cys ArgGlu Lys Ile Lys Lys Arg Arg Ala ThrGlu LeuAsn Glu Glu Ile LysLeu Glu Ser Lys Leu Glu Lys Leu LysAla GluAsn Ala Leu Lys GlnTyr Ser Glu His Glu <210>23 <211>54 <212>PRT

<213>Homo sapiens <400> 23 ArgGlu IleLysLys Ala GluGlu Leu Asn GluLys Lys Thr Glu Ile LysLeu SerLysLeu Leu AlaHis Glu Asn LeuGlu Glu Lys Ala Lys GlnTyr GluIleThr Asn ArgAsn Met Leu GlnThr Ser His Leu Phe Glu Ala Ile Lys Lys Gln <210>24 <211>54 <212>PRT

<213>Homo Sapiens <400> 24 GluSer LysLeuLeu LysAla GluAsn Leu Lys Gln His Ala Glu Tyr SerGlu IleThrAsn HisArg MetLeu Gln Phe Glu Asn Leu Thr Ala IleLys LysGlnMet IleGlu GluAsp Phe Lys Glu Glu Lys Ile Ile ThrAsp PheAsnAsn Asp <210>25 <211>45 <212>PRT

<213>Homo Sapiens <400> 25 GluIle AsnHis Asn Met LeuGln Thr Phe Glu Ala Thr Arg Leu Ile LysLys MetIle Glu Glu LysPhe Ile Lys Glu Ile Gln Glu Asp Thr AspPhe AsnAsp Glu Ile LysLys Arg Glu Asn Tyr Thr <210>26 <211>42 <212>PRT

<213>Homo Sapiens <400> 26 LysLys MetIle Glu Glu Glu Lys Phe Ile Lys Glu Gln Asp Ile Thr AspPhe AsnAsp Tyr Glu Ile Lys Lys Arg Glu Leu Asn Thr Leu Met LysGlu ValLys Ile Glu Ile Asp Asn Ser <210>27 <211>38 <212>PRT

<213>Homo sapiens <400> 27 AsnAsn Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Asp Met Lys Glu AsnVal Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Lys Asn Met Leu LysSer Met Asn Lys Gly <210>28 <211>16 <212>PRT

<213>Homo sapiens <400> 28 Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu <210>29 <211>16 <212>PRT

<213>Homo Sapiens <400> 29 Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn <210>30 <211>16 <212>PRT

<213>Homo Sapiens <400> 30 Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp <210> 31 <211> 16 <212> PRT
<213> Homo sapiens <400> 31 Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met 1 5 ~ 10 15 <210>32 <211>49 <212>PRT

<213>Homo sapiens <400> 32 ArgSer IleThr Cys GluLys Ile Lys AlaThr Glu Arg Arg Lys Glu GluLeu GluGlu Ile LeuGlu Ser Lys LeuLys Asn Lys Lys Leu Ala HisGlu AlaLeu Lys TyrSer Glu I'le AsnHis Asn Glu Gln Thr Arg Asn <210>33 <211>67 <212>PRT

<213>Homo sapiens <400> 33 Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu Ser Lys Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp <210>34 <211>123 <212>PRT

<213>Homo Sapiens 15 , <400> 34 Arg Ser Glu Ile Thr Arg Cys Arg Glu Lys Ile Lys Lys Ala Thr Glu Glu Leu Asn Glu Glu Lys Ile Lys Leu Glu Ser Lys Leu Leu Lys Ala His Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn Nis Arg Asn Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp Lys Phe Ile Lys Glu Ile Thr Asp Phe Asn Asn Asp Tyr Glu Ile Thr Lys Lys Arg Glu Leu Leu Met Lys Glu Asn Ual Lys Ile Glu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met <210>35 <211>34 <212>PRT

<213>Homo sapiens <400> 35 Glu Asn Ala Leu Glu Lys Gln Tyr Ser Glu Ile Thr Asn His Arg Asn 1 5 l0 15 Met Leu Leu Gln Thr Phe Glu Ala Ile Lys Lys Gln Met Ile Glu Glu Glu Asp <210>36 <211>90 <212>PRT

<213>Homo sapiens <400> 36 GluAsnAla LeuGluLysGln TyrSerGlu IleThrAsn HisArgAsn MetLeuLeu GlnThrPheGlu AlaIleLys LysGlnMet IleGluGlu GluAspLys PheIleLysGlu IleThrAsp PheAsnAsn AspTyrGlu IleThrLys LysArgGluLeu LeuMetLys GluAsnUal LysIleGlu Ile Ser Asp Leu Glu Asn Gln Ala Asn Met Leu Lys Ser Gly Met Asn Lys Tyr His Leu Ile Cys Leu Ala Leu Met <210>37 <211>72 <212>PRT

<213>Homo Sapiens <400> 37 LeuGlnThrPheGlu AlaTle LysLysGlnMet IleGluGlu GluAsp LysPheIleLysGlu IleThr AspPheAsnAsn AspTyrGlu IleThr LysLysArgGluLeu LeuMet LysGluAsnUal LysIleGlu IleSer AspLeuGluAsnGln AlaAsn MetLeuLysSer GlyMetAsn LysTyr HisLeuIleCysLeu AlaLeu Met <210>38 <211>23 <212>DNA

<213>Nomo Sapiens <400> 38 gaggagacca taacccccga cag 23 <210>39 <211>23 <212>DNA

<213>Homo sapiens <400> 39 catagctccc accacacgat ttt 23 <210>40 <211>25 <212>DNA

<213>Homo Sapiens <400> 40 caccagacat aatagctgac agact 25 <210>41 <211>21 <212>DNA

<213>Homo sapiens <400> 41 ggtrttgctc agcatgcaca c 21 <210>42 <211>24 <212>DNA

<213>Homo sapiens <400> 42 catgtaggcc atgaggtcca ccac 24 <210>43 <211>22 <212>DNA

<213>Homo sapiens <400> 43 gtccctgttt cagcacatca tc 22 <210>44 <211>25 <212>DNA

<213>Homo sapiens <400> 44 gtcttcctcc tctatcattt gtttc 25

Claims (6)

What is claimed is:
1. An isolated polypeptide comprised of an amino acid sequence selected from the group consisting of SEQ ID NOs: 2-18 and 20-37 or a polypeptide which is at least 90% identical to said polypeptide.
2. An isolated polynucleotide which encodes a polypeptide, wherein said polypeptide is comprised of an amino acid sequence selected from the group consisting of SEQ ID NOs: 2-18 and 20-37 or a polypeptide which is at least 90% identical to said polypeptide.
3. An antibody which specifically binds to a polypeptide, wherein said polypeptide is comprised of an amino acid sequence selected from the group consisting of SEQ ID NOs: 2-18 and 20-37.
4. An anti-idiotypic antibody which specifically binds to an antibody which specifically binds to a polypeptide, wherein said polypeptide is comprised of an amino acid sequence selected from the group consisting of SEQ ID NOs: 2-18 and 20-37.
5. The use of an antibody to a polypeptide, wherein the polypeptide is selected from the group consisting of SEQ ID NOs: 2-18 and 20-37, for the production of a medicament for the treatment of an inflammatory disease.
6. The use of a polypeptide comprised of SEQ ID NO:3 to stimulate the immune system.
CA002429265A 2000-11-17 2001-11-14 Mammalian alpha-helical protein-53 Abandoned CA2429265A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US24968600P 2000-11-17 2000-11-17
US60/249,686 2000-11-17
PCT/US2001/043884 WO2002079248A2 (en) 2000-11-17 2001-11-14 Mammalian alpha-helical protein-53

Publications (1)

Publication Number Publication Date
CA2429265A1 true CA2429265A1 (en) 2002-10-10

Family

ID=22944557

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002429265A Abandoned CA2429265A1 (en) 2000-11-17 2001-11-14 Mammalian alpha-helical protein-53

Country Status (5)

Country Link
EP (1) EP1355937A2 (en)
JP (1) JP2004526446A (en)
AU (1) AU2001297949A1 (en)
CA (1) CA2429265A1 (en)
WO (1) WO2002079248A2 (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1290217A2 (en) * 2000-02-04 2003-03-12 Aeomica, Inc. Methods and apparatus for predicting, confirming, and displaying functional information derived from genomic sequence
AU2001243693A1 (en) * 2000-03-17 2001-10-03 Zymogenetics Inc. Helical protein zalpha51

Also Published As

Publication number Publication date
JP2004526446A (en) 2004-09-02
EP1355937A2 (en) 2003-10-29
WO2002079248A2 (en) 2002-10-10
AU2001297949A1 (en) 2002-10-15
WO2002079248A3 (en) 2003-09-04

Similar Documents

Publication Publication Date Title
US7084253B2 (en) Protease-activated receptor PAR4 (ZCHEMR2)
AU744152B2 (en) Beta-defensins
CA2350623A1 (en) Mammalian chondromodulin-like protein
CA2429265A1 (en) Mammalian alpha-helical protein-53
US20040058354A1 (en) Mammalian alpha-helical protein-53
US6420525B1 (en) Human transcription factor ZGCL-1
US20030207793A1 (en) Secreted alpha-helical protein - 32
CA2360584A1 (en) Mammalian alpha-helical protein - 12
US7122342B1 (en) Protease-activated receptor PAR4 (ZCHEMR2)
CA2374520A1 (en) Secreted alpha-helical protein - 32
CA2415095A1 (en) Secreted alpha-helical protein-36
WO1998054326A1 (en) Human chemokine zchemo-8
WO2001004307A1 (en) Alpha protein - 27
MXPA01007161A (en) Mammalian alpha-helical protein - 12
US20030171542A1 (en) Mammalian secretory peptide - 9
US20030032167A1 (en) Secreted alpha-helical protein zlmda24
AU2023200A (en) Human secretory protein-61
MXPA01004744A (en) Human secretory protein-61
EP1323823A2 (en) Mammalian secretory peptide 9, antibodes against it and their use

Legal Events

Date Code Title Description
FZDE Discontinued