Polynucleotides and polypeptides encoded thereby

Abstract
The present invention provides novel polypeptides, termed PROTX polypeptides, as well as polynucleotides encoding PROTX polypeptides and antibodies that immunospecifically bind to a PROTX or a derivative, variant, mutant, or fragment of a PROTX polypeptide, polynucleotide or antibody. The invention additionally provides methods in which the PROTX polypeptide, polynucleotide and antibody are used in detection and treatment of a broad range of pathological states, as well as to other uses.
Description


FIELD OF THE INVENTION

[0002] The invention relates in general to polynucleotides and polypeptides. The invention relates more particularly to polynucleotide sequences and the membrane-bound or secreted polypeptides encoded by such polynucleotides, as well as vectors, host cells, antibodies and recombinant methods for producing the polypeptides and polynucleotides.



SUMMARY OF THE INVENTION

[0003] The present invention is based in part upon the discovery of novel human polynucleotide sequences and the membrane-bound or secreted polypeptides encoded by these sequences. These human nucleic acids and polypeptides encoded thereby are collectively referred to herein as “PROTX”.


[0004] Accordingly, in one aspect, the invention provides an isolated nucleic acid molecule that encodes a novel polypeptide, or a fragment, homolog, analog or derivative thereof. The nucleic acid can include, e.g., a nucleic acid sequence encoding a polypeptide at least 85% identical to a polypeptide comprising the amino acid sequences of SEQ ID NO:2, 4, 6, or 8, or a polypeptide that is a fragment, homolog, analog or derivative thereof. The nucleic acid can include, e.g., one or more fragments from genomic DNA, or a cDNA molecule, or an RNA molecule. In particular embodiments, the nucleic acid molecule may include the sequence of any of SEQ ID NO:1, 3, 5, or 7.


[0005] Also included in the invention is a vector containing one or more of the nucleic acids described herein, and a cell containing the vectors or nucleic acids described herein.


[0006] The invention is also directed to host cells transformed with a vector comprising any of the nucleic acid molecules described above.


[0007] In another aspect, the invention includes a pharmaceutical composition that includes a PROTX nucleic acid and a pharmaceutically acceptable carrier or diluent.


[0008] In a further aspect, the invention includes a substantially purified PROTX polypeptide, e.g., any of the PROTX polypeptides encoded by a PROTX nucleic acid, and fragments, homologs, analogs, and derivatives thereof. The invention also includes a pharmaceutical composition that includes a PROTX polypeptide and a pharmaceutically acceptable carrier or diluent.


[0009] In a still further aspect, the invention provides an antibody that binds specifically to a PROTX polypeptide. The antibody can be, e.g., a monoclonal or polyclonal antibody, and fragments, homologs, analogs, and derivatives thereof. The invention also includes a pharmaceutical composition including PROTX antibody and a pharmaceutically acceptable carrier or diluent. The invention is also directed to isolated antibodies that bind to an epitope on a polypeptide encoded by any of the nucleic acid molecules described above.


[0010] The invention also includes kits comprising any of the pharmaceutical compositions described above.


[0011] The invention further provides a method for producing a PROTX polypeptide by providing a cell containing a PROTX nucleic acid, e.g., a vector that includes a PROTX nucleic acid, and culturing the cell under conditions sufficient to express the PROTX polypeptide encoded by the nucleic acid. The expressed PROTX polypeptide is then recovered from the cell. Preferably, the cell produces little or no endogenous PROTX polypeptide. The cell can be, e.g., a prokaryotic cell or eukaryotic cell.


[0012] The invention is also directed to methods of identifying PROTX polypeptides or nucleic acids in a sample by contacting the sample with a compound that specifically binds to the polypeptide or nucleic acid, and detecting complex formation, if present.


[0013] The invention further provides methods of identifying a compound that modulates the activity of a PROTX polypeptide by contacting PROTX polypeptide with a compound and determining whether the PROTX polypeptide activity is modified.


[0014] The invention is also directed to compounds that modulate PROTX polypeptide activity identified by contacting a PROTX polypeptide with the compound and determining whether the compound modifies activity of the PROTX polypeptide, binds to the PROTX polypeptide, or binds to a nucleic acid molecule encoding a PROTX polypeptide.


[0015] In another aspect, the invention provides a method of determining the presence of or predisposition to a PROTX-associated disorder in a subject. The method includes providing a sample from the subject and measuring the amount of PROTX polypeptide in the subject sample. The amount of PROTX polypeptide in the subject sample is then compared to the amount of PROTX polypeptide in a control sample. An alteration in the amount of PROTX polypeptide in the subject protein sample relative to the amount of PROTX polypeptide in the control protein sample indicates the subject has pathology related to a dysfunction in the immune system, a tissue proliferation-associated condition, or a neurological disorder. A control sample is preferably taken from a matched individual, i.e., an individual of similar age, sex, or other general condition but who is not suspected of having a dysfunction in the immune system, a tissue proliferation-associated condition, or a neurological disorder. Alternatively, the control sample may be taken from the subject at a time when the subject is not suspected of having a dysfunction in the immune system, a tissue proliferation-associated condition, or a neurological disorder. In some embodiments, the PROTX polypeptide is detected using a PROTX antibody.


[0016] In a further aspect, the invention provides a method of determining the presence of, or predisposition to a PROTX-associated disorder in a subject. The method includes providing a nucleic acid sample, e.g., RNA or DNA, or both, from the subject and measuring the amount of the PROTX nucleic acid in the subject nucleic acid sample. The amount of PROTX nucleic acid sample in the subject nucleic acid is then compared to the amount of PROTX nucleic acid in a control sample. An alteration in the amount of PROTX nucleic acid in the sample relative to the amount of PROTX in the control sample indicates the subject has a dysfunction in the immune system, a tissue proliferation-associated condition, or a neurological disorder.


[0017] In a still further aspect, the invention provides a method of treating or preventing or delaying a PROTX-associated disorder. The method includes administering to a subject in which such treatment or prevention or delay is desired a PROTX nucleic acid, a PROTX polypeptide, or a PROTX antibody in an amount sufficient to treat, prevent, or delay an immune disorder, a tissue proliferation-associated disorder, or a neurological disorder in the subject.


[0018] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.


[0019] Other features and advantages of the invention will be apparent from the following detailed description and claims.







BRIEF DESCRIPTION OF THE DRAWINGS

[0020]
FIG. 1 is a hydropathy plot of a disclosed PROT1 polypeptide sequence.


[0021]
FIG. 2 is a hydropathy plot of a disclosed PROT2 polypeptide sequence.


[0022]
FIG. 3 is a hydropathy plot of a disclosed PROT3 polypeptide sequence.


[0023]
FIG. 4 is a hydropathy plot of a disclosed PROT4 polypeptide sequence.







DETAILED DESCRIPTION OF THE INVENTION

[0024] The invention provides novel polypeptides and nucleotides encoded thereby. Included in the invention are four novel nucleic acid sequences and their encoded polypeptides. The sequences are collectively referred to as “PROTX nucleic acids” or “PROTX polynucleotides” and the corresponding encoded polypeptide is referred to as an “PROTX polypeptide” or “PROTX protein”. Unless indicated otherwise, “PROTX” is meant to refer to any of the sequences disclosed herein.


[0025] Table 1 provides a summary of the PROTX nucleic acids and their encoded polypeptides.
1TABLE 1SEQ IDNOSEQ IDClone IdentificationNucleicNOPROTX No.NumberAcidPolypeptide1AL132780_A122AC016900_A3432826468.0.39564AC015862_A78


[0026] PROTX nucleic acids and their encoded polypeptides, according to the invention, are useful in a variety of applications and contexts. The various PROTX nucleic acids and polypeptides according to the invention are useful, inter alia, as novel members of protein families according to the presence of domains and sequence relatedness to previously described proteins.


[0027] For example, the PROT1 nucleic acid and its encoded polypeptide include structural motifs that are characteristic of LIM domain-containing proteins. LIM domain-containing proteins contribute to cell fate determination, the regulation of cell proliferation and differentiation, and remodeling of the cell cytoskeleton. These proteins are found in the cell nucleus, the cytoplasm, or both. Cytoplasmic LIM proteins are implicated in the cellular response to extracellular stimuli. For example, Ajuba, a LIM protein, interacts with Grb2 and augments mitogen-activated protein kinase activity in fibroblasts, and promotes meiotic maturation of Xenopus oocytes in a Grb2- and Ras-dependent manner.


[0028] Ajuba, although predominantly a cytosolic protein, in contrast to other like proteins does not localize to sites of cellular adhesion to extracellular matrix or interact with the actin cytoskeleton. Removal of the pre-LIM domain of Ajuba, including a putative nuclear export signal, results in an accumulation of the LIM domains in the cell nucleus. The pre-LIM domain contains two putative proline-rich SH3 recognition motifs. Ajuba specifically associated with Grb2 in vitro and in vivo. The interaction between these proteins was mediated by the SH3 domain of Grb2 and the N-terminal proline-rich pre-LIM domain of Ajuba. In fibroblasts expressing Ajuba mitogen-activated protein kinase activity persists despite serum starvation and upon serum stimulation generates levels fivefold higher than that seen in control cells. Finally, when Ajuba is expressed in fully developed Xenopus oocytes, it promotes meiotic maturation in a Grb2- and Ras-dependent manner.


[0029] A cDNA encoding the AJUBA-like protein, PROT1 is useful in gene therapy, and the AJUBA-like protein is useful when administered to a subject in need thereof. By way of a nonlimiting example, the compositions of the present invention have efficacy for treatment of patients suffering from Central Nervous System Disorders, Cancer, Parkinson's Disease, Alzheimer's Disease, Neuromuscular and Cardiac Disorders. The novel nucleic acid encoding this AJUBA-like protein, and the AJUBA-like protein of the invention, or fragments thereof, are useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These materials are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods.


[0030] PROT2 nucleic acids and encoded polypeptides include structural motifs that are characteristic of proteins belonging to the keratin family of proteins. Keratin 8 is a type II keratin and Endo A is the mouse equivalent. Endo B, which is the equivalent of human keratin 18, a type I keratin, is coexpressed with Endo A. Endo A and Endo B are the first intermediate filament (IF) proteins expressed during murine development.


[0031] Keratins 8 and 18 of simple epithelia differ from the keratins of stratified epithelium in tissue expression and regulation. A single active gene for keratin 8 is located on chromosome 12. This chromosome contains several genes for type II keratins and also the gene for keratin 18, the type I keratin that is coexpressed with keratin 8. This location of both members of a keratin pair on a single chromosome is unique among keratin genes.


[0032] Transgenic mice containing the human KRT8 gene, express a moderately increased amount of keratin in simple epithelia compared to normal mice. The mice expressing human KRT8 display progressive exocrine pancreas alterations, including dysplasia and loss of acinar architecture, redifferentiation of acinar to ductal cells, inflammation, fibrosis, and substitution of exocrine by adipose tissue, as well as increased cell proliferation and apoptosis. The phenotype is very similar to that of transgenic mice expressing a dominant-negative mutant TGF-beta type II receptor (TGFbR2). TGFbR2 mutant mice also have elevated KRT8/KRT18 levels. Thus, simple epithelial keratins play a relevant role in the regulation of exocrine pancreas homeostasis and the disruption of mechanisms that normally regulate keratin expression in vivo are related to inflammatory and neoplastic pancreatic disorders. Accordingly, PROT2 nucleic acids and polypeptides are useful for treating patients suffering from inflammatory and neoplastic pancreatic disorders. The novel nucleic acid encoding Cytokeratin 8-like protein, and the Cytokeratin 8-like protein of the invention, or fragments thereof, are also useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. PROT2 nucleic acid and polypeptide are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for the use in therapeutic or diagnostic methods.


[0033] PROT4 nucleic acids and encoded polypeptides include structural motifs that are characteristic of proteins belonging to the endothelin family of proteins. The endothelins are a family of structurally and pharmacologically distinct peptides. Three isoforms of human endothelin have been identified: endothelins-1, -2, and -3. Endothelin-1 is a potent, 21-amino acid vasoconstrictor peptide produced by vascular endothelial cells. Endothelin-1 was originally isolated from the supernatant of porcine aortic endothelial cell cultures and is the most potent vasoconstrictor known. Subsequent cloning and sequence analysis from a human placental cDNA library showed that human endothelin-1 is identical to porcine endothelin. In addition to its vasoconstrictor action, endothelin has effects on the central nervous system and on neuronal excitability. Endothelin-1 is also expressed in distinct neuronal cell types of the dorsal ganglia and spinal cord.


[0034] The PROT4 nucleic acids and proteins are useful in potential therapeutic applications implicated in cardiovascular diseases, neurological diseases, hypertension and/or other pathologies and disorders. For example, a cDNA encoding the endothelin-like protein may be useful in gene therapy, and the endothelin-like protein may be useful when administered to a subject in need thereof. By way of nonlimiting example, the PROT4 compositions will have efficacy for treatment of patients suffering from hypertention, other cardiovascular diseases, neurological diseases and cancer. The novel nucleic acid encoding endothelin-like protein, and the endothelin-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These materials are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. 1. PROT1. Clone AL132780_A, a Novel Ajuba-like Lim Protein


[0035] A PROT1 nucleic acid and polypeptide according to the invention includes the nucleic acid and encoded polypeptide sequence of clone AL132780_A.


[0036] A polynucleotide of the present invention has been identified as clone AL132780_A. AL132780_A is a full-length clone of 1902 nucleotides, including the entire coding sequence of a protein from nucleotides 272 to 1888 (also referred to herein as “AL132780_A protein”). The clone was originally obtained from genomic DNA.


[0037] The nucleotide sequence of AL132780_A is reported in SEQ ID NO:1. The predicted amino acid sequence of the AL132780_A protein is reported in SEQ ID NO:2. The disclosed protein is 538 amino acids, has a moleucular weight of 56933.2. Psort Analysis predicts that the encoded protein localizes to the cytoplasm (certainty of 0.45), the microbody (certainty of 0.3), mitochoncdrial matrix space (certainty of 0.1), and/or lysozomal lumen (certainty of 0.1). A hydrophobocity plot of the encoded protein is shown in FIG. 1.


[0038] The disclosed PROTI nucleic acid sequence has 1571 bases of 1891 (83%) identical to a Mus musculus AJUBA mRNA (GENBANK-ID: MMU79776|acc:U79776) (Table 2A). The full amino acid sequence of the protein of the invention was found to have 502 of 547 amino acid residues (91%) identical to, and 510 of 547 residues (93%) positive with, the 538 amino acid residue protein from Mus musculus (ptnr: SPTREMBL-ACC:P97472) (Table 2B).


[0039] A multiple sequence alignment is given in Table 3, with the protein of the invention being shown on line 2, in a ClustalW analysis comparing the protein of the invention with related protein sequences.


[0040] The AL132780_A nucleic acid has the following sequence:
2ACGCGTGAACAGATAGACCTGCGGACTGGACAGCCGCGGCCAGAGACCCTGCTAGCCCCGCTCAGCCCCAGATGCGCGGGGGGACGCAGCCCCTCCCGCTGGGGGATGCTGTGGGATTCCTGGCGCAGGGCATCCAGGCCGCCCGCTAAGCCCCTGTGCCTCCCCTGTGCCCCTGGGGAACCAGAGTCCGGCTGCAGGGAAAGAGAACCGGCCGCCGAGACGCCGCAGGGTGCCAGGCGGGGAGGGGGCGAGAGGCCCCAGGCCCGAGGGCATGGAGCGGTTAGGAGAGAAGCCAGTCGCCTGCTGGAGAAGTTCGGCCGCAGAAAGGGTGAATCTAGCCGGTCTGGGTCTGACGGGACCCCCGGGCCGGGCAAGGGGCGCCTAAGTGGGTTGGGGGGACCTAGGAAGTCAGGGCCCCGAGGAGCTACTGGGGGACCTGGGGATGAGCCGTTGGAGCCGGCCCGGGAGCAAGGTTCCCTGGACGCTGAGCGAAATCAGCGCGGCTCCTTTGAGGCGCCGCGCTACG~GGCTCTTTTCCCGCGGGGCCGCCGCCCACCCGGGCCTTGCCTCTACCTCAGTCGTTGCCCCCCGATTTTCGGCTGGAGCCCACGGCCCCGGCCCTCAGCCCCCGCTCTAGCTTCGCCAGTAGCTCGGCCAGCGACGCGAGCAAGCCGTCCAGCCCCCGGGGCAGCCTGCTGCTGGACGGGGCGGGGGCTGGCGGAGCTGGAGGTAGCCGGCCCTGCAGCAATCGCACCAGCGGATCAGCATGGGCTACGACCAGCGCCACGGGAGCCCCTTGCCAGCGGGGCCGTGCCTGTTTGGCCCACCCCTGGCCGGAGCACCGGCAGGCTATTCTCCCGGAGGGGTCCCGTCCGCCTACCCGGAGCTCCACGCCGCCCTGGACCGATTGTACGCTCAGCGGCCCGCGGGGTTCGGCTGCCAGGAAAGCCGCCACTCGTATCCCCCGGCCCTGGGCAGCCCTGGAGCTCTAGCCGGGGCCGGAGTGGGAGCGGCGGGGCCCTTGGAGAGACGGGGGGCGAACCCGGACGACACTCTGTGACCGGCTACGGGGACTGCGCCGTGGGCGCCCGGTACCAGGACGAGCTAACAGCTTTGCTTCGCCTGACGGTGGGCACCGGTGGGCGAGAAGCCGGAGCCCGCGGAGAACCCTCGGGGATTGAGCCGTCGGGTCTGGAGGAGCCACCAGGTCCTTTCGTTCCGGAGGCCGCCCGGGCCCGGATGCGGGAGCCAGAGGCCAGGGAGGACTACTTCGGCACCTGTATCAAGTGCAACAAGGCATCTATGGGCAGAGCAATGCCTGCCAGGCCCTGGACAGCCTCTACCACACCCAGTGCTTTGTTTGCTGCTCTTGTGGGCGAACTTTGCGTTGCAAGGCTTTCTACAGTGTCAATGGCTCTGTGTACTGTGAGGAAGATTATCTGTTTTCAGGGTTTCAGGAGGCAGCTGAGAATGCTGTGTCTGTGGTCACTTGATTTTGGAGAAGATCCTACAAGCAATGGGGAAGTCCTATCATCCAGGCTGTTTCCGATGCATTGTTTGCAACAAGTGCCTGGATGGCATCCCCTTCACAGTGGACTTCTCCAACCAAGTATACTGTGTCACCGACTACCACAAAATTATGCTCCTAAGTGTGCAGCCTGTGGCCAACCCATCCTCCCCTCTGAGGGCTGTGAGGACATCGTGAGGGTGATATCCATGGACCGGGATTATCACTTTGAGTGCTACCACTGTGAGGACTGCCGGATGCAGCTGAGTGATGAGGAAGGCTGCTGCTGTTTCCCTCTGGATGGGCACTTGCTCTGCCATGGTTGCCACATGCAGCGGCTCAATGCCCGACAACCCCCTGCCAACTATATCTGAGCTGCAATCACTGC (SEQ ID NO:1)


[0041] The AL132780_A polypeptide has the following sequence:
3MERLGEKASRLLEKFGRRKGESSRSGSDGTPGPGKGRLSGLGGPRKSGPRGATGGPGDE(SEQ ID NO:2)PLEPAREQGSLDAERNQRGSFEAPRYEGSFPAGPPPTRALPLPQSLPPDFRLEPTAPALSPRSSFASSSASDASKPSSPRGSLLLDGAGAGGAGGSRPSCSNRTSGISMGYDQRHGSPLPAGPCLFGPPLAGAPAGYSPGGVPSAYPELHAALDRLYAQRPAGFGCQESRHSYPPALGSPGALAGAGVGAAGPLERRGAQPGRHSVTGYGDCAVGARYQDELTALLRLTVGTGGREAGARGEPSGIEPSGLEEPPGPFVPEAARARMREPEAREDYFGTCIKCNKGIYGQSNACQALDSLYHTQCFVCCSCGRTLRCKAFYSVNGSVYCEEDYLFSGFQEAAEKCCVCGHLILEKILQAMGKSYHPGCFRCIVCNKCLDGIPFTVDFSNQVYCVTDYHKNYAPKCAACGQPILPSEGCEDIVRVISMDRDYHFECYHCEDCRMQLSDEEGCCCFPLDGHLLCHGCHMQRLNARQPPANYI


[0042] The PROT1 sequence disclosed in SEQ ID NO:1 is highly homologous to the Mus musculus Ajuba mRNA (GENBANK-ID:MMU79776|acc:U79776 Mus musculus ajuba (Ajuba) mRNA, complete cds—Mus musculus, 2984 bp). The sequences are identical at 1571 of 1891 nucleoitdes (83%), and are positive at 1571 of 1891 (83%) nucleotides. The results of Nblast alignments are shown below, where “Query” is the disclosed PROTI sequence and “Sbjct” is the mouse Ajuba nucleotide sequence.
4Query:12GATAGACCTGCGGACTGGACAGCCGCGGCCAGAGACCCTGCTAGCCCCGCTCAGCCCCAG 71GA AGACCTGCGGA  GGACAGCCGC  CC   G CCCTGC AGCC CGC C G CCCSbjct:35GACAGACCTGCGGA--GGACAGCCGCTACCGTGGGCCCTGCCAGCCGCGCGCGGACCCTC 92Query:72ATGCGCGGGGGGACGCAGCCCCTCCCGCTGGGGGATG-CTG-TGGGATTCCTGGCGCAGG 129 T CGC GGGG A GC  C     C GC GG G   G C   TGGG   C T  CGC GSbjct:93-T-CGCTGGGGCATGCTTCGGGATC-GCGGGCGCCCGGCACCTGGGCCGC-T--CGCTGA 146Query:130GCATCCAGGCCGCCCGCTAAGCCCCTGTG-CCTC-CCCTGTGCCCCTGGGGAACCAGAGT 187GC  CC  G     C    A CCC  G   CC   CCC G G   CT G G  CC G  TSbjct:147GCCCCCGTGAGTGACTTGCATCCCAGGAATCCGAGCCCCGGGGAACTCGAGC-CCGGCCT 205Query:188CCGGCTG---CAGGGAAAGAGAACCGGCCGCCGAGACGCC-GCAGGGTGCCAGG-C-G-G 240C GGC G   CA  G A G G  CCGG CG  GAG  G C GCAGG   CCAG  C G GSbjct:206CTGGCCGAAACACTGCA-GCGTGCCGGGCGAGGAGGGGGCTGCAGGCC-CCAGATCCGTG 263Query:241GG-AGGGGGCGAG-AGGCCCCAG-GCCCGAGGGCATGGAGCGGTTAGGAGAGAAAGCCAG 297GG A GG  CG   AGG    A  GCC G  G C TG    G   AG  GAGA    C GSbjct:264GGCATGGAACGGTTAGGGGAGAAAGCCAGTCGTC-TGCTA-GAAAAGTTGAGACTCTCGG 321Query:298TCGCCTGCTG-GAGAAGTTCGGCCGCAGAAAGGGTGAATCTAGCCGGTCTGGGTCTGACG 356 C CC GC G G  AAGTTCGGCCGCAGAAAGGG GAA C AGCCG TCTGGGTCTGA GSbjct:322ACTCCGGCAGCGCCAAGTTCGGCCGCAGAAAGGGCGAAGCGAGCCGATCTGGGTCTGATG 381Query:357GGACCCCCGGGCCGGGCAAGGGGCGCCTAAGTGGGTTGGGGGGACCTAGGAAGTCAGGGC 416GGACCCCCGG  C GGCAAGGG CGC TAAG GGGTTGGGGGGACCTAGGAAGTCAGG CSbjct:382GGACCCCCGGAGCAGGCAAGGGACGCTTAAGCGGGTTGGGGGGACCTAGGAAGTCAGGAC 441Query:417CCCGAGGAGCTACTGGGGGACCTGGGGATGAGCCGTTGGAGCCGGCCCGGGAGCAAGGTT 476 CCG GGAGC A TGG GG CCTGG GATGA CC TTGGA CCGGCC GGGAGCAAGGSbjct:442ACCGTGGAGCGAATGGTGGGCCTGGAGATGAACCTTTGGAACCGGCCAGGGAGCAAGGGC 501Query:477CCCTGGACGCTGAGCGAAAT-CAGCGCGGCTCCTTTGAGGCGCCGCGCTACGAAGGCTCT 535CCCTGGACGC GAGCG AA  CA CGCGGCTCCTTTGA GCGC GCGCT CGAAGG TCSbjct:502CCCTGGACGCCGAGCGGAACGCA-CGCGGCTCCTTTGAAGCGCAGCGCTTCGAAGGGTCC 560Query:536TTTCCCGCGGGGCCGCCGCCCACCCGGGCCTTGCCTCTACCTCAGTCGTTGCCCCCCGAT 595TTTCCCG GGGGCCGCCGCCCAC CG GCC TGCCTCT CCTC GTCGT GCC CC GATSbjct:561TTTCCCGGGGGGCCGCCGCCCACTCGAGCCCTGCCTCTGCCTCTGTCGTCGCCTCCTGAT 620Query:596TTTCGGCTGGAGCCCACGGCCCCGGCCCTCAGCCCCCGCTCTAGCTTCGCCAGTAGCTCG 655TTTCGGCTGGAG CCACGGC CC GCCCT AGCCC CGCTC AGCTTCGCCAGTAGCTCGSbjct:621TTTCGGCTGGAGACCACGGCTCCAGCCCTTAGCCCTCGCTCCAGCTTCGCCAGTAGCTCG 680Query:656GCCAGCGACGCGAGCAAGCCGTCCAGCCCCCGGGGCAGCCTGCTGCTGGACGGGGCGGGG 715GCCAGCGA GCGAGCAAGCC TC AGCCCCCGGGGCAGCCTGCTGCTGGACGG GCGGGGSbjct:681GCCAGCGATGCGAGCAAGCCATCTAGCCCCCGGGGCAGCCTGCTGCTGGACGGAGCGGGG 740Query:716GCTGGCGGAGCTGGAGGTAGCCGGCCCTGCAGCAATCGCACCAGCGGCATCAGCATGGGC 775GC  GCGGAGC GGAGGTAGCCGGCC TGCAGCAATCG ACCAGCGGCATCAGCATGGGCSbjct:741GCCAGCGGAGCCGGAGGTAGCCGGCCGTGCAGCAATCGGACCAGCGGCATCAGCATGGGC 800Query:776TACGACCAGCGCCACGGGAGCCCCTTGCCAGCGGGGCCGTGCCTGTTTGGCCCACCCCTG 835TACGACCAGCGCCACGGGAGCCC  TGCC GCGGGGCCGTGCCT TT GGCC  CC CTGSbjct:801TACGACCAGCGCCACGGGAGCCCGCTGCCCGCGGGGCCGTGCCTATTCGGCCTCCCGCTG 860Query:836GCCGGAGCACCGGCAGGCTATTCTCCCGGAGGGGTCCCGTCCGCCTACCCGGAGCTCCAC 895 CC   GC CC GC GGCTA  C CC GGAGGGG  CCGTCCGCCTACCCGGAGCTCCACSbjct:861ACCACTGCTCCTGCGGGCTA--C-CCAGGAGGGGCTCCGTCCGCCTACCCGGAGCTCCAC 917Query:896GCCGCCCTGGACCGATTGTACGCTCAGCGGCCCGCGGGGTTCGGCTGCCAGGAAAGCCGC 955GC GCCCTGGACCGA T T  GCTCA CGG CCG GGG TTCGGCTGCCAGGA AGCCGSbjct:918GCTGCCCTGGACCGACTATGTGCTCATCGGTCCGTGGGATTCGGCTGCCAGGAGAGCCGT 977Query:956CACTCGTATCCCCCGGCCCTGGGCAGCCCTGGAGCTCTAGCCGGGGCCGGAGTGGGAGCG 1015CACTCGTA CCCCCGGCCCTGGGCAGCCC GGAGCTCTA CCGG GCCG  GTGGGA CSbjct:978CACTCGTACCCCCCGGCCCTGGGCAGCCCCGGAGCTCTAACCGGAGCCGTGGTGGGAACA 1037Query:1016GCGGGGCCCTTGGAGAGACGGGGGGCGCAACCCGGACGACACTCTGTGACCGGCTACGGG 1075GCGGG CC TTGGAGAGACG GGGGCGCAACCCGGACGACACTC GT AC GGCTACGGGSbjct:1038GCGGGTCCTTTGGAGAGACGTGGGGCGCAACCCGGACGACACTCGGTTACAGGCTACGGG 1097Query:1076GACTGCGCCGTGGGCGCCCGGTACCAGGACGAGCTAACAGCTTTGCTTCGCCTGACGGTG 1135GACTGCGCCG GGG GCCCG TACCAGGACGAGCTAACAGC TTGCT CG  TGAC GTGSbjct:1098GACTGCGCCGCGGGGGCCCGTTACCAGGACGAGCTAACAGCATTGCTGCGTTTGACCGTG 1157Query:1136GGCACCGGTGGGCGAGAAGCCGGAGCCCGCGGAGAACCCTCGGGGATTGAGCCGTCGGGT 1195G  ACCGGTGGGCGAGAAGCCGG GC CGCGG GAACCCTCGGGGATTGAGCCGTCGGGTSbjct:1158GCTACCGGTGGGCGAGAAGCCGGTGCTCGCGGGGAACCCTCGGGGATTGAGCCGTCGGGT 1217Query:1196CTGGAGGAGCCACCAGGTCCTTTCGTTCCGGAGGCCGCCCGGGCCCGGATGCGGGAGCCA 1255CTGGAGGAG C CC GGTCC TTCGTTCC GAGGCC CCCG  C CGGAT CGGGAGCCASbjct:1218CTGGAGGAGTCTCCTGGTCCCTTCGTTCCAGAGGCCTCCCGATCACGGATACGGGAGCCA 1277Query:1256GAGGCCAGGGAGGACTACTTCGGCACCTGTATCAAGTGCAACAAAGGCATCTATGGGCAG 1315GAGGCCAG GA GA TACTT GGCACCTGTATCAAGTGCAACAAAGG ATCTATGGGCAGSbjct:1278GAGGCCAGAGAAGATTACTTTGGCACCTGTATCAAGTGCAACAAAGGTATCTATGGGCAG 1337Query:1316AGCAATGCCTGCCAGGCCCTGGACAGCCTCTACCACACCCAGTGCTTTGTTTGCTGCTCT 1375AGCAATGCCTGCCAGGCCCTGGACAGCCTCTACCACACCCAGTGCTTTGT TGCTGCTCSbjct:1338AGCAATGCCTGCCAGGCCCTGGACAGCCTCTACCACACCCAGTGCTTTGTCTGCTGCTCC 1397Query:1376TGTGGGCGAACTTTGCGTTGCAAGGCTTTCTACAGTGTCAATGGCTCTGTGTACTGTGAG 1435TGTGG CGAACTTTGCG TGCAAGGCTTTCTACAG GTCAATGGCTCTGT TACTGTGAGSbjct:1398TGTGGACGAACTTTGCGGTGCAAGGCTTTCTACAGCGTCAATGGCTCTGTCTACTGTGAG 1457Query:1436GAAGATTATCTGTTTTCAGGGTTTCAGGAGGCAGCTGAGAAATGCTGTGTCTGTGGTCAC 1495GAAGA TATCTGTTTTCAGGGTTTCAGGAGGCAGCTGAGAA TGCTGTGTCTGTGG CACSbjct:1458GAAGACTATCTGTTTTCAGGGTTTCAGGAGGCAGCTGAGAAGTGCTGTGTCTGTGGCCAC 1517Query:1496TTGATTTTGGAGAAGATCCTACAAGCAATGGGGAAGTCCTATCATCCAGGCTGTTTCCGA 1555TTGATT T GAGAAGATCCT CA GC ATGGGGAAGTCCTATCA CC GGCTG TTCCGASbjct:1518TTGATTCTAGAGAAGATCCTCCAGGCCATGGGGAAGTCCTATCACCCGGGCTGCTTCCGA 1577Query:1556TGCATTGTTTGCAACAAGTGCCTGGATGGCATCCCCTTCACAGTGGACTTCTCCAACCAA 1615TGCAT GT TG AACAAGTGCCTGGA GGC TCCCCTTCAC GTGGACTTCTCCAACCASbjct:1578TGCATCGTATGTAACAAGTGCCTGGACGGCGTCCCCTTCACTGTGGACTTCTCCAACCAG 1637Query:1616GTATACTGTGTCACCGACTACCACAAAAATTATGCTCCTAAGTGTGCAGCCTGTGGCCAA 1675GT TACTGTGT ACCGACTACCACAAAAATTA GC CC AA TGTGCAGCCTG GG CAASbjct:1638GTGTACTGTGTTACCGACTACCACAAAAATTACGCCCCGAAATGTGCAGCCTGCGGACAA 1697Query:1676CCCATCCTCCCCTCTGAGGGCTGTGAGGACATCGTGAGGGTGATATCCATGGACCGGGAT 1735CCCATCCTCCCCTC GAGGGCTGTGAGGACAT GTGAGGGTGATATCCATGGACCG GATSbjct:1698CCCATCCTCCCCTCAGAGGGCTGTGAGGACATTGTGAGGGTGATATCCATGGACCGTGAT 1757Query:1736TATCACTTTGAGTGCTACCACTGTGAGGACTGCCGGATGCAGCTGAGTGATGAGGAAGGC 1795TATCACTTTGAGTGCTACCACTGTGAGGACTGCCG ATGCAGCTGAGTGA GAGGAAGGSbjct:1758TATCACTTTGAGTGCTACCACTGTGAGGACTGCCGCATGCAGCTGAGTGACGAGGAAGGT 1817Query:1796TGCTGCTGTTTCCCTCTGGATGGGCACTTGCTCTGCCATGGTTGCCACATGCAGCGGCTC 1853TGCTGCTGTTTCCCTCT GATGG CA TTGCTCTGCCA GG TG CACAT CAGCG CTCSbjct:1818TGCTGCTGTTTCCCTCTTGATGGACATTTGCTCTGCCACGGCTGTCACATACAGCGCCTC 1877Query:1856AATGCCCGACAACCCCCTGCCAACTATATCTGAGCTGCAATCACTGC 1902 (SEQ ID NO:9)A TGCCCG CA CCC CT CCAACTATATCTGA  T C  T  CTGC (SEQ ID NO:10)Sbjct:1878AGTGCCCGGCAGCCCTCTACCAACTATATCTGATAT-CTTTGTCTGC 1923 (SEQ ID NO:11)


[0043] The disclosed PROT1 polypeptide sequence is also homologous to the moue Ajuba polypeptide sequence. This similarity is shown in the alignement below, where the “Query” polypeptide is the disclosed PROTI polypeptide sequence and the “Sbjct” is the mouse Ajuba Mus Musculus polypeptide sequence. Overall, the disclosed PROT1 polypeptide sequence is identical at 502 of 547 (91%)amino acid residues and is positive at 510 of 547 (93%) residues.
5Query:272MERLGEKASRLLEK----------FGRRKGESSRSGSDGTPGPGKGRLSGLGGPRKSGPR 421MERLGEKASRLLEK          FGRRKGE+SRSGSDGTPG GKGRLSGLGGPRKSG RSbjct:1MERLGEKASRLLEKLRLSDSGSAKFGRRKGEASRSGSDGTPGAGKGRLSGLGGPRKSGHR 60Query:422GATGGPGDEPLEPAREQGSLDAERNQRGSFEAPRYEGSFPAGPPPTRALPLPQSLPPDFR 601GA GGPGDEPLEPAREQG LDAERN RGSFEA R+EGSFP GPPPTRALPLP S PPDFRSbjct:61GANGGPGDEPLEPAREQGPLDAERNARGSFEAQRFEGSFPGGPPPTRALPLPLSSPPDFR 120Query:602LEPTAPALSPRSSFASSSASDASKPSSPRGSLLLDGAGAGGAGGSRPCSNRTSGISMGYD 781LE TAPALSPRSSFASSSASDASKPSSPRGSLLLDGAGA GAGGSRPCSNRTSGISMGYDSbjct:121LETTAPALSPRSSFASSSASDASKPSSPRGSLLLDGAGASGAGGSRPCSNRTSGISMGYD 180Query:782QRHGSPLPAGPCLFGPPLAGAPAGYSPGGVPSAYPELHAALDRLYAQRPAGFGCQESRHS 961QRHGSPLPAGPCLFG PL  APAGY PGG PSAYPELHAALDRL A R  GFGCQESRHSSbjct:181QRHGSPLPAGPCLFGLPLTTAPAGY-PGGAPSAYPELHAALDRLCAHRSVGFGCQESRHS 239Query:962YPPALGSPGALAGAGVGAAGPLERRGAQPGRHSVTGYGDCAVGARYQDELTALLRLTVGT 1141YPPALGSPGAL GA VG AGPLERRGAQPGRHSVTGYGDCA GARYQDELTALLRLTV TSbjct:240YPPALGSPGALTGAVVGTAGPLERRGAQPGRHSVTGYGDCAAGARYQDELTALLRLTVAT 299Query:1142GGREAGARGEPSGIEPSGLEEPPGPFVPEAARARMREPEAREDYFGTCIKCNKGIYGQSN 1321GGREAGARGEPSGIEPSGLEE PGPFVPEA+R+R+REPEAREDYFGTCIKCNKGIYGQSNSbjct:300GGREAGARGEPSGIEPSGLEESPGPFVPEASRSRIREPEAREDYFGTCIKCNKGIYGQSN 359Query:1322ACQALDSLYHTQCFVCCSCGRTLRCKAFYSVNGSVYCEEDYLFSGFQEAAEKCCVCGHLI 1501ACQALDSLYHTQCFVCCSCGRTLRCKAFYSVNGSVYCEEDYLFSGFQEAAEKCCVCGHLISbjct:360ACQALDSLYHTQCFVCCSCGRTLRCKAFYSVNGSVYCEEDYLFSGFQEAAEKCCVCGHLI 419Query:1502LEKILQAMGKSYHPGCFRCIVCNKCLDGIPFTVDFSNQVYCVTDYHKNYAPKCAACGQPI 1681LEKILQAMGKSYHPGCFRCIVCNKCLDG+PFTVDFSNQVYCVTDYHKNYAPKCAACGQPISbjct:420LEKILQAMGKSYHPGCFRCIVCNKCLDGVPFTVDFSNQVYCVTDYHKNYAPKCAACGQPI 479Query:1682LPSEGCEDIVRVISMDRDYHFECYHCEDCRMQLSDEEGCCCFPLDGHLLCHGCHMQRLNA 1861LPSEGCEDIVRVISMDRDYHFECYHCEDCRMQLSDEEGCCCFPLDGHLLCHGCH+QRL+ASbjct:480LPSEGCEDIVRVISMDRDYHFECYHCEDCRMQLSDEEGCCCFPLDGHLLCHGCHIQRLSA 539Query:1862RQPPANYI 1885 (SEQ ID NO:12)RQP  NYI (SEQ ID NO:13)Sbjct:540RQPSTNYI 547 (SEQ ID NO:14)


[0044] The similarity between the disclosed PROT1 polypeptide sequence and the murine Ajuba protein is further shown in the ClutalW alignment below. Black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); greyed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted amino acid residues can potentially be mutated to a much broader extent without altering structure or function. The murine sequence is taken from AJUBA—MUS MUSCULUS: P97472_Best_BlastX_Mouse (SEQ ID NO:15).
612345678910


[0045] PROT2. Clone AC016900_A, a Novel Cytokeratin-8-like Protein


[0046] A PROT2 nucleic acid and polypeptide according to the invention includes the nucleic acid and encoded polypeptide sequence of clone AC016900_A.


[0047] A polynucleotide of the present invention has been identified as clone ACO 16900_A. AC016900_A is a full-length clone of 1474 nucleotides, including the entire coding sequence of a protein from nucleotides 10 to 1443 (also referred to herein as “AC016900_A protein”). The clone was originally obtained from genomic DNA.


[0048] The nucleotide sequence of AC016900_A is reported in SEQ ID NO:3. The disclosed nucleic acid encodes a polypeptide having the amino acid sequenc reported in SEQ ID NO:4. The encoded polypeptide is 477 amino acids in length and has a predicted molecular weight of 52694.5. Psort analysis predicts the protein localizes to the mitochondrial matrix space (certainty of 0.4996), the microbody/peroxisome (certainty of 0.3000), mitochondrial inner membrane (certainty of 0.2182), or mitochondrial intermembrane space (0.2182). A hydropphocity plot of the encoded polypeptide is presented in FIG. 2.


[0049] The disclosed PROT2 nucleic acid sequence has 959 of 1024 bases (93%) identical to a Homo Sapiens species Cytokeratin 8 mRNA (GENBANK-ID: X74929). The amino acid sequence of the disclosed PROT2 protein has 427 of 482 amino acid residues (88%) identical to, and 444 of 482 residues (92%) positive with, the 482 amino acid residue protein from Homo Sapiens (ptnr:SWISSPROT-ACC:P05787) (Table 4B).


[0050] A multiple sequence alignment is given in Table 5, with the protein of the invention being shown on line 5, in a ClustalW analysis comparing the protein of the invention with related protein sequences.


[0051] Based on PSORT and SIGNALP analysis this protein does not appear to have a signal sequence.


[0052] A PROT2 nucleic acid of the invention (AC016900_A) has the following sequence:
7GCCTCTACCATGTCCATCAGGGTGACCCAGAAGTCCTACAAGGTGTCCACCTCTGGCCCCCGGGCCTTCAGCAGCCGCTCCTACACGAGTGGGCCCGGTTCCCGCATCAGCTCCTCGAGCTTCTCCCGAGTGGGCAGCAGCAGCTTCCGAGGTGGCCTGGGTGGAGGCTATGGTGGGACCAGCGGTGTGGGAGGCATCACCACCGTCCCGGTCAACCAAAGCCTGCTGAGCCCCCTTAACCTGGAGGTGGACCCCAATATCCAGGCCATGCGCACCCAGGAGGAGCAGATCAAGACCCTCAATTTGCCTCCTTCGTCGACAAGGTAAGGTCCCTAGAGCAGCAGAACAAGATGCTGGAGACCAAGTGGAGCCTCCTGCAGCAGCAGCAGACGGCTCGGAGCAACATGGACAACATGTTTGAGAGCTACGTCAACAACCTTAGGTGGCAGCTGGAGACTCTGGGCCAGGAGAAGCTGAAGCTGGAGGCGGAGCTTGGCAACATGCAGCCGCTGGTGGAGGACTTCAAGAACAAGTATGAGGATGAGATCAATAAGCGTACAGAGATGGAGAATGAATTTGTCCTCATCAAGAAGGATGCAGATGAAGCTTCCATGAACAAGGTAGAGCTAGGGTCTCGCCTGGAAGGGCTGACTGACGAGATCAACTTCCTCATGCAGCTATATGAAGAGGAGATCCGGGAGCTGCAGTCCCAGATCTCAGACACGTCTGTGGTGCTGTCCATGGACAACAGCTGCTCCCTGGACATGGACAGCATCATTGCTGAGGTCAAGGCACAGTACGAGGAGATCGCCAACTGCAGCCGGGCTGAGGCCGAGAGCATGTACCAGATCAAGTATGAGGAGCTGCAGACGCTGGCTGGGAAGCACGGGGATGACCGGCGGCGCACAAAGACTGAGATCTCCGAGATATACGGGAACATCAGCCAGCTCCAGGCTCATGTTGACCAGAGGGCTTCCCTGGAGGCCGCCATCACAGATGCCGAGCTGCACGGGGATTTGGTGGTTAAGGATGCCAACGCCAAGCTGTCCGAGCTGGAGGCCGCCCTGCAGCGGGCCAAGCAGGACATGGCGTGGCAGCTGCGTGAATACCAGGAGCTGATGAACGTCAAGCTGGCCCTGGACATAGAGATCGCCACCTACAGGAAGCTGCTGGAGGGCAAGGAGAGCCGGCTGGAGTCTGGGATGCAGAACAGGAGTATCCATACGAAGACCACCAGTGGCGGTGGAGGTGTGCTGAGCTCAGCCTATGGGGGTCTCACAAGCCCCGGTCTCAGCTATGGCCTGGGCTCTAGCTTTGGCTCTGGCGCGGGGTCCCCCTCCTTCAGCCGCACCAGTTCCGCCAGGGCCGTGGTCGTGAAGATGATTGAGACCTGCGATGGGAAGCTGGTGTCCGAGTCCTCTGACATCCAGCCAAAGTGAACAGCTGCGGCAGCCCCTCCCAGCCTACCCC (SEQ ID NO:3)


[0053] A PROT2 polypeptide of the invention (AC016900_A) includes the following sequence:
8MSIRVTQKSYKVSTSGPRAFSSRSYTSGPGSRISSSSFSRVGSSSFRGGLGGGYGGTSGVGGITTVPVNQ(SEQ ID NO:4)SLLSPLNLEVDPNIQAMRTQEEQIKTLNKFASFVDKVRSLEQQNKMLETKWSLLQQQQTARSNMDNMFESYVNNLRWQLETLGQEKLKLEAELGNMQPLVEDFKNKYEDEINKRTEMENEFVLIKKDADEASMNKVELGSRLEGLTDEINFLMQLYEEEIRELQSQISDTSVVLSMDNSCSLDMDSIIAEVKAQYEEIANCSRAEAESMYQIKYEELQTLAGKHGDDRRRTKTEISEIYGNISQLQAHVDQRASLEAAITDAELHGDLVVKDANAKLSELEAALQRAKQDMAWQLREYQELMNVKLALDIEIATYRKLLEGKESRLESGMQNRSIHTKTTSGGGGVLSSAYGGLTSPGLSYGLGSSFGSGAGSPSFSRTSSARAVVVKMIETCDGKLVSESSDIQPK


[0054] The similarity between the disclosed PROT2 nucleic acid and cytokeratin8 nucleic acids is shown in the following BLASTN alignment. The compared sequence corresponds to a human mRNA encoding cytokeratin8 (>gb:GENBANK-ID:HSKRT81acc:X74929 H.sapiens KRT8 mRNA for keratin 8—Homo sapiens, 1752 bp (RNA)). For the compared sequences, 959 of 1024 nucleotides are identical. In the alignment shown below, the disclosed PROT2 nucleic acid is shown as the “Query” nucleic acid and the huma cytokeratin8 sequence is listed as the “Sbct” nucleic acid.
9Query:145CCGCTCCTTCTAGGATCTCCGCCTGGTTCGGCCCGCCTGCCTCCACTCCNGCCTCTACCA 204C GCTCCTTCTAGGATCTCCGCCTGGTTCGGCCCGCCTGCCTCCACTCC GCCTC ACCASbjct:1CTGCTCCTTCTAGGATCTCCGCCTGGTTCGGCCCGCCTGCCTCCACTCCTGCCTCCACCA 60Query:205TGTCCATCAGGGTGACCCAGAAGTCCTACAAGGTGTCCACCTCTGGCCCCCGGGCNTTCA 264TGTCCATCAGGGTGACCCAGAAGTCCTACAAGGTGTCCACCTCTGGCCCCCGGGC TTCASbjct:61TGTCCATCAGGGTGACCCAGAAGTCCTACAAGGTGTCCACCTCTGGCCCCCGGGCCTTCA 120Query:265GCAGCCGCTCCTACACGAGTGGGCCCGGTTCCCGCATCAGCTCCTCGAGCTTCTCCCGAG 324GCAGCCGCTCCTACACGAGTGGGCCCGGTTCCCGCATCAGCTCCTCGAGCTTCTCCCGAG Sbjct:121GCAGCCGCTCCTACACGAGTGGGCCCGGTTCCCGCATCAGCTCCTCGAGCTTCTCCCGAG 180Query:325TGGGCAGCAGCAGCTTCCGAGGTGGCCTGGGTGGAGGCTATGGTGGGACCAGCGGTGTGG 384TGGGCAGCAGCA CTT CG GGTGGCCTGGG GG GGCTATGGTGGG CCAGCGG\\TGGSbjct:181TGGGCAGCAGCAACTTTCGCGGTGGCCTGGGCGGCGGCTATGGTGGGGCCAGCGGCATGG 240Query:385GAGGCATCACCACCGTCCCGGTCAACCAAAGCCTGCTGAGCCCCCTTAACCTGGAGGTGG 444GAGGCATCACC C GT\\CGGTCAACCA AGCCTGCTGAGCCCCCTT\\CCTGGAGGTGGSbjct:241GAGGCATCACCGCAGTTACGGTCAACCAGAGCCTGCTGAGCCCCCTTGTCCTGGAGGTGG 300Query:445ACCCCAATATCCAGGCCATGCGCACCCAGGAG--G-AGCAGATCAAGACCCTCAACAA-- 499ACCCCAA ATCCAGGCC TGCGCACCCAGGAG\\G AGCAGATCAAGACCCTCAACAASbjct:301ACCCCAACATCCAGGCCGTGCGCACCCAGGAGAAGGAGCAGATCAAGACCCTCAACAACA 360Query:500-GTTTGCCTCCTTCGTCGACAAGGTAAGGTCCCTAGAGCAGCAGAACAAGATGCTGGAGA 558\GTTTGCCTCCTTC T GACAAGGTA GGT CCT GAGCAGCAGAACAAGATGCTGGAGASbjct:361AGTTTGCCTCCTTCATAGACAAGGTACGGTTCCTGGAGCAGCAGAACAAGATGCTGGAGA 420Query:559CCAAGTGGAGCCTCCTGCAGCAGCAGCAGACGGCTCGGAGCAACATGGACAACATGTTTG 618CCAAGTGGAGCCTCCTGCAGCAGCAG AGACGGCTCG AGCAACATGGACAACATGTT GSbjct:421CCAAGTGGAGCCTCCTGCAGCAGCAGAAGACGGCTCGAAGCAACATGGACAACATGTTCG 480Query:619AGAGCTACGTCAACAACCTTAGGTGGCAGCTGGAGACTCTGGGCCAGGAGAAGCTGAAGC 678AGAGCTAC TCAACAACCTTAGG GGCAGCTGGAGACTCTGGGCCAGGAGAAGCTGAAGCSbjct:481AGAGCTACATCAACAACCTTAGGCGGCAGCTGGAGACTCTGGGCCAGGAGAAGCTGAAGC 540Query:679TGGAGGCGGAGCTTGGCAACATGCAGCCGCTGGTGGAGGACTTCAAGAACAAGTATGAGG 738TGGAGGCGGAGCTTGGCAACATGCAG\\GCTGGTGGAGGACTTCAAGAACAAGTATGAGGSbjct:541TGGAGGCGGAGCTTGGCAACATGCAGGGGCTGGTGGAGGACTTCAAGAACAAGTATGAGG 600Query:739ATGAGATCAATAAGCGTACAGAGATGGAGAATGAATTTGTCCTCATCAAGAAGGATGCAG 798ATGAGATCAATAAGCGTACAGAGATGGAGAA GAATTTGTCCTCATCAAGAAGGATG\\GSbjct:601ATGAGATCAATAAGCGTACAGAGATGGAGAACGAATTTGTCCTCATCAAGAAGGATGTGG 660Query:799ATGAAGCTTCCATGAACAAGGTAGAGCTAGGGTCTCGCCTGGAAGGGCTGACTGACGAGA 858ATGAAGCTT CATGAACAAGGTAGAGCT G GTCTCGCCTGGAAGGGCTGAC GACGAGASbjct:661ATGAAGCTTACATGAACAAGGTAGAGCTGGAGTCTCGCCTGGAAGGGCTGACCGACGAGA 720Query:859TCAACTTCCTCATGCAGCTATATGAAGAGGAGATCCGGGAGCTGCAGTCCCAGATCTCAG 918TCAACTTCCTCA GCAGCTATATGAAGAGGAGATCCGGGAGCTGCAGTCCCAGATCTC GSbjct:721TCAACTTCCTCAGGCAGCTATATGAAGAGGAGATCCGGGAGCTGCAGTCCCAGATCTCGG 780Query:919ACACGTCTGTGGTGCTGTCCATGGACAACAGCTGCTCCCTGGACATGGACAGCATCATTG 978ACAC TCTGTGGTGCTGTCCATGGACAACAGC GCTCCCTGGACATGGACAGCATCATTGSbjct:781ACACATCTGTGGTGCTGTCCATGGACAACAGCCGCTCCCTGGACATGGACAGCATCATTG 840Query:979CTGAGGTCAAGGCACAGTACGAGGAGATCGCCAACTGCAGCCGGGCTGAGGCCGAGAGCA 1038CTGAGGTCAAGGCACAGTACGAGGA AT GCCAAC GCAGCCGGGCTGAGGC GAGAGCASbjct:841CTGAGGTCAAGGCACAGTACGAGGATATTGCCAACCGCAGCCGGGCTGAGGCTGAGAGCA 900Query:1039TGTACCAGATCAAGTATGAGGAGCTGCAGACGCTGGCTGGGAAGCACGGGGATGACCGGC 1098TGTACCAGATCAAGTATGAGGAGCTGCAGA\\CTGGCTGCGAAGCACGGGGATGACC GCSbjct:901TGTACCAGATCAAGTATGAGGAGCTGCAGAGCCTGGCTGGGAAGCACGGGGATGACCTGC 960Query:1099GGCGCACAAAGACTGAGATCTCCGAGATATACGGGAACATCAGCCAGCTCCAGGCTCATG 1158GGCGCACAAAGACTGAGATCTC GAGAT\\AC GGAACATCAGCC GCTCCAGGCT ASbjct:961GGCGCACAAAGACTGAGATCTCTGAGATGAACCGGAACATCAGCCGGCTCCAGGCTGAGA 1020Query:1159TTGA 1162 (SEQ ID NO:16)TTGA (SEQ ID NO:17)Sbjct:1021TTGA 1024 (SEQ ID NO:18)


[0055] A BLASTX alignment between the disclosed PROT2 polypeptide and a human cytokeratin 8 polypeptide is shown below. The disclosed PROT2 polypeptide is identical at 427 of 482 amino acid residues (88%) and is positive at 442 of 482 amino acid residues (92%). In the comparison shown, the disclosed PROT2 polypeptide is indicated as the “Query” polypeptide ant the human cytokeratin8 polypeptide is indicated as the “Sbjct” polypeptide (SWISSPROT-ACC:P05787 keratin, Type II Cytoskeletal 8 (Cytokeratin 8) (Homo sapiens (Human), 482 aa).
10Query:207SIRVTQKSYKVSTSGPRAFSSRSYTSGPGSRISSSSFSRVGSSSFRGGLGGGYGGTSGVG 386SIRVTQKSYKVSTSGPRAFSSRSYTSGPGSRISSSSFSRVGSS+FRGGLGGGYGG SG+GSbjct:1SIRVTQKSYKVSTSGPRAFSSRSYTSGPGSRISSSSFSRVGSSNFRGGLGGGYGGASGMG 60Query:367GITTVPVNQSLLSPLNLEVDPNIQAMRTQE-EQIKTLN-KFASFVDKVRSLEQQNKMLET 560GIT V VNQSLLSPL LEVDPNIQA+RTQE EQIKTLN KFASF+DKVR LEQQNKMLETSbjct:61GITAVTVNQSLLSPLVLEVDPNIQAVRTQEKEQIKTLNNKFASFIDKVRFLEQQNKMLET 120Query:561KWSLLQQQQTARSNMDNMFESYVNNLRWQLETLGQEKLKLEAELGNMQPLVEDFKNKYED 740KWSLLQQQ+TARSNMDNMFESY+NNLR QLETLGQEKLKLEAELGNMQ LVEDFKNKYEDSbjct:121KWSLLQQQKTARSNMDNMFESYINNLRRQLETLGQEKLKLEAELGNMQGLVEDFKNKYED 180Query:741EINKRTEMENEFVLIKKDADEASMNKVELGSRLEGLTDEINFLMQLYEEEIRELQSQISD 920EINKRTEMENEFVLIKKD DEA MNKVEL SRLEGLTDEINFL QLYEEEIREIQSQISDSbjct:181EINKRTEMENEFVLIKKDVDEAYMNKVELESRLEGLTDEINFLRQLYEEEIRELQSQISD 240Query:921TSVVLSMDNSCSLDMDSIIAEVKAQYEEIANCSRAEAESMYQIKYEELQTLAGKHGDDRR 1100TSVVLSMDNS SLDMDSIIAEVKAQYE+IAN SRAEAESMYQIKYEELQ+LAGKHGDD RSbjct:241TSVVLSMDNSRSLDMDSIIAEVKAQYEDIANRSRAEAESMYQIKYEELQSLAGKHGDDLR 300Query:1101RTKTEISEIYGNISQLQAHVD----QRASLEAAITDAELHGDLVVKDANAKLSELEAALQ 1268RTKTEISE+  NIS+LQA ++    QRASLEAAI DAE G+L +KDANAKLSELEAALQSbjct:301RTKTEISEMNRNISRLQAEIEGLKGQRASLEAAIADAEQRGELAIKDANAKLSELEAALQ 360Query:1269RAKQDMAWQLREYQELMNVKLALDIEIATYRKLLEGKESRLESGMQNRSIHTKTTSGGGG 1448RAKQDMA QLREYQELMNVKLALDIEIATYRKLLEG+ESRLESGMQN SIHTKTT G\\GSbjct:361RAKQDMARQLREYQELMNVKLALDIEIATYRKLLEGEESRLESGMQNMSIHTKTTGGYAG 420Query:1449VLSSAYGGLTSPGLSYGLGSSFGSGAGSPSFSRTSSARAVVVKMIETCDGKLVSESSDIQ 1628LSSAYGGLTSPGLSY LGSSFGSGAGS SFSRTSS+RAVVVK IET DGKLVSESSD+Sbjct:421GLSSAYGGLTSPGLSYSLGSSFGSGAGSSSFSRTSSSRAVVVKKIETRDGKLVSESSDVL 480Query:1629PK 1634 (SEQ ID NO:19)PK (SEQ ID NO:20)Sbjct:481PK 482 (SEQ ID NO:21)


[0056] The relationship between the disclosed PROT2 polypeptide and previously described cytokeratin8 polypeptide is further illustrated in the following ClustalW alignment. Black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); greyed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted amino acid residues can potentially be mutated to a much broader extent without altering structure or function.


[0057] For the sequences compared:
11Sequence 1:SWISSPROT-ACC:P05787 K2C8_HUMAN_CK_8(SEQ ID NO:22)482 aaSequence 2:SPTREMBL-ACC:Q61463 Q61463_MOUSE_CK_Endo_A(SEQ ID NO:23)490 aaSequence 3:SWISSPROT-ACC:Q10758 K2C8_RAT_CK_Endo A(SEQ ID NO:24)482 aaSequence 4:SWISSPROT-ACC:P11679 K2C8_MOUSE_CK_Endo_A(SEQ ID NO:25)488aaSequence 5:PROT216900_A477 aa1112131415161718


[0058] 3. PROT3. Clone 2826468.0.39, a Novel Human Antigenic Protein


[0059] A PROT3 nucleic acid and polypeptide according to the invention includes the nucleic acid and encoded polypeptide sequence of clone 2826468.0.39.


[0060] A polynucleotide of the present invention has been identified as clone 2826468.0.39. 2826468.0.39 is a full-length clone of 1481 nucleotides, including the entire coding sequence of a protein from nucleotides 743 to 1180 (also referred to herein as “2826468.0.39 protein”).


[0061] The nucleotide sequence of 2826468.0.39 is reported in SEQ ID NO:5. The predicted amino acid sequence of the 2826468.0.39 protein is reported in SEQ ID NO:6. The encoded protein is 145 amino acids and has a predicted molecular weight of 16213.5. Psort analysis predcits that the encoded protein localizes to the microbody (peroxisome) (certainty of 0.64), cytoplasm (certainty of 0.45), lysosomal lumen (certainty of 0.1731), and/or mitochondrial matrix space (certainty of 0.100). A hydropathy plot of the disclosed PROT3 sequence is presented in FIG. 3.


[0062] The disclosed PROT32826468.0.39 nucleic acid and encoded polypeptide have the following sequences:
121GCCTGGGAAGCTGGGCAGAGGCGTGGCAGCAGGGGGCTGGGAAGGGAGGAGGTGCCCGATGCCAGGATTGAGGGTTTCTT81GGGTCACCTGTTTCCAAAAGCTCCCTATGACCCCAAGCTCCAACCAGGAGAGAAGGGCAGAGAGGCCACAGCCAGGCCAT161CTGGGGAGGGTCTGGCATGGAAGGGTTCTGGGAGGATGGAGTGGTCGGGCCCAATATGTGGCATCCTGTAGGCAGGTCCT241GGGTGCTGTGCCATGGATCTGGCCGGCCCTTCCCCGAAGTATGGTCTTGAGCTAGCCGATCCCCCAACTCTGGGCCACAG321CTTCTCTCTGTGACATGGCCTGGGGGCTGTGGCCCCTCATCCTGGCATGCCCCCAGGAGCTCTTAGTGAGAAGACCAAGG401CTTCCTACCTCCTGGCCCCTGCTGCCCTTGGGTGGGGCCCTCCACCACCCAGAAATGCAAACTGGGCAGCAGGGCGGGTC481TCTGGGGAGCAGGTGTGTTTCCTGAGTGCCCAGGGCTCGGTATTTACAGCAGCAGTGCTGTTGGCCTCTTCTCCTTGGCC561CGGCCCCCTCCCCTGTGTGCCTCCTCCCTGGCCTCCAAGAGGTCTGCAGGGAAGTGTGGGGGCACACAGAGCTCAGATCT641TGCTCAGTTCCCTCCTTGCCAGGCAGCCTTGGGCGGGTGCCAACCCCTTGCTGGGCTGGGGGTGATCATCTTGGCCCACC721TTGCCAGGCGGTTGAGGACCAGATGTGGACAGCACAGGCCCATTGGGGGCCAGGCTGGGACTGGATGTGGGCGGTCTTCC\\\\\\\\\\\\\\\\\\\\\\MetTrpThrAlaGlnAlaHisTrpGlyProGlyTrpAspTrpMetTrpAlaValPheLTCACCTCCCCCAGATTCTCTGAGCCCTGCGCACCACCCCGTTGGTCCAGTCTTGCCTCCTCAGCCAGCCTGGTCTCCCTTeuThrSerProArgPheSerGluProCysAlaProProArgTrpSerSerLeuAlaSerSerAlaSerLeuValSerLeu881GGTCCCCTCTGCCATGGAGGCCTGTGGGACCCTGACTGCAGCTTACCTCACAGCCACCCTCAGCGGCATCCCCACCCCTGGlyProLeuCysHisGlyGlyLeuTrpAspProAspCysSerLeuProHisSerHisProGlnArgHisProHisProCy961CTCTCAGCTCCCAACCACCGCCCGGGTTGTGCTTCTTCGTAAAGGGAGCCGGGCTAGGGGGTCCTTGGTGTCAACCAGACsSerGlnLeuProThrThrAlaArgValValLeuLeuArgLysGlySerArgAlaArgGlySerLeuValSerThrArgP1041CCAGGCACTCACAGCTTGTCCTGAGTCCCTGTCCCTCCCTCCCCACCCACATGTGCCAGGCCCTGTCCAGGGACCCCAAGroArgHisSerGlnLeuValLeuSerProCysProSerLeuProThrHisMetCysGlnAlaLeuSerArgAspProLys1121TTCCCAAGACCCTGCTTCTCAATCTGGCAAATGCCAGACCCATCCAGGTGCCCACACTGATGGGCAGCTAAGCAGGCAGCPheProArgProCysPheSerIleTrpGlnMetProAspProSerArgCysProHis (SEQ ID NO:6)1201GAGGAGTCAGTGCTGCCATCCAAGGAGAAGGTGCCAGGCAGAAGTGTGCTTCTGGCTTCTGGTACTCAGGGTGCCTGGG1281GTGTCCCCCGCACAGAGTGAAGCCGTTCAAGTGAGGGATCAGGCAGGAAGCCATAAGGCTGATGGGGAAGGAGCTGGTGG1361GAGTAACGACCAGGCCACCCGGGCTCAGAGACCATTACCCGCATTACCCCTGTGCCGCGGCTCTTTCTAGTGTCCTCCTC1441AGGTGATGCCCACCCTCCCCTGGGCCCATCCCTGGGTCACA (SEQ ID NO:5)


[0063] 4. PROT4. Clone AC015862_A, a Novel Endothelin-like Protein


[0064] A PROT4 nucleic acid and polypeptide according to the invention includes the nucleic acid and encoded polypeptide sequence of clone AC015862_A.


[0065] A polynucleotide of the present invention has been identified as clone AC015862_A. AC015862_A is a full-length clone of 731 nucleotides, including the entire coding sequence of a secreted protein from nucleotides 71 to 592 (also referred to herein as “AC015862_A protein”). The nucleotide sequence of AC015862_A is reported in SEQ ID NO:7. The predicted amino acid sequence of the encoded AC015862_A protein is reported in SEQ ID NO:8. The encoded PROT4 polypeptide is 173 amino acids and has a predicted molecular weight of 19183.9. Psort analysis predicts that the disclosed PROT4 polypeptide localizes outside the cell (certainty of 0.738), endoplasmic reticulum membrane (certainty of 0.100), endoplasmic reticulum lumen (certainty of 0.100), or lysosome (certainty of 0.100). A hydropathy plot of the disclosed PROT4 polypeptidesequence is presented in FIG. 4.


[0066] The disclsoed PROT4 nucleic acid sequence has 375 of 425 bases (88%) identical to a human endothelin-2 mRNA (GENBANK-ID: M65199). The full amino acid sequence of the protein of the invention was found to have 134 of 178 amino acid residues (75%) identical to, and 143 of 178 residues (80%) positive with, the 178 amino acid residue protein from human (ptnr:SWISSPORT-ACC:P20800).


[0067] A multiple sequence alignment is given below, with the protein of the invention is shown on line 1, in a ClustalW analysis comparing the protein of the invention with related protein sequences.


[0068] The AC015862_A nucleic acid has the following sequence:
13CAGCTTAATAGCAGGACGCTGGCAACAGGCGCTCCCTGCTCCAGTCCAGCCTGCGCGCTCCACCG(SEQ ID NO:7)CCGCTATGGTCTCCGTGCCTAGCACCTGGGTGCTCGTTGCGCTAGCCTTGCTCGTGGGCCTGCATGAAGGGAAGGGCCAGGTTGCTGCCACCTTGGAGCAGCCAGCGTCTTATCTCCATGCCAAGGGCACCCACCTTCGGTTTCGCCGTTGCTCCTGCAGCTCCTGGCTCGCCAAGGAGTGCGTCTACTTCTGCCACTTGGACATCATCTGGGTGAACACTCCTGAGGAAGACACTGGGGTGCCGAGAGCCCTGGGTCTCCCTCCAAGGCGCTTCAGTCTGCAGTGGGCAAGCTGCCAGGACCTTCCCTGCCTATCCTGCAGTAGGAGAGTGCAGATTGAAGCCGGGCCAGTCCCCAGCGGGAAGTCCCCTGCAAACGTGTTCCAGACTGGCAAGACAGGGGCCACTACAGGAGAGCTTCTCCAAAGGCTGAGGGACATTTCCACAGTCAAGAGCCTCTTTGCCAAGCGACAACAGGAGGCCAATGCGGAGCTTCGGTCCACACATTCCAGGTGGAAGGAGAGATAGTGTCGTGAGCTGGAGGGACATTGGGGAGGAAGCCCGCGGGGAGAGAGGAGGAGAAAACTGGGCCAGGGTTGTGGACTCTCCTGCTGCTTTCTGGACGGGGGGCCTTGTCCAGACAGCTGGAACCATTTTCCCAGGATTG


[0069] The AC015862_A polypeptide has the following sequence:
14MVSVPSTWVLVALALLVGLHEGKGQVAATLEQPASYLHAKGTHLRFRRCSCSSWLAKECVYFCHL(SEQ ID NO:8)DIIWVNTPEEDTGVPRALGLPPRRFSLQWASCQDLPCLSCSRRVQIEAGPVPSGKSPANVFQTGKTGATTGELLQRLRDISTVKSLFAKRQQEANAELRSTHSRWKER


[0070] The disclosed PROT4 nucleic acid sequence is homologous to previously described human endothelin 2 nucleic adis. This similarity is shown in the BLASTN alignment shown below. The sequence denoted as the “Sbct” sequence is the disclosed PROT4 nucleic acid. The “Sbct” sequence is the human endothtelin ET2 nucleic acid seuqncde (gb:GENBANK-ID:HUMET2A|acc:M65199 Human endothelin 2 (ET2) mRNA, complete cds—Homo sapiens, 1240 bp (RNA)). The disclosed PROT4 nucleic acid is identical at 375 of 425 (88%) nucleic acids.
15Query:307GCCGAGAGCCCTGGGTC--TCCCT-CCAAGGCGCT-TCAGT-CTGCAGTGGGCAAGCTGC361GCC AGA  CC G G C  TCCCT CCAAGGCGCT TCAGT CT CAGTG  CA G   CSbjct:322GCCAAGACGCCGGCGCCGCTCCCTGCCAAGGCGCTGTCAGTGCTCCAGTGC-CAGGGACC380Query:362CAGGACCT-TCCCTGCCTATCCTGCAGTAGGAGAGTGCAG-ATTGAAGCCGGGCCAGTCC419C G  CCT T CC  CCT TC TGC  T G AG G  C G A TGAAGCCGGG CAGTCCSbjct:381CCG--CCTGTGCCA-CCT-TC-TGCCTTCGAAG-GCCCTGGACTGAAGCCGGGGCAGTCC434Query:420CCAGCGGGAAGTCCCCTGCAAACGTGTTCCAGACTGGCAAGACAGGGGCCACTACAGGAG479C AGC GGAAGTCCCCTGCA ACGTGTTCCAGACTGGCAAGACAGGGGCCACTACAGGAGSbjct:435CAAGCCGGAAGTCCCCTGCAGACGTGTTCCAGACTGGCAAGACAGGGGCCACTACAGGAG494Query:480AGCTTCTCCAAAGGCTGAGGGACATTTCCACAGTCAAGAGCCTCTTTGCCAAGCGACAAC539AGCTTCTCCAAAGGCTGAGGGACATTTCCACAGTCAAGAGCCTCTTTGCCAAGCGACAACSbjct:495AGCTTCTCCAAAGGCTGAGGGACATTTCCACAGTCAAGAGCCTCTTTGCCAAGCGACAAC554Query:540AGGAGGCCAATGCGG-AGCTTCGGTCCACACATTCCAGGTGGAAGGAGAGATAGTGTCGT598AGGAGGCCA TGCGG AGC TCGGTCCACACATTCCAGGTGGA G AGAGATAGTGTCGTSbjct:555AGGAGGCCA-TGCGGGAGCCTCGGTCCACACATTCCAGGTGGAGGAAGAGATAGTGTCGT613Query:599GAGCTGGAGGGACATTGGGGAGGAAGCCCGCGGGGAGAGAGGAGGAGAAAACTGGGCCAG658GAGCTGGAGG ACATTGGG AGGAAGCCCGCGGGGAGAGAGGAGGAGA AA TGG CCAGSbjct:614GAGCTGGAGGAACATTGGGAAGGAAGCCCGCGGGGAGAGAGGAGGAGAGAAGTGG-CCAG672Query:659GG-TTGTGGACTCTCCTGC-TGCTTTCTGGACGGGGGGCCTTG-TCC-AGACAGCTGGA714GG TTGTGGACTCTC TGC TGCTT CTGGAC GGGG CCTTG TCC AGACAGCTGGASbjca:673GGCTTGTGGACTCTC-TGCCTGCTTCCTGGACCGGGG-CCTTGGTCCCAGACAGCTGGAC730Query:715CCATTTTCCCAGGATTG (SEQ ID NO:26)731CCATTT CC AGGATTG (SEQ ID NO:27)Sbjct:731CCATTTGCC-AGGATTG (SEQ ID NO:28)746


[0071] The similarity between the disclosed PROT4 polypeptide sequence and the human endothelin-2 precursor is provided in the following BLASTX alignment. The disclosed PROT4 polypeptide is presented as the “Query” polypetpide, and the human endothelin-2 precursor is presented as the “Sbjct” sequence (ptnr:SWISSPROT-ACC:P20800 ENDOTHELIN-2 PRECURSOR (ET-2). The disclosed PROT4 polypetpide is identical at 134 of 178 amino acids (75%) and is positive at 143 of 178 (80%) amino acid residues.
16Query:71MVSVPSTWVLVALALLVGLHEGKGQVAATLEQPASYLHAKGTHLRFRRCSCSSWLAKECV250MVSVP+TW  VALALLV LHEGKGQ AATLEQPAS  HA+GTHLR RRCSCSSWL KECVSbjct:1MVSVPTTWCSVALALLVALHEGKGQAAATLEQPASSSHAQGTHLRLRRCSCSSWLDKECV60Query:251YFCHLDIIWVNTPEEDTGVPRALGLPPRRF--SL----QWASCQDLPCLS-CSRRVQIEA409YFCHLDIIWVNTPE+    P  LG PPRR   SL    Q +S +D  C + C RR   EASbjct:61YFCHLDIIWVNTPEQTA--PYGLGNPPRRRRRSLPRRCQCSSARDPACATFCLRRPWTEA118Query:410GPVPSGKSPANVFQTGKTGATTGELLQRLRDISTVKSLFAKRQQEANAELRSTHSRWKER589(SEQ ID NO:29)G VPS KSPA+VFQTGKTGATTGELLQRLRDISTVKSLFAKRQQEA  E RSTHSRW++R(SEQ ID NO:30)Sbjct:119GAVPSRKSPADVFQTGKTGATTGELLQRLRDISTVKSLFAKRQQEAMREPRSTHSRWRKR178(SEQ ID NO:31)


[0072] The relatedness between the disclsoed PROT4 polypetpide and endothelin-2 polypeptides is further illustrated in the following ClustalW alignment. Black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); greyed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted amino acid residues can potentially be mutated to a much broader extent without altering structure or function.


[0073] Polypeptide sequences are derived from the following sources:
171. Endothelin_Novell = disclosed PROT4 polypetide sequence2. ET2_HUMAN = (ENDOTHELTN-2 PRECURSOR (ET-2) - human): Accn-P20800; PID-g119615 (SEQ ID NO:32)3. ET2_MOUSE = (ENDOTHELIN-2 PRECURSOR (ET-2) - mouse): Accn-P22389; PID-g119616 (SEQ ID NO:33)4. ET2_RAT = (ENDOTHELTIN-2 PRECURSOR (ET-2) (VASOACTIVE INTESTINAL CONTRACTOR) (VIC): Accn-P23943; PID - g2827760 (SEQ ID NO:34)192021


[0074] Nucleic Acids


[0075] One aspect of the invention pertains to isolated nucleic acid molecules that encode PROTX proteins or biologically active portions thereof, as well as nucleic acid fragments sufficient for use as hybridization probes to identify PROTX-encoding nucleic acids (e.g., PROTX mRNA) and fragments for use as PCR primers for the amplification or mutation of PROTX nucleic acid molecules.


[0076] As used herein, the term “nucleic acid molecule” is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.


[0077] An “isolated” nucleic acid molecule is one that is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an “isolated” nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5′ and 3′ ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated PROTX nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. Moreover, an “isolated” nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or of chemical precursors or other chemicals when chemically synthesized.


[0078] In some embodiments, the PROTX nucleic acids encode a mature form a PROTX polypeptide. As used herein, the term a “mature” form of a polypeptide or protein is the product of a naturally occurring polypeptide or precursor form or PROTX-protein. The naturally occurring polypeptide, precursor or PROTX-protein includes, by way of non-limiting example, the full length gene product, encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or PROTX-protein encoded by an open reading frame described herein. The product “mature” form arises, again by way of non-limiting example, as a result of one or more naturally occurring processing steps as they may take place within the cell, or host cell, in which the gene product arises. Examples of such processing steps leading to a “mature” form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an open reading frame, or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+1 to residue N remaining. Further as used herein, a “mature” form of a polypeptide or protein may arise from a step of post-translational modification other than a proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristylation, or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them.


[0079] A nucleic acid molecule of the present invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7, or a complement of any of these nucleotide sequences, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion of the nucleic acid sequences of SEQ ID NO:1, 3, 5, or 7, as a hybridization probe, PROTX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook et al., eds., MOLECULAR CLONING: A LABORATORY MANUAL 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989; and Ausubel, et al., eds., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, N.Y., 1993.)


[0080] A nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to PROTX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.


[0081] In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7. In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7 or a portion of this nucleotide sequence. A nucleic acid molecule that is complementary to the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7 is one that is sufficiently complementary to the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7 that it can hydrogen bond with little or no mismatches to the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7, thereby forming a stable duplex.


[0082] Moreover, the nucleic acid molecule of the invention can comprise only a portion of the nucleic acid sequence of SEQ ID NO:1, 3, 5, or 7, e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically active portion of a PROTX. Fragments provided herein are defined as sequences of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, respectively, and are at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice. Derivatives are nucleic acid sequences or amino acid sequences formed from the native compounds either directly or by modification or partial substitution. Analogs are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound but differs from it in respect to certain components or side chains. Analogs may be synthetic or from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type. Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species.


[0083] Derivatives and analogs may be full length or other than full length, if the derivative or analog contains a modified nucleic acid or amino acid, as described below. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 30%, 50%, 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the aforementioned proteins under stringent, moderately stringent, or low stringent conditions. See e.g. Ausubel, et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, N.Y., 1993, and below.


[0084] The nucleotide sequence determined from the cloning of a huma PROTX gene allows for the generation of probes and primers designed for use in identifying and/or cloning PROTX homologues in other cell types, e.g. from other tissues, as well as PROTX homologues from other mammals. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ ID NO:1, 3, 5, or 7, or an anti-sense strand nucleotide sequence of SEQ ID NO:1, 3, 5, or 7, or of a naturally occurring mutant of SEQ ID NO:1, 3, 5, or 7.


[0085] Probes based on the huma PROTX nucleotide sequence can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In various embodiments, the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress a PROTX protein, such as by measuring a level of a PROTX-encoding nucleic acid in a sample of cells from a subject e.g., detecting PROTX mRNA levels or determining whether a genomic PROTX gene has been mutated or deleted.


[0086] “A polypeptide having a biologically active portion of PROTX” refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a “biologically active portion of PROTX” can be prepared by isolating a portion of SEQ ID NO:1, 3, 5, or 7 that encodes a polypeptide having a PROTX biological activity (the biological activities of the PROTX proteins are described above), expressing the encoded portion of PROTX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of PROTX.


[0087] PROTX Variants


[0088] The invention further encompasses nucleic acid molecules that differ from the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7 due to degeneracy of the genetic code and thus encode the same PROTX protein as that encoded by the nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NO:2, 4, 6, or 8.


[0089] In addition to the huma PROTX nucleotide sequence shown in SEQ ID NO:1, 3, 5, or 7 it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of PROTX may exist within a population (e.g., the human population). Such genetic polymorphism in the PROTX gene may exist among individuals within a population due to natural allelic variation. As used herein, the terms “gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding a PROTX protein, preferably a mammalia PROTX protein. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the PROTX gene. Any and all such nucleotide variations and resulting amino acid polymorphisms in PROTX that are the result of natural allelic variation and that do not alter the functional activity of PROTX are intended to be within the scope of the invention.


[0090] Moreover, nucleic acid molecules encoding PROTX proteins from other species, and thus that have a nucleotide sequence that differs from the human sequence of SEQ ID NO:1, 3, 5, or 7 are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the PROTX cDNAs of the invention can be isolated based on their homology to the huma PROTX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. For example, a soluble huma PROTX cDNA can be isolated based on its homology to human membrane-bound PROTX cDNA. Likewise, a membrane-bound huma PROTX cDNA can be isolated based on its homology to soluble huma PROTX cDNA.


[0091] Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250 or 500 nucleotides in length. In another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term “hybridizes under stringent conditions” is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.


[0092] Homologs (i.e., nucleic acids encoding PROTX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.


[0093] Stringent conditions are known to those skilled in the art and can be found in CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6× SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA at 65° C., followed by one or more washes in 0.2× SSC, 0.01% BSA at 50° C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NO:1, 3, 5, or 7 corresponds to a naturally-occurring nucleic acid molecule. As used herein, a “naturally-occurring” nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).


[0094] In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency hybridization conditions are hybridization in 6× SSC, 5× Denhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55° C., followed by one or more washes in 1× SSC, 0.1% SDS at 37° C. Other conditions of moderate stringency that may be used are well-known in the art. See, e.g., Ausubel et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY.


[0095] In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7 or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions are hybridization in 35% formamide, 5× SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40° C., followed by one or more washes in 2× SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS at 50° C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, e.g., Ausubel et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981, Proc Natl Acad Sci USA 78: 6789-6792.


[0096] Conservative Mutations


[0097] In addition to naturally-occurring allelic variants of the PROTX sequence that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7, thereby leading to changes in the amino acid sequence of the encoded PROTX protein, without altering the functional ability of the PROTX protein. For example, nucleotide substitutions leading to amino acid substitutions at “non-essential” amino acid residues can be made in the sequence of SEQ ID NO:1, 3, 5, or 7 . A “non-essential” amino acid residue is a residue that can be altered from the wild-type sequence of PROTX without altering the biological activity, whereas an “essential” amino acid residue is required for biological activity. For example, amino acid residues that are conserved among the PROTX proteins of the present invention, are predicted to be particularly unamenable to alteration.


[0098] Another aspect of the invention pertains to nucleic acid molecules encoding PROTX proteins that contain changes in amino acid residues that are not essential for activity. Such PROTX proteins differ in amino acid sequence from SEQ ID NO:2, 4, 6, or 8, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 45% homologous to the amino acid sequence of SEQ ID NO:2, 4, 6, or 8. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NO:2, 4, 6, or 8, more preferably at least about 70% homologous to SEQ ID NO:2, 4, 6, or 8, still more preferably at least about 80% homologous to SEQ ID NO:2, 4, 6, or 8, even more preferably at least about 90% homologous to SEQ ID NO:2, 4, 6, or 8, and most preferably at least about 95% homologous to SEQ ID NO:2, 4, 6, or 8.


[0099] An isolated nucleic acid molecule encoding a PROTX protein homologous to the protein of SEQ ID NO:2, 4, 6, or 8 can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7 such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.


[0100] Mutations can be introduced into SEQ ID NO:1, 3, 5, or 7 by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in PROTX is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of a PROTX coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for PROTX biological activity to identify mutants that retain activity. Following mutagenesis of SEQ ID NO:1, 3, 5, or 7 the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined.


[0101] In one embodiment, a mutant PROTX protein can be assayed for (1) the ability to form protein:protein interactions with other PROTX proteins, other cell-surface proteins, or biologically active portions thereof, (2) complex formation between a mutant PROTX protein and a PROTX ligand; (3) the ability of a mutant PROTX protein to bind to an intracellular target protein or biologically active portion thereof; (e.g. avidin proteins).


[0102] Antisense


[0103] Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:1, 3, 5, or 7 or fragments, analogs or derivatives thereof. An “antisense” nucleic acid comprises a nucleotide sequence that is complementary to a “sense” nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence. In specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire PROTX coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of a PROTX protein of SEQ ID NO:2, 4, 6, or 8, or antisense nucleic acids complementary to a PROTX nucleic acid sequence of SEQ ID NO:1, 3, 5, or 7 are additionally provided.


[0104] In one embodiment, an antisense nucleic acid molecule is antisense to a “coding region” of the coding strand of a nucleotide sequence encoding PROTX. The term “coding region” refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues. In another embodiment, the antisense nucleic acid molecule is antisense to a “noncoding region” of the coding strand of a nucleotide sequence encoding PROTX. The term “noncoding region” refers to 5′ and 3′ sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5′ and 3′ untranslated regions).


[0105] Given the coding strand sequences encoding PROTX disclosed herein (e.g., SEQ ID NO:1, 3, 5, or 7), antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of PROTX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of PROTX mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of PROTX mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.


[0106] Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).


[0107] The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a PROTX protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.


[0108] In yet another embodiment, the antisense nucleic acid molecule of the invention is an a-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids Res 15: 6625-6641). The antisense nucleic acid molecule can also comprise a 2′-o-methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res 15: 6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett 215: 327-330).


[0109] Ribozymes and PNA Moieties


[0110] In still another embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave PROTX mRNA transcripts to thereby inhibit translation of PROTX mRNA. A ribozyme having specificity for a PROTX-encoding nucleic acid can be designed based upon the nucleotide sequence of a PROTX cDNA disclosed herein (i.e., SEQ ID NO:1, 3, 5, or 7). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a PROTX-encoding mRNA. See, e.g., Cech et al. U.S. Pat. No. 4,987,071; and Cech et al. U.S. Pat. No. 5,116,742. Alternatively, PROTX mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al., (1993) Science 261:1411-1418.


[0111] Alternatively, PROTX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the PROTX (e.g. the PROTX promoter and/or enhancers) to form triple helical structures that prevent transcription of the PROTX gene in target cells. See generally, Helene. (1991) Anticancer Drug Des. 6: 569-84; Helene. et al. (1992) Ann. N. Acad. Sci. 660:27-36; and Maher (1992) Bioassays 14: 807-15.


[0112] In various embodiments, the nucleic acids of PROTX can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids (see Hyrup et al. (1996) Bioorg Med Chem 4: 5-23). As used herein, the terms “peptide nucleic acids” or “PNAs” refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996) above; Perry-O'Keefe et al. (1996) PNAS 93: 14670-675.


[0113] PNAs of PROTX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of PROTX can also be used, e.g., in the analysis of single base pair mutations in a gene by, e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., S1 nucleases (Hyrup B. (1996) above); or as probes or primers for DNA sequence and hybridization (Hyrup et al. (1996), above; Perry-O'Keefe (1996), above).


[0114] In another embodiment, PNAs of PROTX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of PROTX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes, e.g., RNase H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup (1996) above). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup (1996) above and Finn et al. (1996) Nucl Acids Res 24: 3357-63. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g., 5′-(4-methoxytrityl)amino-5′-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5′ end of DNA (Mag et al. (1989) Nucl Acid Res 17: 5973-88). PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5′ PNA segment and a 3′ DNA segment (Finn et al. (1996) above). Alternatively, chimeric molecules can be synthesized with a 5′ DNA segment and a 3′ PNA segment. See, Petersen et al. (1975) Bioorg Med Chem Lett 5: 1119-11124.


[0115] In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al., 1989, Proc. Natl. Acad. Sci. U.S.A. 86:6553-6556; Lemaitre et al., 1987, Proc. Natl. Acad Sci. 84:648-652; PCT Publication No. WO88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO89/10134). In addition, oligonucleotides can be modified with hybridization triggered cleavage agents (See, e.g., Krol et al., 1988, BioTechniques 6:958-976) or intercalating agents. (See, e.g., Zon, 1988, Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, etc.


[0116] PROTX Proteins


[0117] One aspect of the invention pertains to isolated PROTX proteins, and biologically active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-PROTX antibodies. In one embodiment, native PROTX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, PROTX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, a PROTX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.


[0118] An “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the PROTX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language “substantially free of cellular material” includes preparations of PROTX protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. In one embodiment, the language “substantially free of cellular material” includes preparations of PROTX protein having less than about 30% (by dry weight) of non-PROTX protein (also referred to herein as a “contaminating protein”), more preferably less than about 20% of non-PROTX protein, still more preferably less than about 10% of non-PROTX protein, and most preferably less than about 5% non-PROTX protein. When the PROTX protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.


[0119] The language “substantially free of chemical precursors or other chemicals” includes preparations of PROTX protein in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. In one embodiment, the language “substantially free of chemical precursors or other chemicals” includes preparations of PROTX protein having less than about 30% (by dry weight) of chemical precursors or non-PROTX chemicals, more preferably less than about 20% chemical precursors or non-PROTX chemicals, still more preferably less than about 10% chemical precursors or non-PROTX chemicals, and most preferably less than about 5% chemical precursors or non-PROTX chemicals.


[0120] Biologically active portions of a PROTX protein include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequence of the PROTX protein, e.g., the amino acid sequence shown in SEQ ID NO:2, 4, 6, or 8, that include fewer amino acids than the full length PROTX proteins, and exhibit at least one activity of a PROTX protein. Typically, biologically active portions comprise a domain or motif with at least one activity of the PROTX protein. A biologically active portion of a PROTX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length.


[0121] Moreover, other biologically active portions, in which other regions of the protein are deleted, can be pepared by recombinant techniques and evaluated for one or more of the functional activities of a native PROTX protein.


[0122] In an embodiment, the PROTX protein has an amino acid sequence shown in SEQ ID NO:2, 4, 6, or 8. In other embodiments, the PROTX protein is substantially homologous to SEQ ID NO:2, 4, 6, or 8 and retains the functional activity of the protein of SEQ ID NO:2, 4, 6, or 8 yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail below. Accordingly, in another embodiment, the PROTX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence of SEQ ID NO:2, 4, 6, or 8 and retains the functional activity of the PROTX proteins of SEQ ID NO:2, 4, 6, or 8.


[0123] Determining Homology Between Two or More Sequences


[0124] To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid “homology” is equivalent to amino acid or nucleic acid “identity”).


[0125] The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch 1970 J Mol Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension penalty of 0.3, the coding region of the analogous nucleic acid sequences referred to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence shown in SEQ ID NO:1, 3, 5, or 7.


[0126] The term “sequence identity” refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term “percentage of sequence identity” is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term “substantial identity” as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.


[0127] Chimeric and Fusion Proteins


[0128] The invention also provides PROTX chimeric or fusion proteins. As used herein, a PROTX “chimeric protein” or “fusion protein” comprises a PROTX polypeptide operatively linked to a non-PROTX polypeptide. An “PROTX polypeptide” refers to a polypeptide having an amino acid sequence corresponding to PROTX, whereas a “non-PROTX polypeptide” refers to a polypeptide having an amino acid sequence corresponding to a protein that is not substantially homologous to the PROTX protein, e.g., a protein that is different from the PROTX protein and that is derived from the same or a different organism. Within a PROTX fusion protein the PROTX polypeptide can correspond to all or a portion of a PROTX protein. In one embodiment, a PROTX fusion protein comprises at least one biologically active portion of a PROTX protein. In another embodiment, a PROTX fusion protein comprises at least two biologically active portions of a PROTX protein. In yet another embodiment, a PROTX fusion protein comprises at least three biologically active portions of a PROTX protein. Within the fusion protein, the term “operatively linked” is intended to indicate that the PROTX polypeptide and the non-PROTX polypeptide are fused in-frame to each other. The non-PROTX polypeptide can be fused to the N-terminus or C-terminus of the PROTX polypeptide.


[0129] In yet another embodiment, the fusion protein is a GST-PROTX fusion protein in which the PROTX sequences are fused to the C-terminus of the GST (i e., glutathione S-transferase) sequences. Such fusion proteins can facilitate the purification of recombinant PROTX.


[0130] In another embodiment, the fusion protein is a PROTX protein containing a heterologous signal sequence at its N-terminus. For example, the native PROTX signal sequence can be removed and replaced with a signal sequence from another protein. In certain host cells (e.g., mammalian host cells), expression and/or secretion of PROTX can be increased through use of a heterologous signal sequence.


[0131] In yet another embodiment, the fusion protein is a PROTX-immunoglobulin fusion protein in which the PROTX are fused to sequences derived from a member of the immunoglobulin protein family. The PROTX-immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a PROTX ligand and a PROTX protein on the surface of a cell, to thereby suppress PROTX-mediated signal transduction in vivo. The PROTX-immunoglobulin fusion proteins can be used to affect the bioavailability of a PROTX cognate ligand. Inhibition of the PROTX ligand/PROTX interaction are useful therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. promoting or inhibiting) cell survival. Moreover, the PROTX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-PROTX antibodies in a subject, to purify PROTX ligands, and in screening assays to identify molecules that inhibit the interaction of PROTX with a PROTX ligand.


[0132] A PROTX chimeric or fusion protein of the invention can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Ausubel et al. (eds.) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A PROTX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the PROTX protein.


[0133] PROTX Agonists and Antagonists


[0134] The present invention also pertains to variants of the PROTX proteins that function as either PROTX agonists (mimetics) or as PROTX antagonists. Variants of the PROTX protein can be generated by mutagenesis, e.g., discrete point mutation or truncation of the PROTX protein. An agonist of the PROTX protein can retain substantially the same, or a subset of, the biological activities of the naturally occurring form of the PROTX protein. An antagonist of the PROTX protein can inhibit one or more of the activities of the naturally occurring form of the PROTX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the PROTX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the PROTX proteins.


[0135] Variants of the PROTX protein that function as either PROTX agonists (mimetics) or as PROTX antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the PROTX protein for PROTX protein agonist or antagonist activity. In one embodiment, a variegated library of PROTX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of PROTX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential PROTX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of PROTX sequences therein. There are a variety of methods which can be used to produce libraries of potential PROTX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an Attorney Ref: 15966-662 appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential PROTX sequences. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang (1983) Tetrahedron 39:3; Itakura et al. (1984) Annu Rev Biochem 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucl Acid Res 11:477.


[0136] Polypeptide Libraries


[0137] In addition, libraries of fragments of the PROTX protein coding sequence can be used to generate a variegated population of PROTX fragments for screening and subsequent selection of variants of a PROTX protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of a PROTX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S1 nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes N-terminal and internal fragments of various sizes of the PROTX protein.


[0138] Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of PROTX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recrusive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify PROTX variants (Arkin and Yourvan (1992) PNAS 89:7811-7815; Delgrave et al. (1993) Protein Engineering 6:327-331).


[0139] Anti-PROTX Antibodies


[0140] An isolated PROTX protein, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind PROTX using standard techniques for polyclonal and monoclonal antibody preparation. The full-length PROTX protein can be used or, alternatively, the invention provides antigenic peptide fragments of PROTX for use as immunogens. The antigenic peptide of PROTX comprises at least 8 amino acid residues of the amino acid sequence shown in SEQ ID NO:2, 4, 6, or 8 and encompasses an epitope of PROTX such that an antibody raised against the peptide forms a specific immune complex with PROTX. Preferably, the antigenic peptide comprises at least 10 amino acid residues, more preferably at least 15 amino acid residues, even more preferably at least 20 amino acid residues, and most preferably at least 30 amino acid residues. Preferred epitopes encompassed by the antigenic peptide are regions of PROTX that are located on the surface of the protein, e.g., hydrophilic regions.


[0141] As disclosed herein, PROTX protein sequence of SEQ ID NO:2, 4, 6, or 8, or derivatives, fragments, analogs or homologs thereof, may be utilized as immunogens in the generation of antibodies that immunospecifically-bind these protein components. The term “antibody” as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen, such as PROTX. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab and F(ab′)2 fragments, and an Fab expression library. In a specific embodiment, antibodies to huma PROTX proteins are disclosed. Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies to a PROTX protein sequence of SEQ ID NO:2, 4, 6, or 8, or derivative, fragment, analog or homolog thereof.


[0142] For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by injection with the native protein, or a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, recombinantly expressed PROTX protein or a chemically synthesized PROTX polypeptide. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, etc.), human adjuvants such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents. If desired, the antibody molecules directed against PROTX can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction.


[0143] The term “monoclonal antibody” or “monoclonal antibody composition”, as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of PROTX. A monoclonal antibody composition thus typically displays a single binding affinity for a particular PROTX protein with which it immunoreacts. For preparation of monoclonal antibodies directed towards a particular PROTX protein, or derivatives, fragments, analogs or homologs thereof, any technique that provides for the production of antibody molecules by continuous cell line culture may be utilized. Such techniques include, but are not limited to, the hybridoma technique (see Kohler & Milstein, 1975 Nature 256: 495-497); the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al., 1983 Immunol Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).


[0144] According to the invention, techniques can be adapted for the production of single-chain antibodies specific to a PROTX protein (see e.g., U.S. Pat. No. 4,946,778). In addition, methodologies can be adapted for the construction of Fab expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a PROTX protein or derivatives, fragments, analogs or homologs thereof. Non-human antibodies can be “humanized” by techniques well known in the art. See e.g., U.S. Pat. No. 5,225,539. Antibody fragments that contain the idiotypes to a PROTX protein may be produced by techniques known in the art including, but not limited to: (i) an F(ab′)2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(ab′)2 fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.


[0145] Additionally, recombinant anti-PROTX antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT International Application No. PCT/US86/02269; European Patent Application No. 184,187; European Patent Application No. 171,496; European Patent Application No. 173,494; PCT International Publication No. WO 86/01533; U.S. Pat. No. 4,816,567; European Patent Application No. 125,023; Better et al.(1988) Science 240:1041-1043; Liu et al. (1987) PNAS 84:3439-3443; Liu et al. (1987) J Immunol. 139:3521-3526; Sun et al. (1987) PNAS 84:214-218; Nishimura et al. (1987) Cancer Res 47:999-1005; Wood et al. (1985) Nature 314:446-449; Shaw et al. (1988) J Natl Cancer Inst 80:1553-1559); Morrison (1985) Science 229:1202-1207; Oi et al. (1986) BioTechniques 4:214; U.S. Pat. No. 5,225,539; Jones et al. (1986) Nature 321:552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J Immunol 141:4053-4060.


[0146] In one embodiment, methodologies for the screening of antibodies that possess the desired specificity include, but are not limited to, enzyme-linked immunosorbent assay (ELISA) and other immunologically-mediated techniques known within the art.


[0147] Anti-PROTX antibodies may be used in methods known within the art relating to the localization and/or quantitation of a PROTX protein (e.g., for use in measuring levels of the PROTX protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like). In a given embodiment, antibodies for PROTX proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antibody derived binding domain, are utilized as pharmacologically-active compounds [hereinafter “Therapeutics”].


[0148] An anti-PROTX antibody (e.g., monoclonal antibody) can be used to isolate PROTX by standard techniques, such as affinity chromatography or immunoprecipitation. An anti-PROTX antibody can facilitate the purification of natural PROTX from cells and of recombinantly produced PROTX expressed in host cells. Moreover, an anti-PROTX antibody can be used to detect PROTX protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the PROTX protein. Anti-PROTX antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131I, 35S or 3H.


[0149] PROTX Recombinant Expression Vectors and Host Cells


[0150] Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding PROTX protein, or derivatives, fragments, analogs or homologs thereof. As used herein, the term “vector” refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “expression vectors”. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, “plasmid” and “vector” can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.


[0151] The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, “operably linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term “regulatory sequence” is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; GENE ExPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., PROTX proteins, mutant forms of PROTX, fusion proteins, etc.).


[0152] The recombinant expression vectors of the invention can be designed for expression of PROTX in prokaryotic or eukaryotic cells. For example, PROTX can be expressed in bacterial cells such as E. coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.


[0153] Expression of proteins in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three purposes: (1) to increase expression of recombinant protein; (2) to increase the solubility of the recombinant protein; and (3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) that fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.


[0154] Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al., (1988) Gene 69:301-315) and pET 11d (Studier et al., GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89).


[0155] One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al., (1992) Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.


[0156] In another embodiment, the PROTX expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerivisae include pYepSec1 (Baldari, et al., (1987) EMBO J 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-943), pJRY88 (Schultz et al., (1987) Gene 54:113-123), pYES2 (Invitrogen Corporation, San Diego, Calif.), and picZ (InVitrogen Corp, San Diego, Calif.).


[0157] Alternatively, PROTX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., SF9 cells) include the pAc series (Smith et al. (1983) Mol Cell Biol 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).


[0158] In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBO J 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells. See, e.g., Chapters 16 and 17 of Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.


[0159] In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev 1:268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv Immunol 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J 8:729-733) and immunoglobulins (Banerji et al. (1983) Cell 33:729-740; Queen and Baltimore (1983) Cell 33:741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle (1989) PNAS 86:5473-5477), pancreas-specific promoters (Edlund et al. (1985) Science 230:912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, e.g., the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the α-fetoprotein promoter (Campes and Tilghman (1989) Genes Dev 3:537-546).


[0160] The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to PROTX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see Weintraub et al., “Antisense RNA as a molecular tool for genetic analysis,” Reviews—Trends in Genetics, Vol. 1(1) 1986.


[0161] Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms “host cell” and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.


[0162] A host cell can be any prokaryotic or eukaryotic cell. For example, PROTX protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.


[0163] Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms “transformation” and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.


[0164] For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding PROTX or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).


[0165] A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) PROTX protein. Accordingly, the invention further provides methods for producing PROTX protein using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding PROTX has been introduced) in a suitable medium such that PROTX protein is produced. In another embodiment, the method further comprises isolating PROTX from the medium or the host cell.


[0166] Transgenic Animals


[0167] The host cells of the invention can also be used to produce nonhuman transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which PROTX-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous PROTX sequences have been introduced into their genome or homologous recombinant animals in which endogenous PROTX sequences have been altered. Such animals are useful for studying the function and/or activity of PROTX and for identifying and/or evaluating modulators of PROTX activity. As used herein, a “transgenic animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a “homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous PROTX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.


[0168] A transgenic animal of the invention can be created by introducing PROTX-encoding nucleic acid into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. The huma PROTX cDNA sequence of SEQ ID NO:1, 3, 5, or 7 can be introduced as a transgene into the genome of a non-human animal. Alternatively, a nonhuman homologue of the huma PROTX gene, such as a mouse PROTX gene, can be isolated based on hybridization to the huma PROTX cDNA (described further above) and used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably linked to the PROTX transgene to direct expression of PROTX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866; 4,870,009; and 4,873,191; and Hogan 1986, In: MANIPULATING THE MOUSE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the PROTX transgene in its genome and/or expression of PROTX mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene encoding PROTX can further be bred to other transgenic animals carrying other transgenes.


[0169] To create a homologous recombinant animal, a vector is prepared which contains at least a portion of a PROTX gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the PROTX gene. The PROTX gene can be a human gene (e.g., the cDNA of SEQ ID NO:1, 3, 5, or 7) but more preferably, is a non-human homologue of a huma PROTX gene. For example, a mouse homologue of huma PROTX gene of SEQ ID NO:1, 3, 5, or 7 can be used to construct a homologous recombination vector suitable for altering an endogenous PROTX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous PROTX gene is functionally disrupted (ie., no longer encodes a functional protein; also referred to as a “knock out” vector).


[0170] Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous PROTX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous PROTX protein). In the homologous recombination vector, the altered portion of the PROTX gene is flanked at its 5′ and 3′ ends by additional nucleic acid of the PROTX gene to allow for homologous recombination to occur between the exogenous PROTX gene carried by the vector and an endogenous PROTX gene in an embryonic stem cell. The additional flanking PROTX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5′ and 3′ ends) are included in the vector. See e.g., Thomas et al. (1987) Cell 51:503 for a description of homologous recombination vectors. The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced PROTX gene has homologously recombined with the endogenous PROTX gene are selected (see e.g., Li et al. (1992) Cell 69:915).


[0171] The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. See e.g., Bradley 1987, In: TERATOCARCINOMAS AND EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley (1991) Curr Opin Biotechnol 2:823-829; PCT International Publication Nos.: WO 90/11354; WO 91/01140; WO 92/0968; and WO 93/04169.


[0172] In another embodiment, transgenic non-human animals can be produced that contain selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage P1. For a description of the cre/loxP recombinase system, see, e.g., Lakso et al. (1992) PNAS 89:6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of “double” transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.


[0173] Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut et al. (1997) Nature 385:810-813. In brief, a cell, e.g., a somatic cell, from the transgenic animal can be isolated and induced to exit the growth cycle and enter G0 phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell, e.g., the somatic cell, is isolated.


[0174] Pharmaceutical Compositions


[0175] The PROTX nucleic acid molecules, PROTX proteins, and anti-PROTX antibodies (also referred to herein as “active compounds”) of the invention, and derivatives, fragments, analogs and homologs thereof, can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.


[0176] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


[0177] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifingal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.


[0178] Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a PROTX protein or anti-PROTX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


[0179] Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.


[0180] For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.


[0181] Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.


[0182] The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.


[0183] In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.


[0184] It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.


[0185] The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Pat. No. 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) PNAS 91:3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.


[0186] The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.


[0187] Uses and Methods of the Invention


[0188] The isolated nucleic acid molecules of the invention can be used to express PROTX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect PROTX mRNA (e.g., in a biological sample) or a genetic lesion in a PROTX gene, and to modulate PROTX activity, as described further below. In addition, the PROTX proteins can be used to screen drugs or compounds that modulate the PROTX activity or expression as well as to treat disorders characterized by insufficient or excessive production of PROTX protein or production of PROTX protein forms that have decreased or aberrant activity compared to PROTX wild type protein (e.g. proliferative disorders such as cancer or preclampsia, immune system disorders and inflammation, neurological disorders, and skin and muscle abnormalities). In addition, the anti-PROTX antibodies of the invention can be used to detect and isolate PROTX proteins and modulate PROTX activity.


[0189] This invention further pertains to novel agents identified by the above described screening assays and uses thereof for treatments as described herein.


[0190] Screening Assays


[0191] The invention provides a method (also referred to herein as a “screening assay”) for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) that bind to PROTX proteins or have a stimulatory or inhibitory effect on, for example, PROTX expression or PROTX activity.


[0192] In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of a membrane-bound form of a PROTX protein or polypeptide or biologically active portion thereof. The test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the “one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam (1997) Anticancer Drug Des 12:145).


[0193] Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al. (1993) Proc Natl Acad Sci U.S.A. 90:6909; Erb et al. (1994) Proc Natl Acad Sci U.S.A. 91:11422; Zuckermann et al. (1994) J Med Chem 37:2678; Cho et al. (1993) Science 261:1303; Carrell et al. (1994) Angew Chem Int Ed Engl 33:2059; Carell et al. (1994) Angew Chem Int Ed Engl 33:2061; and Gallop et al. (1994) J Med Chem 37:1233.


[0194] Libraries of compounds may be presented in solution (e.g., Houghten (1992) Biotechniques 13:412-421), or on beads (Lam (1991) Nature 354:82-84), on chips (Fodor (1993) Nature 364:555-556), bacteria (Ladner U.S. Pat. No. 5,223,409), spores (Ladner USP '409), plasmids (Cull et al. (1992) Proc Natl Acad Sci USA 89:1865-1869) or on phage (Scott and Smith (1990) Science 249:386-390; Devlin (1990) Science 249:404-406; Cwirla et al. (1990) Proc Natl Acad Sci U.S.A. 87:6378-6382; Felici (1991) J Mol Biol 222:301-310; Ladner above.).


[0195] In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of PROTX protein, or a biologically active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to a PROTX protein is determined. The cell, for example, can be of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the PROTX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the PROTX protein or biologically active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 125I, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. In one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of PROTX protein, or a biologically active portion thereof, on the cell surface with a known compound which binds PROTX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with PROTX protein, wherein determining the ability of the test compound to interact with a PROTX protein comprises determining the ability of the test compound to preferentially bind to PROTX or a biologically active portion thereof as compared to the known compound.


[0196] In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of PROTX protein, or a biologically active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the PROTX protein or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of PROTX or a biologically active portion thereof can be accomplished, for example, by determining the ability of the PROTX protein to bind to or interact with a PROTX target molecule. As used herein, a “target molecule” is a molecule with which a PROTX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses a PROTX protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule. A PROTX target molecule can be a non-PROTX molecule or a PROTX protein or polypeptide of the present invention. In one embodiment, a PROTX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound PROTX molecule) through the cell membrane and into the cell. The target, for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with PROTX.


[0197] Determining the ability of the PROTX protein to bind to or interact with a PROTX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the PROTX protein to bind to or interact with a PROTX target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising a PROTX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation.


[0198] In yet another embodiment, an assay of the present invention is a cell-free assay comprising contacting a PROTX protein or biologically active portion thereof with a test compound and determining the ability of the test compound to bind to the PROTX protein or biologically active portion thereof. Binding of the test compound to the PROTX protein can be determined either directly or indirectly as described above. In one embodiment, the assay comprises contacting the PROTX protein or biologically active portion thereof with a known compound which binds PROTX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a PROTX protein, wherein determining the ability of the test compound to interact with a PROTX protein comprises determining the ability of the test compound to preferentially bind to PROTX or biologically active portion thereof as compared to the known compound.


[0199] In another embodiment, an assay is a cell-free assay comprising contacting PROTX protein or biologically active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the PROTX protein or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of PROTX can be accomplished, for example, by determining the ability of the PROTX protein to bind to a PROTX target molecule by one of the methods described above for determining direct binding. In an alternative embodiment, determining the ability of the test compound to modulate the activity of PROTX can be accomplished by determining the ability of the PROTX protein further modulate a PROTX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as previously described.


[0200] In yet another embodiment, the cell-free assay comprises contacting the PROTX protein or biologically active portion thereof with a known compound which binds PROTX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with a PROTX protein, wherein determining the ability of the test compound to interact with a PROTX protein comprises determining the ability of the PROTX protein to preferentially bind to or modulate the activity of a PROTX target molecule.


[0201] The cell-free assays of the present invention are amenable to use of both the soluble form or the membrane-bound form of PROTX. In the case of cell-free assays comprising the membrane-bound form of PROTX, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of PROTX is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-100, Triton® X-114, Thesit®, Isotridecypoly(ethylene glycol ether)n, 3-(3-cholamidopropyl)dimethylamminiol-1-propane sulfonate (CHAPS), 3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy-1-propane sulfonate (CHAPSO), or N-dodecyl—N,N-dimethyl-3-ammonio-1-propane sulfonate.


[0202] In more than one embodiment of the above assay methods of the present invention, it may be desirable to immobilize either PROTX or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to PROTX, or interaction of PROTX with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix. For example, GST-PROTX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or PROTX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of PROTX binding or activity determined using standard techniques.


[0203] Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either PROTX or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated PROTX or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with PROTX or target molecules, but which do not interfere with binding of the PROTX protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or PROTX trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the PROTX or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the PROTX or target molecule.


[0204] In another embodiment, modulators of PROTX expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of PROTX mRNA or protein in the cell is determined. The level of expression of PROTX mRNA or protein in the presence of the candidate compound is compared to the level of expression of PROTX mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of PROTX expression based on this comparison. For example, when expression of PROTX mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of PROTX mRNA or protein expression. Alternatively, when expression of PROTX mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of PROTX MRNA or protein expression. The level of PROTX mRNA or protein expression in the cells can be determined by methods described herein for detecting PROTX mRNA or protein.


[0205] In yet another aspect of the invention, the PROTX proteins can be used as “bait proteins” in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol Chem 268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and Brent WO94/10300), to identify other proteins that bind to or interact with PROTX (“PROTX-binding proteins” or “PROTX-bps”) and modulate PROTX activity. Such PROTX-binding proteins are also likely to be involved in the propagation of signals by the PROTX proteins as, for example, upstream or downstream elements of the PROTX pathway.


[0206] The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for PROTX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein (“prey” or “sample”) is fused to a gene that codes for the activation domain of the known transcription factor. If the “bait” and the “prey” proteins are able to interact, in vivo, forming a PROTX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close PROTXimity. This PROTXimity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with PROTX.


[0207] This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.


[0208] Detection Assays


[0209] Portions or fragments of the cDNA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. These applications are described in the subsections below.


[0210] Chromosome Mapping


[0211] Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the PROTX, sequences, described herein, can be used to map the location of the PROTX genes, respectively, on a chromosome. The mapping of the PROTX sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease.


[0212] Briefly, PROTX genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the PROTX sequences. Computer analysis of the PROTX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the PROTX sequences will yield an amplified fragment.


[0213] Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. (D'Eustachio et al. (1983) Science 220:919-924). Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.


[0214] PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the PROTX sequences to design oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes.


[0215] Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, see Verma et al., HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988).


[0216] Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents corresponding to noncoding regions of the genes actually are preferred for mapping purposes. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.


[0217] Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, for example, in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, for example, Egeland et al. (1987) Nature, 325:783-787.


[0218] Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the PROTX gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymorphisms.


[0219] Tissue Typing


[0220] The PROTX sequences of the present invention can also be used to identify individuals from minute biological samples. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. The sequences of the present invention are useful as additional DNA markers for RFLP (“restriction fragment length polymorphisms,” described in U.S. Pat. No. 5,272,057).


[0221] Furthermore, the sequences of the present invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the PROTX sequences described herein can be used to prepare two PCR primers from the 5′ and 3′ ends of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.


[0222] Panels of corresponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the present invention can be used to obtain such identification sequences from individuals and from tissue. The PROTX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymorphisms (SNPs), which include restriction fragment length polymorphisms (RFLPs).


[0223] Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification purposes. Because greater numbers of polymorphisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. For example, the noncoding sequences of SEQ ID NO:1, 3, 5, or 7 can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as the nucleic acid seqences that code for the amino acid sequences of SEQ ID NO:2, 4, 6, or 8 are used, a more appropriate number of primers for positive individual identification would be 500-2,000.


[0224] Predictive Medicine


[0225] The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trails are used for prognostic (predictive) purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the present invention relates to diagnostic assays for determining PROTX protein and/or nucleic acid expression as well as PROTX activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant PROTX expression or activity. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with PROTX protein, nucleic acid expression or activity. For example, mutations in a PROTX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with PROTX protein, nucleic acid expression or activity.


[0226] Another aspect of the invention provides methods for determining PROTX protein, nucleic acid expression or PROTX activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (referred to herein as “pharmacogenomics”). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent).


[0227] Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of PROTX in clinical trials.


[0228] These and other agents are described in further detail in the following sections.


[0229] Diagnostic Assays


[0230] An exemplary method for detecting the presence or absence of PROTX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting PROTX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes PROTX protein such that the presence of PROTX is detected in the biological sample. An agent for detecting PROTX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to PROTX mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length PROTX nucleic acid, such as the nucleic acid of SEQ ID NO:1, 3, 5, or 7, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to PROTX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein.


[0231] An agent for detecting PROTX protein is an antibody capable of binding to PROTX protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab′)2) can be used. The term “labeled”, with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin. The term “biological sample” is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect PROTX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of PROTX mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of PROTX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence. In vitro techniques for detection of PROTX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of PROTX protein include introducing into a subject a labeled anti-PROTX antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.


[0232] In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A preferred biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.


[0233] In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting PROTX protein, mRNA, or genomic DNA, such that the presence of PROTX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of PROTX protein, mRNA or genomic DNA in the control sample with the presence of PROTX protein, mRNA or genomic DNA in the test sample.


[0234] The invention also encompasses kits for detecting the presence of PROTX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting PROTX protein or mRNA in a biological sample; means for determining the amount of PROTX in the sample; and means for comparing the amount of PROTX in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect PROTX protein or nucleic acid.


[0235] Prognostic Assays


[0236] The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant PROTX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with PROTX protein, nucleic acid expression or activity such as cancer or fibrotic disorders. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the present invention provides a method for identifying a disease or disorder associated with aberrant PROTX expression or activity in which a test sample is obtained from a subject and PROTX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of PROTX protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with aberrant PROTX expression or activity. As used herein, a “test sample” refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.


[0237] Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with aberrant PROTX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder, such as cancer or preclampsia. Thus, the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant PROTX expression or activity in which a test sample is obtained and PROTX protein or nucleic acid is detected (e.g., wherein the presence of PROTX protein or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant PROTX expression or activity.)


[0238] The methods of the invention can also be used to detect genetic lesions in a PROTX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by aberrant cell proliferation and/or differentiation. In various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding a PROTX-protein, or the mis-expression of the PROTX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of (1) a deletion of one or more nucleotides from a PROTX gene; (2) an addition of one or more nucleotides to a PROTX gene; (3) a substitution of one or more nucleotides of a PROTX gene, (4) a chromosomal rearrangement of a PROTX gene; (5) an alteration in the level of a messenger RNA transcript of a PROTX gene, (6) aberrant modification of a PROTX gene, such as of the methylation pattern of the genomic DNA, (7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of a PROTX gene, (8) a non-wild type level of a PROTX-protein, (9) allelic loss of a PROTX gene, and (10) inappropriate post-translational modification of a PROTX-protein. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in a PROTX gene. A preferred biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.


[0239] In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1994) PNAS 91:360-364), the latter of which can be particularly useful for detecting point mutations in a PROTX-gene (see Abravaya et al. (1995) Nucl Acids Res 23:675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to a PROTX gene under conditions such that hybridization and amplification of the PROTX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.


[0240] Alternative amplification methods include: self sustained sequence replication (Guatelli et al., 1990, Proc Natl Acad Sci USA 87:1874-1878), transcriptional amplification system (Kwoh, et al., 1989, Proc Natl Acad Sci USA 86:1173-1177), Q-Beta Replicase (Lizardi et al, 1988, BioTechnology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.


[0241] In an alternative embodiment, mutations in a PROTX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicate mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, for example, U.S. Pat. No. 5,493,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.


[0242] In other embodiments, genetic mutations in PROTX can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotides probes (Cronin et al. (1996) Human Mutation 7: 244-255; Kozal et al. (1996) Nature Medicine 2: 753-759). For example, genetic mutations in PROTX can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin et al. above. Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.


[0243] In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the PROTX gene and detect mutations by comparing the sequence of the sample PROTX with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert (1977) PNAS 74:560 or Sanger (1977) PNAS 74:5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (Naeve et al., (1995) Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT International Publ. No. WO 94/16101; Cohen et al. (1996) Adv Chromatogr 36:127-162; and Griffin et al. (1993) Appl Biochem Biotechnol 38:147-159).


[0244] Other methods for detecting mutations in the PROTX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242). In general, the art technique of “mismatch cleavage” starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type PROTX sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with SI nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al (1988) Proc Natl Acad Sci USA 85:4397; Saleeba et al (1992) Methods Enzymol 217:286-295. In an embodiment, the control DNA or RNA can be labeled for detection.


[0245] In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called “DNA mismatch repair” enzymes) in defined systems for detecting and mapping point mutations in PROTX cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662). According to an exemplary embodiment, a probe based on a PROTX sequence, e.g., a wild-type PROTX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S. Pat. No. 5,459,039.


[0246] In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in PROTX genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc Natl Acad Sci USA: 86:2766, see also Cotton (1993) Mutat Res 285:125-144; Hayashi (1992) Genet Anal Tech Appl 9:73-79). Single-stranded DNA fragments of sample and control PROTX nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet 7:5).


[0247] In yet another embodiment the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of apPROTXimately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:12753).


[0248] Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al. (1989) Proc Natl Acad. Sci USA 86:6230). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.


[0249] Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res 17:2437-2448) or at the extreme 3′ end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11:238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al (1992) Mol Cell Probes 6:1). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc Natl Acad Sci USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3′ end of the 5′ sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.


[0250] The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving a PROTX gene.


[0251] Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which PROTX is expressed may be utilized in the prognostic assays described herein. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.


[0252] Pharmacogenomics


[0253] Agents, or modulators that have a stimulatory or inhibitory effect on PROTX activity (e.g., PROTX gene expression), as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders (e.g., cancer or immune disorders, neurological disorders, muscular dystrophy, or epidermolysis bullosa simplex) associated with aberrant PROTX activity. In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of PROTX protein, expression of PROTX nucleic acid, or mutation content of PROTX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.


[0254] Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See e.g., Eichelbaum, Clin Exp Pharmacol Physiol, 1996, 23:983-985 and Linder, Clin Chem, 1997, 43:254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body (altered drug action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drug metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymorphisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is haemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.


[0255] As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite morphine. The other extreme is the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.


[0256] Thus, the activity of PROTX protein, expression of PROTX nucleic acid, or mutation content of PROTX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymorphic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with a PROTX modulator, such as a modulator identified by one of the exemplary screening assays described herein.


[0257] Monitoring of Effects During Clinical Trials


[0258] Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of PROTX (e.g., the ability to modulate aberrant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to increase PROTX gene expression, protein levels, or upregulate PROTX activity, can be monitored in clinical trails of subjects exhibiting decreased PROTX gene expression, protein levels, or downregulated PROTX activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease PROTX gene expression, protein levels, or downregulate PROTX activity, can be monitored in clinical trails of subjects exhibiting increased PROTX gene expression, protein levels, or upregulated PROTX activity. In such clinical trials, the expression or activity of PROTX and, preferably, other genes that have been implicated in, for example, a cellular proliferation disorder can be used as a “read out” or markers of the immune responsiveness of a particular cell.


[0259] For example, and not by way of limitation, genes, including PROTX, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) that modulates PROTX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of PROTX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of PROTX or other genes. In this way, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.


[0260] In one embodiment, the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of expression of a PROTX protein, mRNA, or genomic DNA in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the PROTX protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the PROTX protein, mRNA, or genomic DNA in the pre-administration sample with the PROTX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of PROTX to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of PROTX to lower levels than detected, i.e., to decrease the effectiveness of the agent.


[0261] Methods of Treatment


[0262] The present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant PROTX expression or activity.


[0263] Disorders


[0264] Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid and nucleic acids that are “dysfunctional” (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to “knockout” endogenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989. Science 244: 1288-1292); or (v) modulators (i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner.


[0265] Diseases and disorders that are characterized by decreased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.


[0266] Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, etc.).


[0267] Prophylactic Methods


[0268] In one aspect, the invention provides a method for preventing, in a subject, a disease or condition associated with an aberrant PROTX expression or activity, by administering to the subject an agent that modulates PROTX expression or at least one PROTX activity. Subjects at risk for a disease that is caused or contributed to by aberrant PROTX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the PROTX aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending on the type of PROTX aberrancy, for example, a PROTX agonist or PROTX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the present invention are further discussed in the following subsections.


[0269] Therapeutic Methods


[0270] Another aspect of the invention pertains to methods of modulating PROTX expression or activity for therapeutic purposes. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of PROTX protein activity associated with the cell. An agent that modulates PROTX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of a PROTX protein, a peptide, a PROTX peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more PROTX protein activity. Examples of such stimulatory agents include active PROTX protein and a nucleic acid molecule encoding PROTX that has been introduced into the cell. In another embodiment, the agent inhibits one or more PROTX protein activity. Examples of such inhibitory agents include antisense PROTX nucleic acid molecules and anti-PROTX antibodies. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by aberrant expression or activity of a PROTX protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., upregulates or downregulates) PROTX expression or activity. In another embodiment, the method involves administering a PROTX protein or nucleic acid molecule as therapy to compensate for reduced or aberrant PROTX expression or activity.


[0271] Stimulation of PROTX activity is desirable in situations in which PROTX is abnormally downregulated and/or in which increased PROTX activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by aberrant cell proliferation and/or differentiation (e.g., cancer). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia).


[0272] The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.


[0273] This invention is further illustrated by the following examples, which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are hereby incorporated by reference.


[0274] Disease Pathways


[0275] Determination of the Biological Effect of the Therapeutic


[0276] In various embodiments of the present invention, suitable in vitro or in vivo assays are utilized to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue.


[0277] In various specific embodiments, in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model systems known in the art may be used prior to administration to human subjects.


[0278] Malignancies


[0279] An aforementioned protein is involved in the regulation of cell proliferation. Accordingly, Therapeutics of the present invention are useful in the therapeutic or prophylactic treatment of diseases or disorders that are associated with cell hyperproliferation and/or loss of control of cell proliferation (e.g., cancers, malignancies and tumors). For a review of such hyperproliferation disorders, see e.g., Fishman, et al., 1985. MEDICINE, 2nd ed., J. B. Lippincott Co., Philadelphia, Pa.


[0280] Therapeutics of the present invention may be assayed by any method known within the art for efficacy in treating or preventing malignancies and related disorders. Such assays include, but are not limited to, in vitro assays utilizing transformed cells or cells derived from the patient's tumor, as well as in vivo assays using animal models of cancer or malignancies. Potentially effective Therapeutics are those that, for example, inhibit the proliferation of tumor-derived or transformed cells in culture or cause a regression of tumors in animal models, in comparison to the controls.


[0281] In the practice of the present invention, once a malignancy or cancer has been shown to be amenable to treatment by modulating (i.e., inhibiting, antagonizing or agonizing) activity, that cancer or malignancy may subsequently be treated or prevented by the administration of a Therapeutic that serves to modulate protein function.


[0282] Premalignant Conditions


[0283] The Therapeutics of the present invention that are effective in the therapeutic or prophylactic treatment of cancer or malignancies may also be administered for the treatment of pre-malignant conditions and/or to prevent the progression of a pre-malignancy to a neoplastic or malignant state. Such prophylactic or therapeutic use is indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia or, most particularly, dysplasia has occurred. For a review of such abnormal cell growth see e.g., Robbins & Angell, 1976. BASIC PATHOLOGY, 2nd ed., W. B. Saunders Co., Philadelphia, Pa.


[0284] Hyperplasia is a form of controlled cell proliferation involving an increase in cell number in a tissue or organ, without significant alteration in its structure or function. For example, it has been demonstrated that endometrial hyperplasia often precedes endometrial cancer. Metaplasia is a form of controlled cell growth in which one type of mature or fully differentiated cell substitutes for another type of mature cell. Metaplasia may occur in epithelial or connective tissue cells. Dysplasia is generally considered a precursor of cancer, and is found mainly in the epithelia. Dysplasia is the most disorderly form of non-neoplastic cell growth, and involves a loss in individual cell uniformity and in the architectural orientation of cells. Dysplasia characteristically occurs where there exists chronic irritation or inflammation, and is often found in the cervix, respiratory passages, oral cavity, and gall bladder.


[0285] Alternatively, or in addition to the presence of abnormal cell growth characterized as hyperplasia, metaplasia, or dysplasia, the presence of one or more characteristics of a transformed or malignant phenotype displayed either in vivo or in vitro within a cell sample derived from a patient, is indicative of the desirability of prophylactic/therapeutic administration of a Therapeutic that possesses the ability to modulate activity of An aforementioned protein. Characteristics of a transformed phenotype include, but are not limited to: (i) morphological changes; (ii) looser substratum attachment; (iii) loss of cell-to-cell contact inhibition; (iv) loss of anchorage dependence; (v) protease release; (vi) increased sugar transport; (vii) decreased serum requirement; (viii) expression of fetal antigens, (ix) disappearance of the 250 kDal cell-surface protein, and the like. See e.g., Richards, et al, 1986. MOLECULAR PATHOLOGY, W. B. Saunders Co., Philadelphia, Pa.


[0286] In a specific embodiment of the present invention, a patient that exhibits one or more of the following predisposing factors for malignancy is treated by administration of an effective amount of a Therapeutic: (i) a chromosomal translocation associated with a malignancy (e.g., the Philadelphia chromosome (bcr/abl) for chronic myelogenous leukemia and t(14;18) for follicular lymphoma, etc.); (ii) familial polyposis or Gardner's syndrome (possible forerunners of colon cancer); (iii) monoclonal gammopathy of undetermined significance (a possible precursor of multiple myeloma) and (iv) a first degree kinship with persons having a cancer or pre-cancerous disease showing a Mendelian (genetic) inheritance pattern (e.g., familial polyposis of the colon, Gardner's syndrome, hereditary exostosis, polyendocrine adenomatosis, Peutz-Jeghers syndrome, neurofibromatosis of Von Recklinghausen, medullary thyroid carcinoma with amyloid production and pheochromocytoma, retinoblastoma, carotid body tumor, cutaneous melanocarcinoma, intraocular melanocarcinoma, xeroderma pigmentosum, ataxia telangiectasia, Chediak-Higashi syndrome, albinism, Fanconi's aplastic anemia and Bloom's syndrome).


[0287] In another embodiment, a Therapeutic of the present invention is administered to a human patient to prevent the progression to breast, colon, lung, pancreatic, or uterine cancer, or melanoma or sarcoma.


[0288] Hyperproliferative and Dysproliferative Disorders


[0289] In one embodiment of the present invention, a Therapeutic is administered in the therapeutic or prophylactic treatment of hyperproliferative or benign dysproliferative disorders. The efficacy in treating or preventing hyperproliferative diseases or disorders of a Therapeutic of the present invention may be assayed by any method known within the art. Such assays include in vitro cell proliferation assays, in vitro or in vivo assays using animal models of hyperproliferative diseases or disorders, or the like. Potentially effective Therapeutics may, for example, promote cell proliferation in culture or cause growth or cell proliferation in animal models in comparison to controls.


[0290] Specific embodiments of the present invention are directed to the treatment or prevention of cirrhosis of the liver (a condition in which scarring has overtaken normal liver regeneration processes); treatment of keloid (hypertrophic scar) formation causing disfiguring of the skin in which the scarring process interferes with normal renewal; psoriasis (a common skin condition characterized by excessive proliferation of the skin and delay in proper cell fate determination); benign tumors; fibrocystic conditions and tissue hypertrophy (e.g., benign prostatic hypertrophy).


[0291] Neurodegenerative Disorders


[0292] PROTX has been implicated in the deregulation of cellular maturation and apoptosis, which are both characteristic of neurodegenerative disease. Accordingly, Therapeutics of the invention, particularly but not limited to those that modulate (or supply) activity of an aforementioned protein, may be effective in treating or preventing neurodegenerative disease. Therapeutics of the present invention that modulate the activity of an aforementioned protein involved in neurodegenerative disorders can be assayed by any method known in the art for efficacy in treating or preventing such neurodegenerative diseases and disorders. Such assays include in vitro assays for regulated cell maturation or inhibition of apoptosis or in vivo assays using animal models of neurodegenerative diseases or disorders, or any of the assays described below. Potentially effective Therapeutics, for example but not by way of limitation, promote regulated cell maturation and prevent cell apoptosis in culture, or reduce neurodegeneration in animal models in comparison to controls.


[0293] Once a neurodegenerative disease or disorder has been shown to be amenable to treatment by modulation activity, that neurodegenerative disease or disorder can be treated or prevented by administration of a Therapeutic that modulates activity. Such diseases include all degenerative disorders involved with aging, especially osteoarthritis and neurodegenerative disorders.


[0294] Disorders Related to Organ Transplantation


[0295] PROTX has been implicated in disorders related to organ transplantation, in particular but not limited to organ rejection. Therapeutics of the invention, particularly those that modulate (or supply) activity, may be effective in treating or preventing diseases or disorders related to organ transplantation. Therapeutics of the invention (particularly Therapeutics that modulate the levels or activity of an aforementioned protein) can be assayed by any method known in the art for efficacy in treating or preventing such diseases and disorders related to organ transplantation. Such assays include in vitro assays for using cell culture models as described below, or in vivo assays using animal models of diseases and disorders related to organ transplantation, see e.g., below. Potentially effective Therapeutics, for example but not by way of limitation, reduce immune rejection responses in animal models in comparison to controls.


[0296] Accordingly, once diseases and disorders related to organ transplantation are shown to be amenable to treatment by modulation of activity, such diseases or disorders can be treated or prevented by administration of a Therapeutic that modulates activity.


[0297] Cardiovascular Disease


[0298] PROTX has been implicated in cardiovascular disorders, including in atherosclerotic plaque formation. Diseases such as cardiovascular disease, including cerebral thrombosis or hemorrhage, ischemic heart or renal disease, peripheral vascular disease, or thrombosis of other major vessel, and other diseases, including diabetes mellitus, hypertension, hypothyroidism, cholesterol ester storage disease, systemic lupus erythematosus, homocysteinemia, and familial protein or lipid processing diseases, and the like, are either directly or indirectly associated with atherosclerosis. Accordingly, Therapeutics of the invention, particularly those that modulate (or supply) activity or formation may be effective in treating or preventing atherosclerosis-associated diseases or disorders. Therapeutics of the invention (particularly Therapeutics that modulate the levels or activity) can be assayed by any method known in the art, including those described below, for efficacy in treating or preventing such diseases and disorders.


[0299] A vast array of animal and cell culture models exist for processes involved in atherosclerosis. A limited and non-exclusive list of animal models includes knockout mice for premature atherosclerosis (Kurabayashi and Yazaki, 1996, Int. Angiol. 15: 187-194), transgenic mouse models of atherosclerosis (Kappel et al., 1994, FASEB J. 8: 583-592), antisense oligonucleotide treatment of animal models (Callow, 1995, Curr. Opin. Cardiol. 10: 569-576), transgenic rabbit models for atherosclerosis (Taylor, 1997, Ann. N.Y. Acad. Sci 811: 146-152), hypercholesterolemic animal models (Rosenfeld, 1996, Diabetes Res. Clin. Pract. 30 Suppl.: 1-11), hyperlipidemic mice (Paigen et al., 1994, Curr. Opin. Lipidol. 5: 258-264), and inhibition of lipoxygenase in animals (Sigal et al., 1994, Ann. N.Y. Acad. Sci. 714: 211-224). In addition, in vitro cell models include but are not limited to monocytes exposed to low density lipoprotein (Frostegard et al., 1996, Atherosclerosis 121: 93-103), cloned vascular smooth muscle cells (Suttles et al., 1995, Exp. Cell Res. 218: 331-338), endothelial cell-derived chemoattractant exposed T cells (Katz et al., 1994, J. Leukoc. Biol. 55: 567-573), cultured human aortic endothelial cells (Farber et al., 1992, Am. J. Physiol. 262: H1088-1085), and foam cell cultures (Libby et al., 1996, Curr Opin Lipidol 7: 330-335). Potentially effective Therapeutics, for example but not by way of limitation, reduce foam cell formation in cell culture models, or reduce atherosclerotic plaque formation in hypercholesterolemic mouse models of atherosclerosis in comparison to controls.


[0300] Accordingly, once an atherosclerosis-associated disease or disorder has been shown to be amenable to treatment by modulation of activity or formation, that disease or disorder can be treated or prevented by administration of a Therapeutic that modulates activity.


[0301] Cytokine and Cell Proliferation/Differentiation Activity


[0302] A PROTX protein of the present invention may exhibit cytokine, cell proliferation (either inducing or inhibiting) or cell differentiation (either inducing or inhibiting) activity or may induce production of other cytokines in certain cell populations. Many protein factors discovered to date, including all known cytokines, have exhibited activity in one or more factor dependent cell proliferation assays, and hence the assays serve as a convenient confirmation of cytokine activity. The activity of a protein of the present invention is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DAIG, T10, B9, B9/11, BaF3, MC9/G, M+ (preB M+), 2E8, RB5, DA1, 123, T1165, HT2, CTLL2, TF-1, Mo7e and CMK.


[0303] The activity of a protein of the invention may, among other means, be measured by the following methods: Assays for T-cell or thymocyte proliferation include without limitation those described in: CURRENT PROTOCOLS IN IMMUNOLOGY, Ed by Coligan et al., Greene Publishing Associates and Wiley-Interscience (Chapter 3 and Chapter 7); Takai et al., J Immunol 137:3494-3500, 1986; Bertagnoili et al, J Immunol 145:1706-1712, 1990; Bertagnolli et al., Cell Immunol 133:327-341, 1991; Bertagnolli, et al., J Immunol 149:3778-3783, 1992; Bowman et al., J Immunol 152:1756-1761, 1994.


[0304] Assays for cytokine production and/or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described by Kruisbeek and Shevach, In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Vol 1, pp. 3.12.1-14, John Wiley and Sons, Toronto 1994; and by Schreiber, In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan eds., Vol 1, pp. 6.8.1-8, John Wiley and Sons, Toronto 1994.


[0305] Assays for proliferation and differentiation of hematopoietic and lymphopoietic cells include, without limitation, those described by Bottomly et al., In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Vol 1, pp. 6.3.1-6.3.12, John Wiley and Sons, Toronto 1991; deVries et al., J Exp Med 173:1205-1211, 1991; Moreau et al., Nature 336:690-692, 1988; Greenberger et al., Proc Natl Acad Sci U.S.A. 80:2931-2938, 1983; Nordan, In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Vol 1, pp. 6.6.1-5, John Wiley and Sons, Toronto 1991; Smith et al., Proc Natl Acad Sci U.S.A. 83:1857-1861, 1986; Measurement of human Interleukin 11-Bennett, et al. In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Vol 1, pp. 6.15.1 John Wiley and Sons, Toronto 1991; Ciarletta, et al., In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Vol 1, pp. 6.13.1, John Wiley and Sons, Toronto 1991.


[0306] Assays for T-cell clone responses to antigens (which will identify, among others, proteins that affect APC-T cell interactions as well as direct T-cell effects by measuring proliferation and cytokine production) include, without limitation, those described In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds., Greene Publishing Associates and Wiley-Interscience (Chapter 3, Chapter 6, Chapter 7); Weinberger et al, Proc Natl Acad Sci USA 77:6091-6095, 1980; Weinberger et al., Eur J Immun 11:405-411, 1981; Takai et al., J Immunol 137:3494-3500, 1986; Takai et al., J Immunol 140:508-512, 1988.


[0307] Immune Stimulating or Suppressing Activity


[0308] A PROTX protein of the present invention may also exhibit immune stimulating or immune suppressing activity, including without limitation the activities for which assays are described herein. A protein are useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency (SCID)), e.g., in regulating (up or down) growth and proliferation of T and/or B lymphocytes, as well as effecting the cytolytic activity of NK cells and other cell populations. These immune deficiencies may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders. More specifically, infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpesviruses, mycobacteria, Leishmania species, malaria species, and various fungal infections such as candidiasis. Of course, in this regard, a protein of the present invention may also be useful where a boost to the immune system generally may be desirable, i.e., in the treatment of cancer.


[0309] Autoimmune disorders which may be treated using a protein of the present invention include, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitus, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease. Such a protein of the present invention may also to be useful in the treatment of allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems. Other conditions, in which immune suppression is desired (including, for example, organ transplantation), may also be treatable using a protein of the present invention.


[0310] Using the proteins of the invention it may also be possible to immune responses, in a number of ways. Down regulation may be in the form of inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response. The functions of activated T cells may be inhibited by suppressing T cell responses or by inducing specific tolerance in T cells, or both. Immunosuppression of T cell responses is generally an active, non-antigen-specific, process which requires continuous exposure of the T cells to the suppressive agent. Tolerance, which involves inducing non-responsiveness or energy in T cells, is distinguishable from immunosuppression in that it is generally antigen-specific and persists after exposure to the tolerizing agent has ceased. Operationally, tolerance can be demonstrated by the lack of a T cell response upon re-exposure to specific antigen in the absence of the tolerizing agent.


[0311] Down regulating or preventing one or more antigen functions (including without limitation B lymphocyte antigen functions (such as, for example, B7), e.g., preventing high level lymphokine synthesis by activated T cells, are useful in situations of tissue, skin and organ transplantation and in graft-versus-host disease (GVHD). For example, blockage of T cell function should result in reduced tissue destruction in tissue transplantation. Typically, in tissue transplants, rejection of the transplant is initiated through its recognition as foreign by T cells, followed by an immune reaction that destroys the transplant. The administration of a molecule which inhibits or blocks interaction of a B7 lymphocyte antigen with its natural ligand(s) on immune cells (such as a soluble, monomeric form of a peptide having B7-2 activity alone or in conjunction with a monomeric form of a peptide having an activity of another B lymphocyte antigen (e.g., B7-1, B7-3) or blocking antibody), prior to transplantation can lead to the binding of the molecule to the natural ligand(s) on the immune cells without transmitting the corresponding costimulatory signal. Blocking B lymphocyte antigen function in this matter prevents cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant. Moreover, the lack of costimulation may also be sufficient to energize the T cells, thereby inducing tolerance in a subject. Induction of long-term tolerance by B lymphocyte antigen-blocking reagents may avoid the necessity of repeated administration of these blocking reagents. To achieve sufficient immunosuppression or tolerance in a subject, it may also be necessary to block the function of B lymphocyte antigens.


[0312] The efficacy of particular blocking reagents in preventing organ transplant rejection or GVHD can be assessed using animal models that are predictive of efficacy in humans. Examples of appropriate systems which can be used include allogeneic cardiac grafts in rats and xenogeneic pancreatic islet cell grafts in mice, both of which have been used to examine the immunosuppressive effects of CTLA4Ig fusion proteins in vivo as described in Lenschow et al., Science 257:789-792 (1992) and Turka et al., Proc Natl Acad Sci USA, 89:11102-11105 (1992). In addition, murine models of GVHD (see Paul ed., FUNDAMENTAL IMMUNOLOGY, Raven Press, New York, 1989, pp. 846-847) can be used to determine the effect of blocking B lymphocyte antigen function in vivo on the development of that disease.


[0313] Blocking antigen function may also be therapeutically useful for treating autoimmune diseases. Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against self tissue and which promote the production of cytokines and auto-antibodies involved in the pathology of the diseases. Preventing the activation of autoreactive T cells may reduce or eliminate disease symptoms. Administration of reagents which block costimulation of T cells by disrupting receptor:ligand interactions of B lymphocyte antigens can be used to inhibit T cell activation and prevent production of auto-antibodies or T cell-derived cytokines which may be involved in the disease process. Additionally, blocking reagents may induce antigen-specific tolerance of autoreactive T cells which could lead to long-term relief from the disease. The efficacy of blocking reagents in preventing or alleviating autoimmune disorders can be determined using a number of well-characterized animal models of human autoimmune diseases. Examples include murine experimental autoimmune encephalitis, systemic lupus erythematosis in MRL/lpr/lpr mice or NZB hybrid mice, murine autoimmune collagen arthritis, diabetes mellitus in NOD mice and BB rats, and murine experimental myasthenia gravis (see Paul ed., FUNDAMENTAL IMMUNOLOGY, Raven Press, New York, 1989, pp. 840-856).


[0314] Upregulation of an antigen function (preferably a B lymphocyte antigen function), as a means of up regulating immune responses, may also be useful in therapy. Upregulation of immune responses may be in the form of enhancing an existing immune response or eliciting an initial immune response. For example, enhancing an immune response through stimulating B lymphocyte antigen function are useful in cases of viral infection. In addition, systemic viral diseases such as influenza, the common cold, and encephalitis might be alleviated by the administration of stimulatory forms of B lymphocyte antigens systemically.


[0315] Alternatively, anti-viral immune responses may be enhanced in an infected patient by removing T cells from the patient, costimulating the T cells in vitro with viral antigen-pulsed APCs either expressing a peptide of the present invention or together with a stimulatory form of a soluble peptide of the present invention and reintroducing the in vitro activated T cells into the patient. Another method of enhancing anti-viral immune responses would be to isolate infected cells from a patient, transfect them with a nucleic acid encoding a protein of the present invention as described herein such that the cells express all or a portion of the protein on their surface, and reintroduce the transfected cells into the patient. The infected cells would now be capable of delivering a costimulatory signal to, and thereby activate, T cells in vivo.


[0316] In another application, up regulation or enhancement of antigen function (preferably B lymphocyte antigen function) are useful in the induction of tumor immunity. Tumor cells (e.g., sarcoma, melanoma, lymphoma, leukemia, neuroblastoma, carcinoma) transfected with a nucleic acid encoding at least one peptide of the present invention can be administered to a subject to overcome tumor-specific tolerance in the subject. If desired, the tumor cell can be transfected to express a combination of peptides. For example, tumor cells obtained from a patient can be transfected ex vivo with an expression vector directing the expression of a peptide having B7-2-like activity alone, or in conjunction with a peptide having B7-1-like activity and/or B7-3-like activity. The transfected tumor cells are returned to the patient to result in expression of the peptides on the surface of the transfected cell. Alternatively, gene therapy techniques can be used to target a tumor cell for transfection in vivo.


[0317] The presence of the peptide of the present invention having the activity of a B lymphocyte antigen(s) on the surface of the tumor cell provides the necessary costimulation signal to T cells to induce a T cell mediated immune response against the transfected tumor cells. In addition, tumor cells which lack MHC class I or MHC class II molecules, or which fail to reexpress sufficient amounts of MHC class I or MHC class II molecules, can be transfected with nucleic acid encoding all or a portion of (e.g., a cytoplasmic-domain truncated portion) of an MHC class I a chain protein and b 2 microglobulin protein or an MHC class II a chain protein and an MHC class II b chain protein to thereby express MHC class I or MHC class II proteins on the cell surface. Expression of the appropriate class I or class II MHC in conjunction with a peptide having the activity of a B lymphocyte antigen (e.g., B7-1, B7-2, B7-3) induces a T cell mediated immune response against the transfected tumor cell. Optionally, a gene encoding an antisense construct which blocks expression of an MHC class II associated protein, such as the invariant chain, can also be cotransfected with a DNA encoding a peptide having the activity of a B lymphocyte antigen to promote presentation of tumor associated antigens and induce tumor specific immunity. Thus, the induction of a T cell mediated immune response in a human subject may be sufficient to overcome tumor-specific tolerance in the subject.


[0318] The activity of a protein of the invention may, among other means, be measured by the following methods: Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds. Greene Publishing Associates and Wiley-Interscience (Chapter 3, Chapter 7); Herrmann et al., Proc Natl Acad Sci USA 78:2488-2492, 1981; Herrmann et al., J Immunol 128:1968-1974, 1982; Handa et al., J Immunol 135:1564-1572, 1985; Takai et al., J Immunol 137:3494-3500, 1986; Takai et al., J Immunol 140:508-512, 1988; Herrmann et al., Proc Natl Acad Sci USA 78:2488-2492, 1981; Herrmann et al., J Immunol 128:1968-1974,1982; Handa et al., J Immunol 135:1564-1572, 1985; Takai et al., J Immunol 137:3494-3500, 1986; Bowman et al., J Virology 61:1992-1998; Takai et al., J Immunol 140:508-512, 1988; Bertagnolli et al., Cell Immunol 133:327-341, 1991; Brown et al., J Immunol 153:3079-3092, 1994.


[0319] Assays for T-cell-dependent immunoglobulin responses and isotype switching (which will identify, among others, proteins that modulate T-cell dependent antibody responses and that affect Th1/Th2 profiles) include, without limitation, those described in: Maliszewski, J Immunol 144:3028-3033, 1990; and Mond and Brunswick In: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan etal., eds., Vol 1, pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto 1994.


[0320] Mixed lymphocyte reaction (MLR) assays (which will identify, among others, proteins that generate predominantly Thl and CTL responses) include, without limitation, those described in: CURRENT PROTOCOLS IN IMMUNOLOGY. Coligan et al., eds. Greene Publishing Associates and Wiley-Interscience (Chapter 3, Chapter 7); Takai et al., J Immunol 137:3494-3500, 1986; Takai et al., J Immunol 140:508-512, 1988; Bertagnolli et al., J Immunol 149:3778-3783, 1992.


[0321] Dendritic cell-dependent assays (which will identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J Immunol 134:536-544, 1995; Inaba et al., J Exp Med 173:549-559, 1991; Macatonia et al., J Immunol 154:5071-5079, 1995; Porgador et al., J Exp Med 182:255-260, 1995; Nair et al., J Virol 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., J Exp Med 169:1255-1264, 1989; Bhardwaj et al., J Clin Investig 94:797-807, 1994; and Inaba et al., J Exp Med 172:631-640, 1990.


[0322] Assays for lymphocyte survival/apoptosis (which will identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis) include, without limitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Res 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, J Immunol 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., Internat J Oncol 1:639-648, 1992.


[0323] Assays for proteins that influence early steps of T-cell commitment and development include, without limitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., Cell Immunol 155: 111-122, 1994; Galy et al., Blood 85:2770-2778, 1995; Toki et al., Proc Nat Acad Sci USA 88:7548-7551, 1991.


[0324] Hematopoiesis Regulating Activity


[0325] A PROTX protein of the present invention are useful in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell deficiencies. Even marginal biological activity in support of colony forming cells or of factor-dependent cell lines indicates involvement in regulating hematopoiesis, e.g. in supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines, thereby indicating utility, for example, in treating various anemias or for use in conjunction with irradiation/chemotherapy to stimulate the production of erythroid precursors and/or erythroid cells; in supporting the growth and proliferation of myeloid cells such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo-suppression; in supporting the growth and proliferation of megakaryocytes and consequently of platelets thereby allowing prevention or treatment of various platelet disorders such as thrombocytopenia, and generally for use in place of or complimentary to platelet transfusions; and/or in supporting the growth and proliferation of hematopoietic stem cells which are capable of maturing to any and all of the above-mentioned hematopoietic cells and therefore find therapeutic utility in various stem cell disorders (such as those usually treated with transplantation, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem cell compartment post irradiation/chemotherapy, either in-vivo or ex-vivo (i.e., in conjunction with bone marrow transplantation or with peripheral progenitor cell transplantation (homologous or heterologous)) as normal cells or genetically manipulated for gene therapy.


[0326] The activity of a protein of the invention may, among other means, be measured by the following methods:


[0327] Suitable assays for proliferation and differentiation of various hematopoietic lines are cited above.


[0328] Assays for embryonic stem cell differentiation (which will identify, among others, proteins that influence embryonic differentiation hematopoiesis) include, without limitation, those described in: Johansson et al. Cellular Biology 15:141-151, 1995; Keller et al., Molecular and Cellular Biology 13:473-486, 1993; McClanahan et al., Blood 81:2903-2915, 1993.


[0329] Assays for stem cell survival and differentiation (which will identify, among others, proteins that regulate lympho-hematopoiesis) include, without limitation, those described in: Methylcellulose colony forming assays, Freshney, M. G. In CULTURE OF HEMATOPOIETIC CELLS. Freshney, et al. eds. Vol pp. 265-268, Wiley-Liss, Inc., New York, N.Y 1994; Hirayama et al., Proc Natl Acad Sci USA 89:5907-5911, 1992; McNiece and Briddeli. In CULTURE OF HEMATOPOIETIC CELLS. Freshney, et al eds. Vol pp. 23-39, Wiley-Liss, Inc., New York, N.Y. 1994; Neben et al., Exp Hematol 22:353-359, 1994; Ploemacher In CULTURE OF HEMATOPOIETIC CELLS. Freshney, et al. eds. Vol pp. 1-21, Wiley-Liss, Inc., New York, N.Y. 1994; Spoonceret al., In CULTURE OF HEMATOPOIETIC CELLS. Freshhey, et al. eds. Vol pp. 163-179, Wiley-Liss, Inc., New York, N.Y. 1994; Sutherland, In CULTURE OF HEMATOPOIETIC CELLS. Freshney, et al. eds. Vol pp. 139-162, Wiley-Liss, Inc., New York, N.Y. 1994.


[0330] Tissue Growth Activity


[0331] A PROTX protein of the present invention also may have utility in compositions used for bone, cartilage, tendon, ligament and/or nerve tissue growth or regeneration, as well as for wound healing and tissue repair and replacement, and in the treatment of burns, incisions and ulcers.


[0332] A protein of the present invention, which induces cartilage and/or bone growth in circumstances where bone is not normally formed, has application in the healing of bone fractures and cartilage damage or defects in humans and other animals. Such a preparation employing a protein of the invention may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints. De novo bone formation induced by an osteogenic agent contributes to the repair of congenital, trauma induced, or oncologic resection induced craniofacial defects, and also is useful in cosmetic plastic surgery.


[0333] A protein of this invention may also be used in the treatment of periodontal disease, and in other tooth repair processes. Such agents may provide an environment to attract bone-forming cells, stimulate growth of bone-forming cells or induce differentiation of progenitors of bone-forming cells. A protein of the invention may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and/or cartilage repair or by blocking inflammation or processes of tissue destruction (collagenase activity, osteoclast activity, etc.) mediated by inflammatory processes.


[0334] Another category of tissue regeneration activity that may be attributable to the protein of the present invention is tendon/ligament formation. A protein of the present invention, which induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normally formed, has application in the healing of tendon or ligament tears, deformities and other tendon or ligament defects in humans and other animals. Such a preparation employing a tendon/ligament-like tissue inducing protein may have prophylactic use in preventing damage to tendon or ligament tissue, as well as use in the improved fixation of tendon or ligament to bone or other tissues, and in repairing defects to tendon or ligament tissue. De novo tendon/ligament-like tissue formation induced by a composition of the present invention contributes to the repair of congenital, trauma induced, or other tendon or ligament defects of other origin, and is also useful in cosmetic plastic surgery for attachment or repair of tendons or ligaments. The compositions of the present invention may provide an environment to attract tendon- or ligament-forming cells, stimulate growth of tendon- or ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/ligament cells or progenitors ex vivo for return in vivo to effect tissue repair. The compositions of the invention may also be useful in the treatment of tendonitis, carpal tunnel syndrome and other tendon or ligament defects. The compositions may also include an appropriate matrix and/or sequestering agent as a career as is well known in the art.


[0335] The protein of the present invention may also be useful for proliferation of neural cells and for regeneration of nerve and brain tissue, i.e. for the treatment of central and peripheral nervous system diseases and neuropathies, as well as mechanical and traumatic disorders, which involve degeneration, death or trauma to neural cells or nerve tissue. More specifically, a protein may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome. Further conditions which may be treated in accordance with the present invention include mechanical and traumatic disorders, such as spinal cord disorders, head trauma and cerebrovascular diseases such as stroke. Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a protein of the invention.


[0336] Proteins of the invention may also be useful to promote better or faster closure of non-healing wounds, including without limitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.


[0337] It is expected that a protein of the present invention may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth, skeletal or cardiac) and vascular (including vascular endothelium) tissue, or for promoting the growth of cells comprising such tissues. Part of the desired effects may be by inhibition or modulation of fibrotic scarring to allow normal tissue to regenerate. A protein of the invention may also exhibit angiogenic activity.


[0338] A protein of the present invention may also be useful for gut protection or regeneration and treatment of lung or liver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage.


[0339] A protein of the present invention may also be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or cells; or for inhibiting the growth of tissues described above.


[0340] The activity of a protein of the invention may, among other means, be measured by the following methods:


[0341] Assays for tissue generation activity include, without limitation, those described in: International Patent Publication No. WO95/16035 (bone, cartilage, tendon); International Patent Publication No. WO95/05846 (nerve, neuronal); International Patent Publication No. WO91/07491 (skin, endothelium).


[0342] Assays for wound healing activity include, without limitation, those described in: Winter, Epidermal Wound Healing, pp. 71-112 (Maibach, H I and Rovee, D T, eds.), Year Book Medical Publishers, Inc., Chicago, as modified by Eaglstein and Menz, J. Invest. Dermatol 71:382-84 (1978).


[0343] Activin/Inhibin Activity


[0344] A PROTX protein of the present invention may also exhibit activin- or inhibin-related activities. Inhibins are characterized by their ability to inhibit the release of follicle stimulating hormone (FSH), while activins and are characterized by their ability to stimulate the release of follicle stimulating hormone (FSH). Thus, a protein of the present invention, alone or in heterodimers with a member of the inhibin a family, are useful as a contraceptive based on the ability of inhibins to decrease fertility in female mammals and decrease spermatogenesis in male mammals. Administration of sufficient amounts of other inhibins can induce infertility in these mammals. Alternatively, the protein of the invention, as a homodimer or as a heterodimer with other protein subunits of the inhibin-b group, are useful as a fertility inducing therapeutic, based upon the ability of activin molecules in stimulating FSH release from cells of the anterior pituitary. See, for example, U.S. Pat. No. 4,798,885. A protein of the invention may also be useful for advancement of the onset of fertility in sexually immature mammals, so as to increase the lifetime reproductive performance of domestic animals such as cows, sheep and pigs.


[0345] The activity of a protein of the invention may, among other means, be measured by the following methods:


[0346] Assays for activin/inhibin activity include, without limitation, those described in: Vale et al., Endocrinology 91:562-572, 1972; Ling et al., Nature 321:779-782, 1986; Vale et al., Nature 321:776-779, 1986; Mason et al., Nature 318:659-663, 1985; Forage et al., Proc Natl Acad Sci USA 83:3091-3095, 1986.


[0347] Chemotactic/Chemokinetic Activity


[0348] A protein of the present invention may have chemotactic or chemokinetic activity (e.g., act as a chemokine) for mammalian cells, including, for example, monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells. Chemotactic and chemokinetic proteins can be used to mobilize or attract a desired cell population to a desired site of action. Chemotactic or chemokinetic proteins provide particular advantages in treatment of wounds and other trauma to tissues, as well as in treatment of localized infections. For example, attraction of lymphocytes, monocytes or neutrophils to tumors or sites of infection may result in improved immune responses against the tumor or infecting agent.


[0349] A protein or peptide has chemotactic activity for a particular cell population if it can stimulate, directly or indirectly, the directed orientation or movement of such cell population. Preferably, the protein or peptide has the ability to directly stimulate directed movement of cells. Whether a particular protein has chemotactic activity for a population of cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.


[0350] The activity of a protein of the invention may, among other means, be measured by following methods:


[0351] Assays for chemotactic activity (which will identify proteins that induce or prevent chemotaxis) consist of assays that measure the ability of a protein to induce the migration of cells across a membrane as well as the ability of a protein to induce the adhesion of one cell population to another cell population. Suitable assays for movement and adhesion include, without limitation, those described in: CURRENT PROTOCOLS IN IMMUNOLOGY, Coligan et al., eds. (Chapter 6.12, Measurement of alpha and beta Chemokines 6.12.1-6.12.28); Taub et al. J Clin Invest 95:1370-1376, 1995; Lind et al. APMIS 103:140-146, 1995; Muller et al Eur J Immunol 25: 1744-1748; Gruberet al. J Immunol 152:5860-5867, 1994; Johnston et al. J Immunol 153: 1762-1768, 1994.


[0352] Hemostatic and Thrombolytic Activity


[0353] A protein of the invention may also exhibit hemostatic or thrombolytic activity. As a result, such a protein is expected to be useful in treatment of various coagulation disorders (including hereditary disorders, such as hemophilias) or to enhance coagulation and other hemostatic events in treating wounds resulting from trauma, surgery or other causes. A protein of the invention may also be useful for dissolving or inhibiting formation of thromboses and for treatment and prevention of conditions resulting therefrom (such as, for example, infarction of cardiac and central nervous system vessels (e.g., stroke).


[0354] The activity of a protein of the invention may, among other means, be measured by the following methods:


[0355] Assay for hemostatic and thrombolytic activity include, without limitation, those described in: Linet et al., J. Clin. Pharmacol. 26:131-140, 1986; Burdick et al., Thrombosis Res. 45:413-419, 1987; Humphrey et al., Fibrinolysis 5:71-79 (1991); Schaub, Prostaglandins 35:467-474, 1988.


[0356] Receptor/Ligand Activity


[0357] A protein of the present invention may also demonstrate activity as receptors, receptor ligands or inhibitors or agonists of receptor/ligand interactions. Examples of such receptors and ligands include, without limitation, cytokine receptors and their ligands, receptor kinases and their ligands, receptor phosphatases and their ligands, receptors involved in cell—cell interactions and their ligands (including without limitation, cellular adhesion molecules (such as selectins, integrins and their ligands) and receptor/ligand pairs involved in antigen presentation, antigen recognition and development of cellular and humoral immune responses). Receptors and ligands are also useful for screening of potential peptide or small molecule inhibitors of the relevant receptor/ligand interaction. A protein of the present invention (including, without limitation, fragments of receptors and ligands) may themselves be useful as inhibitors of receptor/ligand interactions.


[0358] The activity of a protein of the invention may, among other means, be measured by the following methods:


[0359] Suitable assays for receptor-ligand activity include without limitation those described in: Current Protocols in Immunology, Ed by J. E. coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 7.28, Measurement of Cellular Adhesion under static conditions 7.28.1-7.28.22), Takai et al., Proc Natl Acad Sci USA 84:6864-6868, 1987; Bierer et al., J. Exp. Med. 168:1145-1156, 1988; Rosenstein et al., J. Exp. Med. 169:149-160 1989; Stoltenborg et al., J Immunol Methods 175:59-68, 1994; Stitt et al., Cell 80:661-670, 1995.


[0360] Anti-inflammatory Activity


[0361] Proteins of the present invention may also exhibit anti-inflammatory activity. The anti-inflammatory activity may be achieved by providing a stimulus to cells involved in the inflammatory response, by inhibiting or promoting cell—cell interactions (such as, for example, cell adhesion), by inhibiting or promoting chemotaxis of cells involved in the inflammatory process, inhibiting or promoting cell extravasation, or by stimulating or suppressing production of other factors which more directly inhibit or promote an inflammatory response. Proteins exhibiting such activities can be used to treat inflammatory conditions including chronic or acute conditions), including without limitation inflammation associated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine-induced lung injury, inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1. Proteins of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material.


[0362] Tumor Inhibition Activity


[0363] In addition to the activities described above for immunological treatment or prevention of tumors, a protein of the invention may exhibit other anti-tumor activities. A protein may inhibit tumor growth directly or indirectly (such as, for example, via ADCC). A protein may exhibit its tumor inhibitory activity by acting on tumor tissue or tumor precursor tissue, by inhibiting formation of tissues necessary to support tumor growth (such as, for example, by inhibiting angiogenesis), by causing production of other factors, agents or cell types which inhibit tumor growth, or by suppressing, eliminating or inhibiting factors, agents or cell types which promote tumor growth.


[0364] Other Activities


[0365] A protein of the invention may also exhibit one or more of the following additional activities or effects: inhibiting the growth, infection or function of, or killing, infectious agents, including, without limitation, bacteria, viruses, fungi and other parasites; effecting (suppressing or enhancing) bodily characteristics, including, without limitation, height, weight, hair color, eye color, skin, fat to lean ratio or other tissue pigmentation, or organ or body part size or shape (such as, for example, breast augmentation or diminution, change in bone form or shape); effecting biorhythms or circadian cycles or rhythms; effecting the fertility of male or female subjects; effecting the metabolism, catabolism, anabolism, processing, utilization, storage or elimination of dietary fat, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional factors or component(s); effecting behavioral characteristics, including, without limitation, appetite, libido, stress, cognition (including cognitive disorders), depression (including depressive disorders) and violent behaviors; providing analgesic effects or other pain reducing effects; promoting differentiation and growth of embryonic stem cells in lineages other than hematopoietic lineages; hormonal or endocrine activity; in the case of enzymes, correcting deficiencies of the enzyme and treating deficiency-related diseases; treatment of hyperproliferative disorders (such as, for example, psoriasis); immunoglobulin-like activity (such as, for example, the ability to bind antigens or complement); and the ability to act as an antigen in a vaccine composition to raise an immune response against such protein or another material or entity which is cross-reactive with such protein.


[0366] The invention will be further illustrated in the following non-limiting examples.



EXAMPLES


Example 1


Quantitative Expression Analysis of PROTX Nucleic Acids

[0367] The quantitative expression of various clones was assessed in about 40 normal and about 54 tumor samples (the samples are identified in the Tables below) by real time quantitative PCR (TAQMAN®) performed on a Perkin-Elmer Biosystems ABI PRISM® 7700 Sequence Detection System.


[0368] First, 96 RNA samples were normalized to P-actin and GAPDH. RNA (˜50 ng total or ˜1 ng polyA+) was converted to cDNA using the TAQMAN® Reverse Transcription Reagents Kit (PE Biosystems, Foster City, Calif.; cat # N808-0234) and random hexamers according to the manufacturer's protocol. Reactions were performed in 20 ul and incubated for 30 min. at 48° C. cDNA (5 ul) was then transferred to a separate plate for the TAQMAN® reaction using β-actin and GAPDH TAQMAN® Assay Reagents (PE Biosystems; cat. #'s 4310881E and 4310884E, respectively) and TAQMAN® universal PCR Master Mix (PE Biosystems; cat # 4304447) according to the manufacturer's protocol. Reactions were performed in 25 ul using the following parameters: 2 min. at 50° C.; 10 min. at 95° C.; 15 sec. at 95° C./1 min. at 60° C. (40 cycles). Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being represented as 2 to the power of delta CT. The percent relative expression is then obtained by taking the reciprocal of this RNA difference and multiplying by 100. The average CT values obtained for p-actin and GAPDH were used to normalize RNA samples. The RNA sample generating the highest CT value required no further diluting, while all other samples were diluted relative to this sample according to their β-actin/GAPDH average CT values.


[0369] Normalized RNA (5 ul) was converted to cDNA and analyzed via TAQMAN® using One Step RT-PCR Master Mix Reagents (PE Biosystems; cat. # 4309169) and gene-specific primers according to the manufacturer's instructions. Probes and primers were designed for each assay according to Perkin Elmer Biosystem's Primer Express Software package (version I for Apple Computer's Macintosh Power PC) using the sequence of the subject clone as input. Default settings were used for reaction conditions and the following parameters were set before selecting primers: primer concentration=250 nM, primer melting temperature (Tm) range=58°-60° C., primer optimal Tm=59° C., maximum primer difference=2° C., probe does not have 5′ G, probe Tm must be 10° C. greater than primer Tm, amplicon size 75 bp to 100 bp. The probes and primers selected (see below) were synthesized by Synthegen (Houston, Tex., USA). Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5′ and 3′ ends of the probe, respectively. Their final concentrations were: forward and reverse primers, 900 nM each, and probe, 200nM.


[0370] PCR conditions: Normalized RNA from each tissue and each cell line was spotted in each well of a 96 well PCR plate (Perkin Elmer Biosystems). PCR cocktails including two probes (PROTX-specific and another gene-specific probe multiplexed with the PROTX probe) were set up using 1× TaqMan™ PCR Master Mix for the PE Biosystems 7700, with 5 mM MgCl2, dNTPs (dA, G, C, U at 1:1:1:2 ratios), 0.25 U/ml AmpliTaq Gold™ (PE Biosystems), and 0.4 U/l RNase inhibitor, and 0.25 U/μl reverse transcriptase. Reverse transcription was performed at 48° C. for 30 minutes followed by amplification/PCR cycles as follows: 95° C. 10 min, then 40 cycles of 95° C. for 15 seconds, 60° C. for 1 minute.
18TABLE 2Clone Identification No: AL132780_A (PROT1)Probe Name: Ag2260SEQ IDPrimersSequencesNOForward5′-CAAAATCAAGTGACCACAGACA-3′35ProbeFAM-5′-36ATTTCTCAGCTGCCTCCTGAAACCCT-3′-TAMRAReverse5′-TCAATGGCTCTGTGTACTGTGA-3′37


[0371]

19





TABLE 3










Expression Data for Clone No. AL132780_A (PROT1)











% Relative

% Relative


Tissue_Name/Run_Name
Expression
Tissue_Name/Run_Name
Expression













Liver adenocarcinoma
33.92
Normal Colon GENPAK 061003
10.15


Heart (fetal)
50
83219 CC Well to Mod Diff(ODO3866)
7.38


Pancreas
6
83220 CC NAT (ODO3866)
1.78


Pancreatic ca. CAPAN 2
14.97
83221 CC Gr.2 rectosigmoid (ODO3868)
7.08


Adrenal gland
1.46
83222 CC NAT (ODO3868)
0.38


Thyroid
15.07
83235 CC Mod Diff(ODO3920)
6.98


Salivary gland
3.19
83236 CC NAT (ODO3920)
1.72


Pituitary gland
0.89
83237 CC Gr.2 ascend colon (ODO3921)
10.88


Brain (fetal)
0.41
83238 CC NAT (ODO3921)
3.9


Brain (whole)
0.68
83241 CC from Partial Hepatectomy (ODO4309)
20.73


Brain (amygdala)
1
83242 Liver NAT (ODO4309)
61.99


Brain (cerebellum)
0.43
87472 Colon mets to lung (OD04451-01)
11.03


Brain (hippocampus)
2.92
87473 Lung NAT (OD04451-02)
14.56


Brain (thalamus)
1.14
Normal Prostate Clontech A+ 6546-1
23.33


Cerebral Cortex
1.27
84140 Prostate Cancer (OD04410)
10.73


Spinal cord
3.12
84141 Prostate NAT (OD04410)
17.56


CNS ca. (glio/astro) U87-MG
5.91
87073 Prostate Cancer (OD04720-01)
25.88


CNS ca. (glio/astro) U-118-MG
37.11
87074 Prostate NAT (OD04720-02)
38.96


CNS ca. (astro) SW1783
24.66
Normal Lung GENPAK 061010
36.1


CNS ca.* (neuro; met) SK-N-AS
6.16
83239 Lung Met to Muscle (ODO4286)
20.59


CNS ca. (astro) SF-539
20.31
83240 Muscle NAT (ODO4286)
2.22


CNS ca. (astro) SNB-75
34.39
84136 Lung Malignant Cancer (OD03126)
14.56


CNS ca. (glio) SNB-19
10.15
84137 Lung NAT (OD03126)
41.47


CNS ca. (glio) U251
18.95
84871 Lung Cancer (OD04404)
62.85


CNS ca. (glio) SF-295
23
84872 Lung NAT (OD04404)
18.3


Heart
2.06
84875 Lung Cancer (OD04565)
27.93


Skeletal muscle
0
85950 Lung Cancer (OD04237-01)
40.05


Bone marrow
0.03
85970 Lung NAT (OD04237-02)
15.39


Thymus
3.54
83255 Ocular Mel Met to Liver (ODO4310)
7.86


Spleen
2.13
83256 Liver NAT (ODO4310)
37.89


Lymph node
1.63
84139 Melanoma Mets to Lung (OD04321)
8.48


Colorectal
1.12
84138 Lung NAT (OD04321)
39.5


Stomach
5.08
Normal Kidney GENPAK 061008
72.2


Small intestine
2.61
83786 Kidney Ca, Nuclear grade 2 (OD04338)
52.12


Colon ca. SW480
27.36
83787 Kidney NAT (OD04338)
36.86


Colon ca.* (SW480 met)SW620
13.58
83788 Kidney Ca Nuclear grade 1/2 (OD04339)
21.46


Colon ca. HT29
9.67
83789 Kidney NAT (OD04339)
58.24


Colon ca. HCT-116
12.24
83790 Kidney Ca, Clear cell type (OD04340)
100


Colon ca. CaCo-2
66.9
83791 Kidney NAT (OD04340)
31.43


83219 CC Well to Mod Diff
5.22
83792 Kidney Ca, Nuclear grade 3 (OD04348)
13.68


(ODO3866)


Colon ca. HCC-2998
20.45
83793 Kidney NAT (OD04348)
40.9


Gastric ca.* (liver met) NCI-N87
22.22
87474 Kidney Cancer (OD04622-01)
5.95


Bladder
5.01
87475 Kidney NAT (OD04622-03)
24.15


Trachea
13.68
85973 Kidney Cancer (OD04450-01)
44.75


Kidney
6.7
85974 Kidney NAT (OD04450-03)
33.22


Kidney (fetal)
17.56
Kidney Cancer Clontech 8120607
26.06


Renal ca. 786-0
45.69
Kidney NAT Clontech 8120608
22.07


Renal ca. A498
36.6
Kidney Cancer Clontech 8120613
6.38


Renal ca. RXF 393
15.5
Kidney NAT Clontech 8120614
49.65


Renal ca. ACHN
59.87
Kidney Cancer Clontech 9010320
25.17


Renal ca. UO-31
77.38
Kidney NAT Clontech 9010321
73.71


Renal ca. TK-10
42.63
Normal Uterus GENPAK 061018
4.64


Liver
4.04
Uterus Cancer GENPAK 064011
14.66


Liver (fetal)
9.21
Normal Thyroid Clontech A+ 6570-1
17.31


Liver ca. (hepatoblast) HepG2
71.2
Thyroid Cancer GENPAK 064010
33.68


Lung
12.67
Thyroid Cancer INVITROGEN A302152
21.92


Lung (fetal)
20.73
Thyroid NAT INVITROGEN A302153
22.53


Lung ca. (small cell) LX-1
22.38
Normal Breast GENPAK 061019
23.33


Lung ca. (small cell) NCI-H69
5.75
84877 Breast Cancer (OD04566)
10.66


Lung ca. (s.cell var.) SHP-77
2.61
85975 Breast Cancer (OD04590-01)
5.95


Lung ca. (large cell)NCI-H460
10.37
85976 Breast Cancer Mets (OD04590-03)
5.29


Lung ca. (non-sm. cell) A549
10.66
87070 Breast Cancer Metastasis (OD04655-05)
2.24


Lung ca. (non-s.cell) NCI-H23
30.57
GENPAK Breast Cancer 064006
7.43


Lung ca (non-s.cell) HOP-62
28.92
Breast Cancer Clontech 9100266
18.05


Lung ca. (non-s.cl) NCI-H522
52.49
Breast NAT Clontech 9100265
25.7


Lung ca. (squam.) SW 900
20.03
Breast Cancer INVITROGEN A209073
24.15


Lung ca. (squam.) NCI-H596
0.26
Breast NAT INVITROGEN A2090734
23.98


Mammary gland
9.94
Normal Liver GENPAK 061009
12.07


Breast ca.* (pl. effusion) MCF-7
12.16
Liver Cancer GENPAK 064003
39.5


Breast ca.* (pl.ef) MDA-MB-231
88.88
Liver Cancer Research Genetics RNA 1025
15.07


Breast ca.* (pl. effusion) T47D
23.82
Liver Cancer Research Genetics RNA 1026
18.56


Breast ca. BT-549
23.65
Paired Liver Cancer Tissue Research Genetics
20.88




RNA 6004-T


Breast ca. MDA-N
2.59
Paired Liver Tissue Research Genetics RNA 6004-N
7.18


Ovary
18.05
Paired Liver Cancer Tissue Research Genetics
14.16




RNA 6005-T


Ovarian ca. OVCAR-3
24.32
Paired Liver Tissue Research Genetics RNA 6005-N
9.94


Ovarian ca. OVCAR-4
9.41
Normal Bladder GENPAK 061001
35.11


Ovarian ca. OVCAR-5
40.33
Bladder Cancer Research Genetics RNA 1023
7.43


Ovarian ca. OVCAR-8
100
Bladder Cancer INVITROGEN A302173
19.08


Ovarian ca. IGROV-1
16.04
87071 Bladder Cancer (OD04718-01)
28.72


Ovarian ca.* (ascites) SK-OV-3
41.18
87072 Bladder Normal Adjacent (OD04718-03)
2.66


Uterus
12.59
Normal Ovary Res. Gen.
14.06


Plancenta
13.03
Ovarian Cancer GENPAK 064008
20.31


Prostate
11.58
87492 Ovary Cancer (OD04768-07)
60.29


Prostate ca.* (bone met)PC-3
39.78
87493 Ovary NAT (OD04768-08)
4.74


Testis
10.15
Normal Stomach GENPAK 061017
4.04


Melanoma Hs688(A).T
47.96
NAT Stomach Clontech 9060359
4.54


Melanoma* (met) Hs688(B).T
45.38
Gastric Cancer Clontech 9060395
11.91


Melanoma UACC-62
1.5
NAT Stomach Clontech 9060394
9.02


Melanoma M14
1.07
Gastric Cancer Clontech 9060397
45.69


Melanoma LOX IMVI
2.63
NAT Stomach Clontech 9060396
2.45


Melanoma* (met) SK-MEL-5
4.9
Gastric Cancer GENPAK 064005
20.03


Adipose
2.63






ca. = carcinoma




*= established from metastasis




met = metastasis




s cell var = small cell variant




non-s = non-sm = non-small




squam = squamous




pl. eff = pl effusion = pleural effusion




glio = glioma




astro = astrocytoma




neuro = neuroblastoma








[0372] It is seen from the Table above that clone AL132780_A is expressed in most normal and cancer cells assayed. It is especially prominent in 83790 Kidney Ca, Clear cell type (OD04340) and Ovarian ca. OVCAR-8. This observation indicates that the clone of PROTI has a role in cell proliferation and potential utility as a marker to identify and/or stage tumors. It may furthermore be a target for a specific monoclonal antibody that could be used to treat various cancers, especially kidney cancer and ovarian cancer.
20TABLE 4Clone Identification No: AC016900_A (PROT2)Probe Name: Ag382SEQ IDPrimersSequencesNOForward5′-GGAGGTGGACCCCAATATCC-3′38ProbeTET-5′-ATCTGCTCCTCCTGGGTGCGCA-3′-39TAMRAReverse5′-CGACGAAGGAGGCAAACTTG-3′40


[0373]

21





TABLE 5










Expression Data for Clone AC016900_A













% Rel.
% Rel.

% Rel.
% Rel.


Tissue_Name/Run_Name
Expr.
Expr.
Tissue_Name/Run_Name
Expr.
Expr.















Adipose
0
2.98
Normal Colon GENPAK 061003
28.92
7.33


Adrenal gland
0
0.11
83219 CC Well to Mod Diff
0
33.92





(ODO3866)


Bladder
33.45
34.87
83220 CC NAT (ODO3866)
19.61
36.35


Brain (amygdala)
0
0
83221 CC Gr.2 rectosigmoid
20.45
40.61





(ODO3868)


Brain (cerebellum)
0
0
83222 CC NAT (ODO3868)
1.51
4.27


Brain (hippocampus)
0
0
83235 CC Mod Diff (ODO3920)
46.98
100


Brain (substantia nigra)
0
0
83236 CC NAT (ODO3920)
14.26
38.69


Brain (thalamus)
0
0
83237 CC Gr.2 ascend colon
24.49
88.27





(ODO3921)


Cerebral Cortex
0
0
83238 CC NAT (ODO3921)
3.28
8.08


Brain (fetal)
0
0
83241 CC from Partial Hepatectomy
14.86
85.26





(ODO4309)


Brain (whole)
0
0
83242 Liver NAT (ODO4309)
6.43
11.66


CNS ca. (glio/astro) U-118-
0
0
87472 Colon mets to lung (OD04451-
16.84
16.96


MG


01)


CNS ca. (astro) SF-539
0
0
87473 Lung NAT (OD04451-02)
3.82
5.11


CNS ca. (astro) SNB-
0
0
Normal Prostate Clontech A+ 6546-
1.39
2.76


75


1


CNS ca. (astro)
0
0
84140 Prostate Cancer (OD04410)
7.97
45.38


SW1783


CNS ca. (glio)
0.01
0.89
84141 Prostate NAT (OD04410)
3.77
6.47


U251


CNS ca. (glio) SF-
0
0
87073 Prostate Cancer (OD04720-01)
16.96
25.35


295


CNS ca. (glio) SNB-
0
0
87074 Prostate NAT (OD04720-02)
2.49
4.42


19


CNS ca. (glio/astro) U87-
0
0
Normal Lung GENPAK 061010
1.14
3.28


MG




CNS ca.* (neuro; met) SK-N-
0
0
83239 Lung Met to Muscle
1.13
0.16


AS


(ODO4286)


Mammary gland
2.4
4.27
83240 Muscle NAT (ODO4286)
1.48
1.05


Breast ca. BT-
0
0
84136 Lung Malignant Cancer
3.28
19.89


549


(OD03126)


Breast ca. MDA-
0
0
84137 Lung NAT (OD03126)
0.8
13.4


N


Breast ca.* (pl. effusion)
20.73
21.92
84871 Lung Cancer (OD04404)
1.42
3.74


T47D


Breast ca.* (pl. effusion)
71.2
57.43
84872 Lung NAT (OD04404)
0.92
1.45


MCF-7


Breast ca.* (pl.ef) MDA-MB-
6.08
8.9
84875 Lung Cancer (OD04565)
1.49
2.74


231


Small intestine
8.36
9.21
85950 Lung Cancer (OD04237-01)
5.33
13.97


Colorectal
4.61
6.34
85970 Lung NAT (OD04237-02)
4.07
13.49


Colon ca.
41.47
28.13
83255 Ocular Mel Met to Liver
0.71
1.14


HT29


(ODO4310)


Colon ca. CaCo-2
33.92
38.69
83256 Liver NAT (ODO4310)
8.48
23


Colon ca. HCT-
27.93
31.43
84139 Melanoma Mets to Lung
3.15
1.37


15


(OD04321)


Colon ca. HCT-
14.86
15.07
84138 Lung NAT (OD04321)
2.49
9.15


116


Colon ca. HCC-2998
30.35
37.63
Normal Kidney GENPAK 061008
0.94
7.48


Colon ca. SW480
13.3
18.69
83786 Kidney Ca, Nuclear grade 2
2.16
5.01





(OD04338)


Colon ca.* (SW480
30.35
38.42
83787 Kidney NAT (OD04338)
2.7
3.17


met)SW620


Stomach
1.13
4.51
83788 Kidney Ca Nuclear grade 1/2
17.92
4.18





(OD04339)


Gastric ca.* (liver met) NCI-
62.85
48.3
83789 Kidney NAT (OD04339)
6.79
4.74


N87


Heart
3.37
5.48
83790 Kidney Ca, Clear cell type
5.26
8.02





(OD04340)


Fetal Skeletal
0
0
83791 Kidney NAT (OD04340)
3.06
4.8


Skeletal muscle
0
0
83792 Kidney Ca, Nuclear grade 3
1.65
0.01





(OD04348)


Endothelial cells
0
0
83793 Kidney NAT (OD04348)
4.45
11.27


Endothelial cells (treated)
1.98
4.36
87474 Kidney Cancer (OD04622-01)
3.04
7.28


Kidney
4.61
7.18
87475 Kidney NAT (OD04622-03)
2.43
2.42


Kidney (fetal)
1.54
3.4
85973 Kidney Cancer (OD04450-01)
3.3
10.58


Renal ca. 786-0
4.74
4.27
85974 Kidney NAT (OD04450-03)
2.12
5.4


Renal ca. A498
8.3
8.96
Kidney Cancer Clontech 8120607
2.38
2.34


Renal ca. ACHN
1.98
5.67
Kidney NAT Clontech 8120608
1.58
4.15


Renal ca. TK-10
12.33
14.86
Kidney Cancer Clontech 8120613
1.98
7.23


Renal ca. UO-31
5.37
6.21
Kidney NAT Clontech 8120614
1.47
3.12


Renal ca. RXF 393
0
0
Kidney Cancer Clontech 9010320
0.95
0.34


Liver
7.59
10.51
Kidney NAT Clontech 9010321
3.06
2.96


Liver (fetal)
3.35
5.79
Normal Uterus GENPAK 061018
0.41
0.23


Liver ca. (hepatoblast) HepG2
12.5
16.38
Uterus Cancer GENPAK 064011
2.78
3.87


Lung
0.63
1.71
Normal Thyroid Clontech A+ 6570-
2.47
0.01





1**


Lung (fetal)
1.31
3.69
Thyroid Cancer GENPAK 064010
4.01
1.67


Lung ca. (non-s.cell) HOP-62
6.65
8.78
Thyroid Cancer INVITROGEN
4.77
2.37





A302152


Lung ca. (large cell)NCI-H460
0
8.08
Thyroid NAT INVITROGEN
3.56
12.16





A302153


Lung ca. (non-s.cell) NCI-H23
0
0.01
Normal Breast GENPAK 061019
1.18
3.47


Lung ca. (non-s.cl) NCI-H522
0
0.35
84877 Breast Cancer (OD04566)
5.71
18.69


Lung ca. (non-sm. cell) A549
32.09
43.53
85975 Breast Cancer (OD04590-01)
5.48
4.74


Lung ca. (s.cell var.) SHP-77
0.59
2.13
85976 Breast Cancer Mets (OD04590-
6.43
6.79





03)


Lung ca. (small cell) LX-1
100
100
87070 Breast Cancer Metastasis
4.33
3.17





(OD04655-05)


Lung ca. (small cell) NCI-
7.03
0
GENPAK Breast Cancer 064006
6.43
8.19


H69


Lung ca. (squam.) SW 900
8.48
10.96
Breast Cancer Clontech 9100266
5.29
5.4


Lung ca. (squam.) NCI-H596
4.8
8.54
Breast NAT Clontech 9100265
1.47
3.72


Lymph node
0
0
Breast Cancer INVITROGEN
5.11
8.19





A209073


Spleen
0
0
Breast NAT INVITROGEN A2090734
1.46
2.65


Thymus
0
0.04
Normal Liver GENPAK 061009
3.17
6.7


Ovary
0
0.04
Liver Cancer GENPAK 064003
2.98
0.74


Ovarian ca. IGROV-
10.73
11.11
Liver Cancer Research Genetics RNA
5.29
13.49


1


1025


Ovarian ca. OVCAR-
21.17
28.52
Liver Cancer Research Genetics RNA
2.59
5.15


3


1026


Ovarian ca. OVCAR-
4.42
5.87
Paired Liver Cancer Tissue Research
2.45
5.48


4


Genetics RNA 6004-T


Ovarian ca. OVCAR-
36.1
31.64
Paired Liver Tissue Research Genetics
3.96
3.3


5


RNA 6004-N


Ovarian ca. OVCAR-
4.7
4.58
Paired Liver Cancer Tissue Research
2.12
2.24


8


Genetics RNA 6005-T


Ovarian ca.* (ascites) SK-OV-
2.35
3.93
Paired Liver Tissue Research Genetics
3.64
4.15


3


RNA 6005-N


Pancreas
7.54
10.15
Normal Bladder GENPAK 061001
12.94
8.42


Pancreatic ca. CAPAN 2
27.17
25
Bladder Cancer Research Genetics
7.18
10.37





RNA 1023


Pituitary gland
6.43
6.52
Bladder Cancer INVITROGEN
2.9
0.3





A302173


Plancenta
26.61
34.87
87071 Bladder Cancer (OD04718-01)
18.43
4.36


Prostate
5.91
6.56
87072 Bladder Normal Adjacent
3.08
0.63





(OD04718-03)


Prostate ca.* (bone met)PC-3
3.12
4.24
Normal Ovary Res. Gen.
0.29
0.31


Salavary gland
8.25
11.91
Ovarian Cancer GENPAK 064008
3.15
1.24


Trachea
4.07
5.56
87492 Ovary Cancer (OD04768-07)
16.04
10.29


Spinal cord
0
0
87493 Ovary NAT (OD04768-08)
1.37
0.14


Testis
0
0
Normal Stomach GENPAK 061017
8.02
0.04


Thyroid
0
0
NAT Stomach Clontech 9060359
33.22
4.74


Uterus
0
0
Gastric Cancer Clontech 9060395
19.48
5.01


Melanoma M14
0
0.2
NAT Stomach Clontech 9060394
33.45
11.91


Melanoma LOX IMVI
0
0
Gastric Cancer Clontech 9060397
100
13.97


Melanoma UACC-62
0
0
NAT Stomach Clontech 9060396
51.05
40.9


Melanoma SK-MEL-28
6.43
6.38
Gastric Cancer GENPAK 064005
39.23
43.23


Melanoma* (met) SK-MEL-5
0.48
1.45


Melanoma Hs688(A).T
0
0


Melanoma* (met) Hs688(B).T
0
0






ca. = carcinoma




*= established from metastasis




met = metastasis




s cell var = small cell variant




non-s = non-sm = non-small




squam = squamous




pl. eff = pl effusion = pleural effusion




glio = glioma




astro = astrocytoma




neuro = neuroblastoma








[0374] It is seen from the above Table that clone AC016900_A is highly expressed in certain cell lines and weakly or not at all in many others. It is highly expressed in 83235 CC Mod Diff (OD03920), Gastric Cancer Clontech 9060397, and Lung ca. (small cell) LX-1. This observation indicates that the clone of PROT2 has a role in cell proliferation and potential utility as a marker to identify and/or stage tumors. It may furthermore be a target for a specific monoclonal antibody that could be used to treat various cancers, especially colon cancer, gastric cancer and lung cancer.
22TABLE 6Clone Identification No: 2826468.0.39 (PROT3)Probe Name: Ag2632SEQ IDPrimersSequencesNOForward5′-ACCACCCAGAAATGCAAACT-3′41ProbeFAM-5′-42GTCTCTGGGGAGCAGGTGTGTTTCCT-3′-TAMRAReverse5′-CTGTAAATACCGAGCCCTGG-3′43


[0375]

23





TABLE 7










Probe Name: Ag2808











SEQ ID


Primers
Sequences
NO





Forward
5′-ACCACCCAGAAATGCAAACT-3′
44


Probe
FAM-5′-
45



GTCTCTGGGGAGCAGGTGTGTTTCCT-3′-



TAMRA


Reverse
5′-CTGTAAATACCGAGCCCTGG-3′
46










[0376]

24





TABLE 8










Expression Data for Clone 2826468.0.39











Ag2632

Ag2808


Tissue_Name/Run_Name
% Rel Expr.
Tissue_Name/Run_Name
% Rel. Expr.













Liver adenocarcinoma
15.5
Normal Colon GENPAK 061003
40.9


Heart (fetal)
2.24
83219 CC Well to Mod Diff (ODO3866)
35.6


Pancreas
5.4
83220 CC NAT (ODO3866)
13.12


Pancreatic ca. CAPAN 2
15.18
83221 CC Gr.2 rectosigmoid (ODO3868)
6.61


Adrenal gland
22.85
83222 CC NAT (ODO3868)
0


Thyroid
4.42
83235 CC Mod Diff (ODO3920)
77.92


Salivary gland
8.08
83236 CC NAT (ODO3920)
20.31


Pituitary gland
34.87
83237 CC Gr.2 ascend colon (ODO3921)
85.26


Brain (fetal)
6.7
83238 CC NAT (ODO3921)
45.06


Brain (whole)
15.18
83241 CC from Partial Hepatectomy
93.95




(ODO4309)


Brain (amygdala)
12.16
83242 Liver NAT (ODO4309)
7.38


Brain (cerebellum)
14.76
87472 Colon mets to lung (OD04451-01)
38.96


Brain (hippocampus)
100
87473 Lung NAT (OD04451-02)
3.28


Brain (thalamus)
13.58
Normal Prostate Clontech A+ 6546-1
7.59


Cerebral Cortex
13.68
84140 Prostate Cancer (OD04410)
12.33


Spinal cord
6.84
84141 Prostate NAT (OD04410)
2.74


CNS ca. (glio/astro) U87-MG
0
87073 Prostate Cancer (OD04720-01)
28.32


CNS ca. (glio/astro) U-118-MG
4.64
87074 Prostate NAT (OD04720-02)
100


CNS ca. (astro) SW1783
7.91
Normal Lung GENPAK 061010
81.23


CNS ca.* (neuro; met) SK-N-AS
21.76
83239 Lung Met to Muscle (ODO4286)
10.96


CNS ca. (astro) SF-539
3.77
83240 Muscle NAT (ODO4286)
4.7


CNS ca. (astro) SNB-75
19.89
84136 Lung Malignant Cancer (OD03126)
53.96


CNS ca. (glio) SNB-19
14.76
84137 Lung NAT (OD03126)
24.32


CNS ca. (glio) U251
4.77
84871 Lung Cancer (OD04404)
0


CNS ca. (glio) SF-295
30.35
84872 Lung NAT (OD04404)
12.59


Heart
1.99
84875 Lung Cancer (OD04565)
4.58


Skeletal muscle
0
85950 Lung Cancer (OD04237-01)
13.3


Bone marrow
14.06
85970 Lung NAT (OD04237-02)
5.52


Thymus
30.35
83255 Ocular Mel Met to Liver (ODO4310)
2.06


Spleen
55.86
83256 Liver NAT (ODO4310)
3.47


Lymph node
53.96
84139 Melanoma Mets to Lung (OD04321)
48.3


Colorectal
22.07
84138 Lung NAT (OD04321)
13.03


Stomach
29.32
Normal Kidney GENPAK 061008
28.72


Small intestine
40.9
83786 Kidney Ca, Nuclear grade 2
17.31




(OD04338)


Colon ca. SW480
21.17
83787 Kidney NAT (OD04338)
19.48


Colon ca.* (SW480 met)SW620
10.96
83788 Kidney Ca Nuclear grade 1/2
33.45




(OD04339)


Colon ca. HT29
10.29
83789 Kidney NAT (OD04339)
12.67


Colon ca. HCT-116
2.72
83790 Kidney Ca, Clear cell type (OD04340)
16.72


Colon ca. CaCo-2
64.62
83791 Kidney NAT (OD04340)
35.85


83219 CC Well to Mod Diff
6.12
83792 Kidney Ca, Nuclear grade 3
6.38


(ODO3866)

(OD04348)


Colon ca. HCC-2998
14.36
83793 Kidney NAT (OD04348)
35.36


Gastric ca.* (liver met) NCI-N87
21.02
87474 Kidney Cancer (OD04622-01)
5.52


Bladder
3.87
87475 Kidney NAT (OD04622-03)
0


Trachea
13.12
85973 Kidney Cancer (OD04450-01)
92.66


Kidney
2.63
85974 Kidney NAT (OD04450-03)
23


Kidney (fetal)
9.28
Kidney Cancer Clontech 8120607
0


Renal ca. 786-0
12.33
Kidney NAT Clontech 8120608
11.34


Renal ca. A498
42.34
Kidney Cancer Clontech 8120613
16.72


Renal ca. RXF 393
4.94
Kidney NAT Clontech 8120614
44.44


Renal ca. ACHN
2.19
Kidney Cancer Clontech 9010320
7.97


Renal ca. UO-31
6.21
Kidney NAT Clontech 9010321
57.04


Renal ca. TK-10
23.33
Normal Uterus GENPAK 061018
3.93


Liver
3.17
Uterus Cancer GENPAK 064011
5.37


Liver (fetal)
22.38
Normal Thyroid Clontech A+ 6570-1
5.22


Liver ca. (hepatoblast) HepG2
21.76
Thyroid Cancer GENPAK 064010
13.3


Lung
32.76
Thyroid Cancer INVITROGEN A302152
8.9


Lung (fetal)
18.56
Thyroid NAT INVITROGEN A302153
9.21


Lung ca. (small cell) LX-1
4.33
Normal Breast GENPAK 061019
41.18


Lung ca. (small cell) NCI-H69
14.26
84877 Breast Cancer (OD04566)
27.55


Lung ca. (s.cell var.) SHP-77
6.65
85975 Breast Cancer (OD04590-01)
48.97


Lung ca. (large cell)NCI-H460
1.05
85976 Breast Cancer Mets (OD04590-03)
61.13


Lung ca. (non-sm. cell) A549
5.75
87070 Breast Cancer Metastasis (OD04655-
50.35




05)


Lung ca. (non-s.cell) NCI-H23
13.87
GENPAK Breast Cancer 064006
5.56


Lung ca (non-s.cell) HOP-62
87.06
Breast Cancer Clontech 9100266
13.58


Lung ca. (non-s.cl) NCI-H522
12.5
Breast NAT Clontech 9100265
0


Lung ca. (squam.) SW 900
5.91
Breast Cancer INVITROGEN A209073
10.37


Lung ca. (squam.) NCI-H596
8.9
Breast NAT INVITROGEN A2090734
40.33


Mammary gland
3.56
Normal Liver GENPAK 061009
0


Breast ca.* (pl. effusion) MCF-7
2.65
Liver Cancer GENPAK 064003
1.77


Breast ca.* (pl.ef) MDA-MB-231
8.19
Liver Cancer Research Genetics RNA 1025
10.29


Breast ca.* (pl. effusion) T47D
0
Liver Cancer Research Genetics RNA 1026
2.42


Breast ca. BT-549
4.15
Paired Liver Cancer Tissue Research Genetics
8.48




RNA 6004-T


Breast ca. MDA-N
0
Paired Liver Tissue Research Genetics RNA
12.85




6004-N


Ovary
3.79
Paired Liver Cancer Tissue Research Genetics
3.35




RNA 6005-T


Ovarian ca. OVCAR-3
23.16
Paired Liver Tissue Research Genetics RNA
0




6005-N


Ovarian ca. OVCAR-4
3.04
Normal Bladder GENPAK 061001
34.63


Ovarian ca. OVCAR-5
35.36
Bladder Cancer Research Genetics RNA 1023
11.03


Ovarian ca. OVCAR-8
9.54
Bladder Cancer INVITROGEN A302173
12.94


Ovarian ca. IGROV- 1
0
87071 Bladder Cancer (OD04718-01)
3.74


Ovarian ca.* (ascites) SK-OV-3
0
87072 Bladder Normal Adjacent (OD04718-
14.06




03)


Uterus
5.67
Normal Ovary Res. Gen.
1.63


Plancenta
12.94
Ovarian Cancer GENPAK 064008
37.89


Prostate
14.46
87492 Ovary Cancer (OD04768-07)
3.42


Prostate ca.* (bone met)PC-3
0
87493 Ovary NAT (OD04768-08)
3.98


Testis
26.24
Normal Stomach GENPAK 061017
4.36


Melanoma Hs688(A).T
5.08
NAT Stomach Clontech 9060359
5.08


Melanoma* (met) Hs688(B).T
8.19
Gastric Cancer Clontech 9060395
14.26


Melanoma UACC-62
1.83
NAT Stomach Clontech 9060394
18.95


Melanoma M14
1.05
Gastric Cancer Clontech 9060397
64.17


Melanoma LOX IMVI
28.13
NAT Stomach Clontech 9060396
0


Melanoma* (met) SK-MEL-5
5.87
Gastric Cancer GENPAK 064005
12.33


Adipose
4.64






ca. = carcinoma




*= established from metastasis




met = metastasis




s cell var = small cell variant




non-s = non-sm = non-small




squam = squamous




pl. eff = pl effusion = pleural effusion




glio = glioma




astro = astrocytoma




neuro = neuroblastoma








[0377] It is seen from the Table above that clone 2826468.0.39 is expressed in most normal and cancer cells assayed. It is especially prominent in 83241 CC from Partial Hepatectomy (OD04309), Brain (hippocampus), 87074 Prostate NAT (OD04720-02), and 85973 Kidney Cancer (OD04450-01). This observation indicates that the clone of PROT3 has a role in cell proliferation and potential utility as a marker to identify and/or stage tumors. It may furthermore be a target for a specific monoclonal antibody that could be used to treat various cancers, especially kidney cancer.
25TABLE 9Clone Identification No: AC015862_A (PROT4)Probe Name: Ag713SEQ IDPrimersSequencesNOForward5′-AGGGACATTTCCACAGTCAAG-3′47ProbeFAM-5′-48CTCTTTGCCAAGCGACAACAGGAG-3′-TAMRAReverse5′-TCTCTCCTTCCACCTGGAAT-3′49


[0378]

26





TABLE 10










Expression Data for Clone AC015862_A












% Rel.
% Rel.

% Rel.


Tissue_Name/Run_Name
Expr.
Expr.
Tissue_Name/Run_Name
Expr.














Endothelial cells
0
0
Normal Colon GENPAK 061003
9.15


Endothelial cells (treated)
0
0
83219 CC Well to Mod Diff
2.63





(ODO3866)


Pancreas
0.4
0
83220 CC NAT (ODO3866)
1.55


Pancreatic ca. CAPAN 2
0.14
0
83221 CC Gr.2 rectosigmoid
0





(ODO3868)


Adrenal Gland (new lot*)
0
0
83222 CC NAT (ODO3868)
0


Thyroid
0
0
83235 CC Mod Diff (ODO3920)
0


Salavary gland
0
0
83236 CC NAT (ODO3920)
2.24


Pituitary gland
0
0
83237 CC Gr.2 ascend colon
0





(ODO3921)


Brain (fetal)
0
0
83238 CC NAT (ODO3921)
1.9


Brain (whole)
0
0
83241 CC from Partial Hepatectomy
2.35





(ODO4309)


Brain (amygdala)
0
0
83242 Liver NAT (ODO4309)
0


Brain (cerebellum)
0
0
87472 Colon mets to lung (OD04451-
0





01)


Brain (hippocampus)
0
0
87473 Lung NAT (OD04451-02)
0


Brain (thalamus)
0
0
Normal Prostate Clontech A+ 6546-1
5.08


Cerebral Cortex
0
0
84140 Prostate Cancer (OD04410)
0


Spinal cord
0
0
84141 Prostate NAT (OD04410)
2.54


CNS ca. (glio/astro) U87-MG
0
0
87073 Prostate Cancer (OD04720-01)
1.82


CNS ca. (glio/astro) U-118-
0
0
87074 Prostate NAT (OD04720-02)
3.26


MG


CNS ca. (astro) SW1783
0
0
Normal Lung GENPAK 061010
1.65


CNS ca.* (neuro; met) SK-N-
0
0
83239 Lung Met to Muscle (ODO4286)
0


AS


CNS ca. (astro) SF-539
0
0
83240 Muscle NAT (ODO4286)
0


CNS ca. (astro) SNB-75
0
0
84136 Lung Malignant Cancer
0





(OD03126)


CNS ca. (glio) SNB-19
0
0
84137 Lung NAT (OD03126)
0


CNS ca. (glio) U251
0
0
84871 Lung Cancer (OD04404)
17.92


CNS ca. (glio) SF-295
0
0
84872 Lung NAT (OD04404)
0


Heart
0
0
84875 Lung Cancer (OD04565)
11.11


Skeletal Muscle (new lot*)
0
0
85950 Lung Cancer (OD04237-01)
2.86


Bone marrow
0
0
85970 Lung NAT (OD04237-02)
3.79


Thymus
0
0
83255 Ocular Mel Met to Liver
0





(ODO4310)


Spleen
0
0
83256 Liver NAT (ODO4310)
0


Lymph node
0
0
84139 Melanoma Mets to Lung
0





(OD04321)


Colorectal
0
0
84138 Lung NAT (OD04321)
3.37


Stomach
0
0
Normal Kidney GENPAK 061008
0


Small intestine
22.22
12.33
83786 Kidney Ca, Nuclear grade 2
0





(OD04338)


Colon ca. SW480
0
0
83787 Kidney NAT (OD04338)
0


Colon ca.* (SW480 met)SW620
0
0
83788 Kidney Ca Nuclear grade 1/2
4.15





(OD04339)


Colon ca. HT29
0
0
83789 Kidney NAT (OD04339)
0


Colon ca. HCT-116
0
0
83790 Kidney Ca, Clear cell type
27.93





(OD04340)


Colon ca. CaCo-2
0
0
83791 Kidney NAT (OD04340)
100


83219 CC Well to Mod Diff
0
0
83792 Kidney Ca, Nuclear grade 3
0


(ODO3866)


(OD04348)


Colon ca. HCC-2998
0
0
83793 Kidney NAT (OD04348)
18.43


Gastric ca.* (liver met) NCI-N87
0
0
87474 Kidney Cancer (OD04622-01)
0


Bladder
10.58
0
87475 Kidney NAT (OD04622-03)
3.15


Trachea
100
100
85973 Kidney Cancer (OD04450-01)
1.6


Kidney
0
0.08
85974 Kidney NAT (OD04450-03)
0


Kidney (fetal)
3.1
0.9
Kidney Cancer Clontech 8120607
0


Renal ca. 786-0
40.05
35.36
Kidney NAT Clontech 8120608
0


Renal ca. A498
8.78
0.08
Kidney Cancer Clontech 8120613
0


Renal ca. RXF 393
0
0
Kidney NAT Clontech 8120614
0


Renal ca. ACHN
58.64
53.96
Kidney Cancer Clontech 9010320
0


Renal ca. UO-31
0.88
0.07
Kidney NAT Clontech 9010321
2.74


Renal ca. TK-10
13.03
0.4
Normal Uterus GENPAK 061018
0


Liver
0
0
Uterus Cancer GENPAK 064011
3


Liver (fetal)
0
0
Normal Thyroid Clontech A+ 6570-1
0


Liver ca. (hepatoblast) HepG2
0
0
Thyroid Cancer GENPAK 064010
2.74


Lung
3.59
0.09
Thyroid Cancer INVITROGEN
0





A302152


Lung (fetal)
0.03
0
Thyroid NAT INVITROGEN A302153
0


Lung ca. (small cell) LX-1
1.78
0.03
Normal Breast GENPAK 061019
4.42


Lung ca. (small cell) NCI-H69
0.15
0
84877 Breast Cancer (OD04566)
0


Lung ca. (s.cell var.) SHP-77
0
0
85975 Breast Cancer (OD04590-01)
0


Lung ca. (large cell)NCI-H460
0
0
85976 Breast Cancer Mets (OD04590-
0





03)


Lung ca. (non-sm. cell) A549
0
0
87070 Breast Cancer Metastasis
7.64





(OD04655-05)


Lung ca. (non-s.cell) NCI-H23
0
0
GENPAK Breast Cancer 064006
4.39


Lung ca (non-s.cell) HOP-62
0
0
Breast Cancer Clontech 9100266
4.33


Lung ca. (non-s.cl) NCI-H522
0
0
Breast NAT Clontech 9100265
0


Lung ca. (squam.) SW 900
0
0
Breast Cancer INVITROGEN A209073
0


Lung ca. (squam.) NCI-H596
40.05
27.55
Breast NAT INVITROGEN A2090734
0


Mammary gland
0.64
0.8
Normal Liver GENPAK 061009
0


Breast ca.* (pl. effusion) MCF-7
0
0
Liver Cancer GENPAK 064003
0


Breast ca.* (pl.ef) MDA-MB-
0.71
0
Liver Cancer Research Genetics RNA
0


231


1025


Breast ca.* (pl. effusion) T47D
0
0
Liver Cancer Research Genetics RNA
0





1026


Breast ca. BT-549
0
0
Paired Liver Cancer Tissue Research
0





Genetics RNA 6004-T


Breast ca. MDA-N
0
0
Paired Liver Tissue Research Genetics
0





RNA 6004-N


Ovary
0
0
Paired Liver Cancer Tissue Research
0





Genetics RNA 6005-T


Ovarian ca. OVCAR-3
50.35
36.1
Paired Liver Tissue Research Genetics
0





RNA 6005-N


Ovarian ca. OVCAR-4
95.93
89.5
Normal Bladder GENPAK 061001
2.54


Ovarian ca. OVCAR-5
0
0
Bladder Cancer Research Genetics RNA
0





1023


Ovarian ca. OVCAR-8
0
0
Bladder Cancer INVITROGEN
3.74





A302173


Ovarian ca. IGROV-1
0
0
87071 Bladder Cancer (OD04718-01)
2.66


Ovarian ca.* (ascites) SK-OV-3
1.44
0.02
87072 Bladder Normal Adjacent
0





(OD04718-03)


Uterus
0
0
Normal Ovary Res. Gen.
0


Plancenta
0.05
0
Ovarian Cancer GENPAK 064008
0


Prostate
0.71
0
87492 Ovary Cancer (OD04768-07)
0


Prostate ca.* (bone met)PC-3
0
0
87493 Ovary NAT (OD04768-08)
0


Testis
0
0
Normal Stomach GENPAK 061017
0


Melanoma Hs688(A).T
0
0
NAT Stomach Clontech 9060359
0


Melanoma* (met) Hs688(B).T
0
0
Gastric Cancer Clontech 9060395
4.45


Melanoma UACC-62
0
0
NAT Stomach Clontech 9060394
0


Melanoma M14
0
0
Gastric Cancer Clontech 9060397
1.82


Melanoma LOX IMVI
0
0
NAT Stomach Clontech 9060396
0


Melanoma* (met) SK-MEL-5
0
0
Gastric Cancer GENPAK 064005
0


Adipose
0
0






ca. = carcinoma




*= established from metastasis




met = metastasis




s cell var = small cell variant




non-s = non-sm = non-small




squam = squamous




pl. eff = pl effusion = pleural effusion




glio = glioma




astro = astrocytoma




neuro = neuroblastoma








[0379] It is seen from the above Table that clone AC015862_A is highly expressed in certain cell lines and weakly or not at all in many others. It most prominently expressed in Trachea, 83791 Kidney NAT (OD04340), Ovarian ca. OVCAR-4, Ovarian ca. OVCAR-3, Renal ca. 786-0, Renal ca. ACHN, and Lung ca. (squam.) NCI-H596. This observation indicates that the clone of PROT4 has a role in cell proliferation and potential utility as a marker to identify and/or stage tumors. It may furthermore be a target for a specific monoclonal antibody that could be used to treat various cancers, especially renal cancer, ovarian cancer and lung cancer.



Other Embodiments

[0380] Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims. In particular, it is contemplated by the inventor that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. Therefore, other aspects, advantages, and modifications are within the scope of the following claims.


Claims
  • 1. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of: a) a mature form of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8; b) a variant of a mature form of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8, wherein any amino acid in the mature form is changed to a different amino acid, provided that no more than 5% of the amino acid residues in the sequence of the mature form are so changed; c) the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8; d) a variant of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8 wherein any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 5% of the amino acid residues in the sequence are so changed; and e) a fragment of any of a) through d).
  • 2. The polypeptide of claim 1 that is a naturally occurring allelic variant of the sequence given by SEQ ID NO: 2, 4, 6, and 8.
  • 3. The polypeptide of claim 2, wherein the variant is the translation of a single nucleotide polymorphism.
  • 4. The polypeptide of claim 1 that is a variant polypeptide described therein, wherein any amino acid specified in the chosen sequence is changed to provide a conservative substitution.
  • 5. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of: a) a mature form of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8; b) a variant of a mature form of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8 wherein any amino acid in the mature form of the chosen sequence is changed to a different amino acid, provided that no more than 5% of the amino acid residues in the sequence of the mature form are so changed; c) the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8; d) a variant of the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8, in which any amino acid specified in the chosen sequence is changed to a different amino acid, provided that no more than 5% of the amino acid residues in the sequence are so changed; e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8 or any variant of said polypeptide wherein any amino acid of the chosen sequence is changed to a different amino acid, provided that no more than 5% of the amino acid residues in the sequence are so changed; and f) the complement of any of said nucleic acid molecules.
  • 6. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally occurring allelic nucleic acid variant.
  • 7. The nucleic acid molecule of claim 5 that encodes a variant polypeptide, wherein the variant polypeptide has the polypeptide sequence of a naturally occurring polypeptide variant.
  • 8. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises a single nucleotide polymorphism encoding said variant polypeptide.
  • 9. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of a) the nucleotide sequence given by SEQ ID NO:1, 3, 5, and 7; b) a nucleotide sequence wherein one or more nucleotides in the nucleotide sequence given by SEQ ID NO:1, 3, 5, and 7 is changed from that given by the chosen sequence to a different nucleotide provided that no more than 5% of the nucleotides are so changed; c) a nucleic acid fragment of the sequence given by SEQ ID NO:1, 3, 5, and 7; and d) a nucleic acid fragment wherein one or more nucleotides in the nucleotide sequence given by SEQ ID NO:1, 3, 5, and 7 is changed from that given by the chosen sequence to a different nucleotide provided that no more than 5% of the nucleotides are so changed.
  • 10. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule hybridizes under stringent conditions to the nucleotide sequence given by SEQ ID NO:1, 3, 5, and 7, or a complement of said nucleotide sequence.
  • 11. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises a nucleotide sequence in which any nucleotide specified in the coding sequence of the chosen nucleotide sequence is changed from that given by the chosen sequence to a different nucleotide provided that no more than 5% of the nucleotides in the chosen coding sequence are so changed, an isolated second polynucleotide that is a complement of the first polynucleotide, or a fragment of any of them.
  • 12. A vector comprising the nucleic acid molecule of claim 11.
  • 13. The vector of claim 12, further comprising a promoter operably linked to said nucleic acid molecule.
  • 14. A cell comprising the vector of claim 12.
  • 15. An antibody that binds immunospecifically to the polypeptide of claim 1.
  • 16. The antibody of claim 15, wherein said antibody is a monoclonal antibody.
  • 17. The antibody of claim 15, wherein said antibody is a humanized antibody.
  • 18. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising: (a) providing said sample; (b) introducing said sample to an antibody that binds immunospecifically to the polypeptide; and (c) determining the presence or amount of antibody bound to said polypeptide, thereby determining the presence or amount of polypeptide in said sample.
  • 19. A method for determining the presence or amount of the nucleic acid molecule of claim 5 in a sample, the method comprising: (a) providing said sample; (b) introducing said sample to a probe that binds to said nucleic acid molecule; and (c) determining the presence or amount of said probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
  • 20. A method of identifying an agent that binds to the polypeptide of claim 1, the method comprising: (a) introducing said polypeptide to said agent; and (b) determining whether said agent binds to said polypeptide.
  • 21. A method for identifying a potential therapeutic agent for use in treatment of a pathology, wherein the pathology is related to aberrant expression or aberrant physiological interactions of the polypeptide of claim 1, the method comprising: (a) providing a cell expressing the polypeptide of claim 1 and having a property or function ascribable to the polypeptide; (b) contacting the cell with a composition comprising a candidate substance; and (c) determining whether the substance alters the property or function ascribable to the polypeptide; whereby, if an alteration observed in the presence of the substance is not observed when the cell is contacted with a composition devoid of the substance, the substance is identified as a potential therapeutic agent.
  • 22. A method for modulating the activity of the polypeptide of claim 1, the method comprising introducing a cell sample expressing the polypeptide of said claim with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
  • 23. A method of treating or preventing a pathology associated with the polypeptide of claim 1, said method comprising administering the polypeptide of claim 1 to a subject in which such treatment or prevention is desired in an amount sufficient to treat or prevent said pathology in said subject.
  • 24. The method of claim 23, wherein said subject is a human.
  • 25. A method of treating or preventing a pathology associated with the polypeptide of claim 1, said method comprising administering to a subject in which such treatment or prevention is desired a PROTX nucleic acid in an amount sufficient to treat or prevent said pathology in said subject.
  • 26. The method of claim 25, wherein said subject is a human.
  • 27. A method of treating or preventing a pathology associated with the polypeptide of claim 1, said method comprising administering to a subject in which such treatment or prevention is desired a PROTX antibody in an amount sufficient to treat or prevent said pathology in said subject.
  • 28. The method of claim 27, wherein the subject is a human.
  • 29. A pharmaceutical composition comprising the polypeptide of claim 1 and a pharmaceutically acceptable carrier.
  • 30. A pharmaceutical composition comprising the nucleic acid molecule of claim 5 and a pharmaceutically acceptable carrier.
  • 31. A pharmaceutical composition comprising the antibody of claim 15 and a pharmaceutically acceptable carrier.
  • 32. A kit comprising in one or more containers, the pharmaceutical composition of claim 29.
  • 33. A kit comprising in one or more containers, the pharmaceutical composition of claim 30.
  • 34. A kit comprising in one or more containers, the pharmaceutical composition of claim 31.
  • 35. A method for screening for a modulator of activity or of latency or predisposition to a pathology associated with the polypeptide of claim 1, said method comprising: a) administering a test compound to a test animal at increased risk for a pathology associated with the polypeptide of claim 1, wherein said test animal recombinantly expresses the polypeptide of claim 1;b) measuring the activity of said polypeptide in said test animal after administering the compound of step (a); and c) comparing the activity of said protein in said test animal with the activity of said polypeptide in a control animal not administered said polypeptide, wherein a change in the activity of said polypeptide in said test animal relative to said control animal indicates the test compound is a modulator of latency of, or predisposition to, a pathology associated with the polypeptide of claim 1.
  • 36. The method of claim 35, wherein said test animal is a recombinant test animal that expresses a test protein transgene or expresses said transgene under the control of a promoter at an increased level relative to a wild-type test animal, and wherein said promoter is not the native gene promoter of said transgene.
  • 37. A method for determining the presence of or predisposition to a disease associated with altered levels of the polypeptide of claim 1 in a first mammalian subject, the method comprising: a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and b) comparing the amount of said polypeptide in the sample of step (a) to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease, wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to said disease.
  • 38. A method for determining the presence of or predisposition to a disease associated with altered levels of the nucleic acid molecule of claim 5 in a first mammalian subject, the method comprising: a) measuring the amount of the nucleic acid in a sample from the first mammalian subject; and b) comparing the amount of said nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
  • 39. A method of treating a pathological state in a mammal, the method comprising administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising the amino acid sequence given by SEQ ID NO: 2, 4, 6, and 8 or a biologically active fragment thereof.
  • 40. A method of treating a pathological state in a mammal, the method comprising administering to the mammal the antibody of claim 15 in an amount sufficient to alleviate the pathological state.
RELATED APPLICATIONS

[0001] This application claims priority to U.S. Ser. No. 60/180,880 filed Feb. 8, 2000, USSN 60/181,044 filed Feb. 8, 2000, U.S. Ser. No. 60/181,656 filed Feb. 10, 2000 and U.S. Ser. No. 60/182,795 filed Feb. 15, 2000. The contents of these applications are incorporated herein by reference in their entireties.

Provisional Applications (4)
Number Date Country
60180880 Feb 2000 US
60181044 Feb 2000 US
60181656 Feb 2000 US
60182795 Feb 2000 US