Cathepsin V-like polypeptides

Information

  • Patent Grant
  • 6783969
  • Patent Number
    6,783,969
  • Date Filed
    Monday, March 5, 2001
    23 years ago
  • Date Issued
    Tuesday, August 31, 2004
    20 years ago
Abstract
The present invention provides novel nucleic acids, novel polypeptide sequences encoded by these nucleic acids and uses thereof.
Description




1. BACKGROUND OF THE INVENTION




1.1 Technical Field




The present invention provides novel polynucleotides and proteins encoded by such polynucleotides, along with uses for these polynucleotides and proteins, for example in therapeutic, diagnostic and research methods.




1.2 Background




Technology aimed at the discovery of protein factors (including e.g., cytokines, such as lymphokines, interferons, CSFs, chemokines, and interleukins) has matured rapidly over the past decade. The now routine hybridization cloning and expression cloning techniques clone novel polynucleotides “directly” in the sense that they rely on information directly related to the discovered protein (i.e., partial DNA/amino acid sequence of the protein in the case of hybridization cloning; activity of the protein in the case of expression cloning). More recent “indirect” cloning techniques such as signal sequence cloning, which isolates DNA sequences based on the presence of a now well-recognized secretory leader sequence motif, as well as various PCR-based or low stringency hybridization-based cloning techniques, have advanced the state of the art by making available large numbers of DNA/amino acid sequences for proteins that are known to have biological activity, for example, by virtue of their secreted nature in the case of leader sequence cloning, by virtue of their cell or tissue source in the case of PCR-based techniques, or by virtue of structural similarity to other genes of known biological activity.




Identified polynucleotide and polypeptide sequences have numerous applications in, for example, diagnostics, forensics, gene mapping; identification of mutations responsible for genetic disorders or other traits, to assess biodiversity, and to produce many other types of data and products dependent on DNA and amino acid sequences.




2. SUMMARY OF THE INVENTION




The compositions of the present invention include novel isolated polypeptides, novel isolated polynucleotides encoding such polypeptides, including recombinant DNA molecules, cloned genes or degenerate variants thereof, especially naturally occurring variants such as allelic variants, antisense polynucleotide molecules, and antibodies that specifically recognize one or more epitopes present on such polypeptides, as well as hybridomas producing such antibodies.




The compositions of the present invention additionally include vectors, including expression vectors, containing the polynucleotides of the invention, cells genetically engineered to contain such polynucleotides and cells genetically engineered to express such polynucleotides.




The present invention relates to a collection or library of at least one novel nucleic acid sequence assembled from expressed sequence tags (ESTs) isolated mainly by sequencing by hybridization (SBM), and in some cases, sequences obtained from one or more public databases. The invention relates also to the proteins encoded by such polynucleotides, along with therapeutic, diagnostic and research utilities for these polynucleotides and proteins. These nucleic acid sequences are designated as SEQ ID NO: 1-948 and are provided in the Sequence Listing. In the nucleic acids provided in the Sequence Listing, A is adenine; C is cytosine; G is guanine; T is thymine; and N is any of the four bases. In the amino acids provided in the Sequence Listing, * corresponds to the stop codon.




The nucleic acid sequences of the present invention also include, nucleic acid sequences that hybridize to the complement of SEQ ID NO: 1-948 under stringent hybridization conditions; nucleic acid sequences which are allelic variants or species homologues of any of the nucleic acid sequences recited above, or nucleic acid sequences that encode a peptide comprising a specific domain or truncation of the peptides encoded by SEQ ID NO: 1-948. A polynucleotide comprising a nucleotide sequence having at least 90% identity to an identifying sequence of SEQ ID NO: 1-948 or a degenerate variant or fragment thereof. The identifying sequence can be 100 base pairs in length.




The nucleic acid sequences of the present invention also include the sequence information from the nucleic acid sequences of SEQ ID NO: 1-948. The sequence information can be a segment of any one of SEQ ID NO: 1-948 that uniquely identifies or represents the sequence information of SEQ ID NO: 1-948.




A collection as used in this application can be a collection of only one polynucleotide. The collection of sequence information or identifying information of each sequence can be provided on a nucleic acid array. In one embodiment, segments of sequence information is provided on a nucleic acid array to detect the polynucleotide that contains the segment. The array can be designed to detect full-match or mismatch to the polynucleotide that contains the segment. The collection can also be provided in a computer-readable format.




This invention also includes the reverse or direct complement of any of the nucleic acid sequences recited above; cloning or expression vectors containing the nucleic acid sequences; and host cells or organisms transformed with these expression vectors. Nucleic acid sequences (or their reverse or direct complements) according to the invention have numerous applications in a variety of techniques known to those skilled in the art of molecular biology, such as use as hybridization probes, use as primers for PCR, use in an array, use in computer-readable media, use in sequencing full-length genes, use for chromosome and gene mapping, use in the recombinant production of protein, and use in the generation of anti-sense DNA or RNA, their chemical analogs and the like.




In a preferred embodiment, the nucleic acid sequences of SEQ ID NO: 1-948 or novel segments or parts of the nucleic acids of the invention are used as primers in expression assays that are well known in the art. In a particularly preferred embodiment, the nucleic acid sequences of SEQ ID NO: 1-948 or novel segments or parts of the nucleic acids provided herein are used in diagnostics for identifying expressed genes or, as well known in the art and exemplified by Vollrath et al., Science 258:52-59 (1992), as expressed sequence tags for physical mapping of the human genome.




The isolated polynucleotides of the invention include, but are not limited to, a polynucleotide comprising any one of the nucleotide sequences set forth in SEQ ID NO: 1-948; a polynucleotide comprising any of the full length protein coding sequences of SEQ ID NO: 1-948; and a polynucleotide comprising any of the nucleotide sequences of the mature protein coding sequences of SEQ ID NO: 1-948. The polynucleotides of the present invention also include, but are not limited to, a polynucleotide that hybridizes under stringent hybridization conditions to (a) the complement of any one of the nucleotide sequences set forth in SEQ ID NO: 1-948; (b) a nucleotide sequence encoding any one of the amino acid sequences set forth in the Sequence Listing; (c) a polynucleotide which is an allelic variant of any polynucleotides recited above; (d) a polynucleotide which encodes a species homolog (e.g. orthologs) of any of the proteins recited above; or (e) a polynucleotide that encodes a polypeptide comprising a specific domain or truncation of any of the polypeptides comprising an amino acid sequence set forth in the Sequence Listing.




The isolated polypeptides of the invention include, but are not limited to, a polypeptide comprising any of the amino acid sequences set forth in the Sequence Listing; or the corresponding full length or mature protein. Polypeptides of the invention also include polypeptides with biological activity that are encoded by (a) any of the polynucleotides having a nucleotide sequence set forth in SEQ ID NO: 1-948; or (b) polynucleotides that hybridize to the complement of the polynucleotides of (a) under stringent hybridization conditions. Biologically or immunologically active variants of any of the polypeptide sequences in the Sequence Listing, and “substantial equivalents” thereof (e.g., with at least about 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid sequence identity) that preferably retain biological activity are also contemplated. The polypeptides of the invention may be wholly or partially chemically synthesized but are preferably produced by recombinant means using the genetically engineered cells (e.g. host cells) of the invention.




The invention also provides compositions comprising a polypeptide of the invention. Polypeptide compositions of the invention may further comprise an acceptable carrier, such as a hydrophilic, e.g., pharmaceutically acceptable, carrier.




The invention also provides host cells transformed or transfected with a polynucleotide of the invention.




The invention also relates to methods for producing a polypeptide of the invention comprising growing a culture of the host cells of the invention in a suitable culture medium under conditions permitting expression of the desired polypeptide, and purifying the polypeptide from the culture or from the host cells. Preferred embodiments include those in which the protein produced by such process is a mature form of the protein.




Polynucleotides according to the invention have numerous applications in a variety of techniques known to those skilled in the art of molecular biology. These techniques include use as hybridization probes, use as oligomers, or primers, for PCR, use for chromosome and gene mapping, use in the recombinant production of protein, and use in generation of anti-sense DNA or RNA, their chemical analogs and the like. For example, when the expression of an mRNA is largely restricted to a particular cell or tissue type, polynucleotides of the invention can be used as hybridization probes to detect the presence of the particular cell or tissue mRNA in a sample using, e.g., in situ hybridization.




In other exemplary embodiments, the polynucleotides are used in diagnostics as expressed sequence tags for identifying expressed genes or, as well known in the art and exemplified by Vollrath et al., Science 258:52-59 (1992), as expressed sequence tags for physical mapping of the human genome.




The polypeptides according to the invention can be used in a variety of conventional procedures and methods that are currently applied to other proteins. For example, a polypeptide of the invention can be used to generate an antibody that specifically binds the polypeptide. Such antibodies, particularly monoclonal antibodies, are useful for detecting or quantitating the polypeptide in tissue. The polypeptides of the invention can also be used as molecular weight markers, and as a food supplement.




Methods are also provided for preventing, treating, or ameliorating a medical condition which comprises the step of administering to a mammalian subject a therapeutically effective amount of a composition comprising a polypeptide of the present invention and a pharmaceutically acceptable carrier.




In particular, the polypeptides and polynucleotides of the invention can be utilized, for example, in methods for the prevention and/or treatment of disorders involving aberrant protein expression or biological activity.




The present invention further relates to methods for detecting the presence of the polynucleotides or polypeptides of the invention in a sample. Such methods can, for example, be utilized as part of prognostic and diagnostic evaluation of disorders as recited herein and for the identification of subjects exhibiting a predisposition to such conditions. The invention provides a method for detecting the polynucleotides of the invention in a sample, comprising contacting the sample with a compound that binds to and forms a complex with the polynucleotide of interest for a period sufficient to form the complex and under conditions sufficient to form a complex and detecting the complex such that if a complex is detected, the polynucleotide of interest is detected. The invention also provides a method for detecting the polypeptides of the invention in a sample comprising contacting the sample with a compound that binds to and forms a complex with the polypeptide under conditions and for a period sufficient to form the complex and detecting the formation of the complex such that if a complex is formed, the polypeptide is detected.




The invention also provides kits comprising polynucleotide probes and/or monoclonal antibodies, and optionally quantitative standards, for carrying out methods of the invention. Furthermore, the invention provides methods for evaluating the efficacy of drugs, and monitoring the progress of patients, involved in clinical trials for the treatment of disorders as recited above.




The invention also provides methods for the identification of compounds that modulate (i.e., increase or decrease) the expression or activity of the polynucleotides and/or polypeptides of the invention. Such methods can be utilized, for example, for the identification of compounds that can ameliorate symptoms of disorders as recited herein. Such methods can include, but are not limited to, assays for identifying compounds and other substances that interact with (e.g., bind to) the polypeptides of the invention. The invention provides a method for identifying a compound that binds to the polypeptides of the invention comprising contacting the compound with a polypeptide of the invention in a cell for a time sufficient to form a polypeptide/compound complex, wherein the complex drives expression of a reporter gene sequence in the cell; and detecting the complex by detecting the reporter gene sequence expression such that if expression of the reporter gene is detected the compound the binds to a polypeptide of the invention is identified.




The methods of the invention also provides methods for treatment which involve the administration of the polynucleotides or polypeptides of the invention to individuals exhibiting symptoms or tendencies. In addition, the invention encompasses methods for treating diseases or disorders as recited herein comprising administering compounds and other substances that modulate the overall activity of the target gene products. Compounds and other substances can effect such modulation either on the level of target gene/protein expression or target protein activity.




The polypeptides of the present invention and the polynucleotides encoding them are also useful for the same functions known to one of skill in the art as the polypeptides and polynucleotides to which they have homology (set forth in Table 2); for which they have a signature region (as set forth in Table 3); or for which they have homology to a gene family (as set forth in Table 4). If no homology is set forth for a sequence, then the polypeptides and polynucleotides of the present invention are useful for a variety of applications, as described herein, including use in arrays for detection.











3. DETAILED DESCRIPTION OF THE INVENTION




3.1 Definitions




It must be noted that as used herein and in the appended claims, the singular forms “a”, “an” and “the” include plural references unless the context clearly dictates otherwise.




The term “active” refers to those forms of the polypeptide which retain the biologic and/or immunologic activities of any naturally occurring polypeptide. According to the invention, the terms “biologically active” or “biological activity” refer to a protein or peptide having structural, regulatory or biochemical functions of a naturally occurring molecule. Likewise “immunologically active” or “immunological activity” refers to the capability of the natural, recombinant or synthetic polypeptide to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.




The term “activated cells” as used in this application are those cells which are engaged in extracellular or intracellular membrane trafficking, including the export of secretory or enzymatic molecules as part of a normal or disease process.




The terms “complementary” or “complementarity” refer to the natural binding of polynucleotides by base pairing. For example, the sequence 5′-AGT-3′ binds to the complementary sequence 3′-TCA-5′. Complementarity between two single-stranded molecules may be “partial” such that only some of the nucleic acids bind or it may be “complete” such that total complementarity exists between the single stranded molecules. The degree of complementarity between the nucleic acid strands has significant effects on the efficiency and strength of the hybridization between the nucleic acid strands.




The term “embryonic stem cells (ES)” refers to a cell that can give rise to many differentiated cell types in an embryo or an adult, including the germ cells. The term “germ line stem cells (GSCs)” refers to stem cells derived from primordial stem cells that provide a steady and continuous source of germ cells for the production of gametes. The term “primordial germ cells (PGCs)” refers to a small population of cells set aside from other cell lineages particularly from the yolk sac, mesenteries, or gonadal ridges during embryogenesis that have the potential to differentiate into germ cells and other cells. PGCs are the source from which GSCs and ES cells are derived The PGCs, the GSCs and the ES cells are capable of self-renewal. Thus these cells not only populate the germ line and give rise to a plurality of terminally differentiated cells that comprise the adult specialized organs, but are able to regenerate themselves.




The term “expression modulating fragment,” EMF, means a series of nucleotides which modulates the expression of an operably linked ORF or another EMF.




As used herein, a sequence is said to “modulate the expression of an operably linked sequence” when the expression of the sequence is altered by the presence of the EMF. EMFs include, but are not limited to, promoters, and promoter modulating sequences (inducible elements). One class of EMFs are nucleic acid fragments which induce the expression of an operably linked ORF in response to a specific regulatory factor or physiological event.




The terms “nucleotide sequence” or “nucleic acid” or “polynucleotide” or “oligonculeotide” are used interchangeably and refer to a heteropolymer of nucleotides or the sequence of these nucleotides. These phrases also refer to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA) or to any DNA-like or RNA-like material. In the sequences herein A is adenine, C is cytosine, T is thymine, G is guanine and N is A, C, G or T (U). It is contemplated that where the polynucleotide is RNA, the T (thymine) in the sequences provided herein is substituted with U (uracil). Generally, nucleic acid segments provided by this invention may be assembled from fragments of the genome and short oligonucleotide linkers, or from a series of oligonucleotides, or from individual nucleotides, to provide a synthetic nucleic acid which is capable of being expressed in a recombinant transcriptional unit comprising regulatory elements derived from a microbial or viral operon, or a eukaryotic gene.




The terms “oligonucleotide fragment” or a “polynucleotide fragment”, “portion,” or “segment” or “probe” or “primer” are used interchangeably and refer to a sequence of nucleotide residues which are at least about 5 nucleotides, more preferably at least about 7 nucleotides, more preferably at least about 9 nucleotides, more preferably at least about 11 nucleotides and most preferably at least about 17 nucleotides. The fragment is preferably less than about 500 nucleotides, preferably less than about 200 nucleotides, more preferably less than about 100 nucleotides, more preferably less than about 50 nucleotides and most preferably less than 30 nucleotides. Preferably the probe is from about 6 nucleotides to about 200 nucleotides, preferably from about 15 to about 50 nucleotides, more preferably from about 17 to 30 nucleotides and most preferably from about 20 to 25 nucleotides. Preferably the fragments can be used in polymerase chain reaction (PCR), various hybridization procedures or microarray procedures to identify or amplify identical or related parts of mRNA or DNA molecules. A fragment or segment may uniquely identify each polynucteotide sequence of the present invention. Preferably the fragment comprises a sequence substantially similar to any one of SEQ ID NOs: 1-948.




Probes may, for example, be used to determine whether specific mRNA molecules are present in a cell or tissue or to isolate similar nucleic acid sequences from chromosomal DNA as described by Walsh et al. (Walsh, P. S. et al., 1992, PCR Methods Appl 1:241-250). They may be labeled by nick translation, Klenow fill-in reaction, PCR, or other methods well known in the art. Probes of the present invention, their preparation and/or labeling are elaborated in Sambrook, J. et al., 1989, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, NY; or Ausubel, F. M. et al., 1989, Current Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., both of which are incorporated herein by reference in their entirety.




The nucleic acid sequences of the present invention also include the sequence information from the nucleic acid sequences of SEQ ID NOs: 1-948. The sequence information can be a segment of any one of SEQ ID NOs: 1-948 that uniquely identifies or represents the sequence information of that sequence of SEQ ID NO: 1-948. One such segment can be a twenty-mer nucleic acid sequence because the probability that a twenty-mer is fully matched in the human genome is 1 in 300. In the human genome, there are three billion base pairs in one set of chromosomes. Because 4


20


possible twenty-mers exist, there are 300 times more twenty-mers than there are base pairs in a set of human chromosomes. Using the same analysis, the probability for a seventeen-mer to be fully matched in the human genome is approximately 1 in 5. When these segments are used in arrays for expression studies, fifteen-mer segments can be used. The probability that the fifteen-mer is fully matched in the expressed sequences is also approximately one in five because expressed sequences comprise less than approximately 5% of the entire genome sequence.




Similarly, when using sequence information for detecting a single mismatch, a segment can be a twenty-five mer. The probability that the twenty-five mer would appear in a human genome with a single mismatch is calculated by multiplying the probability for a full match (1÷4


25


) times the increased probability for mismatch at each nucleotide position (3×25). The probability that an eighteen mer with a single mismatch can be detected in an array for expression studies is approximately one in five. The probability that a twenty-mer with a single mismatch can be detected in a human genome is approximately one in five.




The term “open reading frame,” ORF, means a series of nucleotide triplets coding for amino acids without any termination codons and is a sequence translatable into protein.




The terms “operably linked” or “operably associated” refer to functionally related nucleic acid sequences. For example, a promoter is operably associated or operably linked with a coding sequence if the promoter controls the transcription of the coding sequence. While operably linked nucleic acid sequences can be contiguous and in the same reading friame, certain genetic elements e.g. repressor genes are not contiguously linked to the coding sequence but still control transcription/translation of the coding sequence.




The term “pluripotent” refers to the capability of a cell to differentiate into a number of differentiated cell types that are present in an adult organism. A pluripotent cell is restricted in its differentiation capability in comparison to a totipotent cell.




The terms “polypeptide” or “peptide” or “amino acid sequence” refer to an oligopeptide, peptide, polypeptide or protein sequence or fragment thereof and to naturally occurring or synthetic molecules. A polypeptide “fragment,” “portion,” or “segment” is a stretch of amino acid residues of at least about 5 amino acids, preferably at least about 7 amino acids, more preferably at least about 9 amino acids and most preferably at least about 17 or more amino acids. The peptide preferably is not greater than about 200 amino acids, more preferably less than 150 amino acids and most preferably less than 100 amino acids. Preferably the peptide is from about 5 to about 200 amino acids. To be active, any polypeptide must have sufficient length to display biological and/or immunological activity.




The term “naturally occurring polypeptide” refers to polypeptides produced by cells that have not been genetically engineered and specifically contemplates various polypeptides arising from post-translational modifications of the polypeptide including, but not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation.




The term “translated protein coding portion” means a sequence which encodes for the full length protein which may include any leader sequence or any processing sequence.




The term “mature protein coding sequence” means a sequence which encodes a peptide or protein without a signal or leader sequence. The “mature protein portion” means that portion of the protein which does not include a signal or leader sequence. The peptide may have been produced by processing in the cell which removes any leader/signal sequence. The mature protein portion may or may not include the initial methionine residue. The methionine residue may be removed from the protein during processing in the cell. The peptide may be produced synthetically or the protein may have been produced using a polynucleotide only encoding for the mature protein coding sequence.




The term “derivative” refers to polypeptides chemically modified by such techniques as ubiquitination, labeling (e.g., with radionuclides or various enzymes), covalent polymer attachment such as pegylation (derivatization with polyethylene glycol) and insertion or substitution by chemical synthesis of amino acids such as omithine, which do not normally occur in human proteins.




The term “variant” (or “analog”) refers to any polypeptide differing from naturally occurring polypeptides by amino acid insertions, deletions, and substitutions, created using, e g.; recombinant DNA techniques. Guidance in determining which amino acid residues may be replaced, added or deleted without abolishing activities of interest, may be found by comparing the sequence of the particular polypeptide with that of homologous peptides and minimizing the number of amino acid sequence changes made in regions of high homology (conserved regions) or by replacing amino acids with consensus sequence.




Alternatively, recombinant variants encoding these same or similar polypeptides may be synthesized or selected by making use of the “redundancy” in the genetic code. Various codon substitutions, such as the silent changes which produce various restriction sites, may be introduced to optimize cloning into a plasmid or viral vector or expression in a particular prokaryotic or eukaryotic system. Mutations in the polynucleotide sequence may be reflected in the polypeptide or domains of other peptides added to the polypeptide to modify the properties of any part of the polypeptide, to change characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate.




Preferably, amino acid “substitutions” are the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, i.e., conservative amino acid replacements. “Conservative” amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. “Insertions” or “deletions” are preferably in the range of about 1 to 20 amino acids, more preferably 1 to 10 amino acids. The variation allowed may be experimentally determined by systematically malking insertions, deletions, or substitutions of amino acids in a polypeptide molecule using recombinant DNA techniques and assaying the resulting recombinant variants for activity.




Alternatively, where alteration of function is desired, insertions, deletions or non-conservative alterations can be engineered to produce altered polypeptides. Such alterations can, for example, alter one or more of the biological functions or biochemical characteristics of the polypeptides of the invention. For example, such alterations may change polypeptide characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate. Further, such alterations can be selected so as to generate polypeptides that are better suited for expression, scale up and the like in the host cells chosen for expression. For example, cysteine residues can be deleted or substituted with another amino acid residue in order to eliminate disulfide bridges.




The terms “purified” or “substantially purified” as used herein denotes that the indicated nucleic acid or polypeptide is present in the substantial absence of other biological macromolecules, e.g., polynucleotides, proteins, and the like. In one embodiment, the polynucleotide or polypeptide is purified such that it constitutes at least 95% by weight, more preferably at least 99% by weight, of the indicated biological macromolecules present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 1000 daltons, can be present).




The term “isolated” as used herein refers to a nucleic acid or polypeptide separated from at least one other component (e.g., nucleic acid or polypeptide) present with the nucleic acid or polypeptide in its natural source. In one embodiment, the nucleic acid or polypeptide is found in the presence of (if anything) only a solvent, buffer, ion, or other component normally present in a solution of the same. The terms “isolated” and “purified” do not encompass nucleic acids or polypeptides present in their natural source.




The term “recombinant,” when used herein to refer to a polypeptide or protein, means that a polypeptide or protein is derived from recombinant (e.g., microbial, insect, or mammalian) expression systems. “Microbial” refers to recombinant polypeptides or proteins made in bacterial or fungal (e.g., yeast) expression systems. As a product, “recombinant microbial” defines a polypeptide or protein essentially free of native endogenous substances and unaccompanied by associated native glycosylation. Polypeptides or proteins expressed in most bacterial cultures, e.g.,


E. coli


, will be free of glycosylation modifications; polypeptides or proteins expressed in yeast will have a glycosylation pattern in general different from those expressed in mammalian cells.




The term “recombinant expression vehicle or vector” refers to a plasmid or phage or virus or vector, for expressing a polypeptide from a DNA (RNA) sequence. An expression vehicle can comprise a transcriptional unit comprising an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription initiation and termiination sequences. Structural units intended for use in yeast or eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell. Alternatively, where recombinant protein is expressed without a leader or transport sequence, it may include an amino terminal methionine residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final product.




The term “recombinant expression system” means host cells which have stably integrated a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit extrachromosomally. Recombinant expression systems as defined herein will express heterologous polypeptides or proteins upon induction of the regulatory elements linked to the DNA segment or synthetic gene to be expressed. This term also means host cells which have stably integrated a recombinant genetic element or elements having a regulatory role in gene expression, for example, promoters or enhancers. Recombinant expression systems as defined herein will express polypeptides or proteins endogenous to the cell upon induction of the regulatory elements linked to the endogenous DNA segment or gene to be expressed. The cells can be prokaryotic or eukaryotic.




The term “secreted” includes a protein that is transported across or through a membrane, including transport as a result of signal sequences in its amino acid sequence when it is expressed in a suitable host cell. “Secreted” proteins include without limitation proteins secreted wholly (e.g., soluble proteins) or partially (e.g., receptors) from the cell in which they are expressed. “Secreted” proteins also include without limitation proteins that are transported across the membrane of the endoplasmic reticulum. “Secreted” proteins are also intended to include proteins containing non-typical signal sequences (e.g. Interleukin-1 Beta, see Krasney, P. A. and Young, P. R. (1992) Cytokine 4(2):134-143) and factors released from damaged cells (e.g. Interleukin-1 Receptor Antagonist, see Arend, W. P. et. al. (1998) Annu. Rev. Immunol. 16:27-55)




Where desired, an expression vector may be designed to contain a “signal or leader sequence” which will direct the polypeptide through the membrane of a cell. Such a sequence may be naturally present on the polypeptides of the present invention or provided from heterologous protein sources by recombinant DNA techniques.




The term “stringent” is used to refer to conditions that are commonly understood in the art as stringent. Stringent conditions can include highly stringent conditions (i.e., hybridization to filter-bound DNA in 0.5 M NaHPO


4


, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65° C., and washing in 0.1×SSC/0.1% SDS at 68° C.), and moderately stringent conditions (i.e., washing in 0.2×SSC/0.1% SDS at 42° C.). Other exemplary hybridization conditions are described herein in the examples.




In instances of hybridization of deoxyoligonucleotides, additional exemplary stringent hybridization conditions include washing in 6×SSC/0.05% sodium pyrophosphate at 37° C. (for 14-base oligonucleotides), 48° C. (for 17-base oligos), 55° C. (for 20-base oligonucleotides), and 60° C. (for 23-base oligonucleotides).




As used herein, “substantially equivalent” or “substantially similar” can refer both to nucleotide and amino acid sequences, for example a mutant sequence, that varies from a reference sequence by one or more substitutions, deletions, or additions, the net effect of which does not result in an adverse functional dissimilarity between the reference and subject sequences. Typically, such a substantially equivalent sequence varies from one of those listed herein by no more than about 35% (i.e., the number of individual residue substitutions, additions, and/or deletions in a substantially equivalent sequence, as compared to the corresponding reference sequence, divided by the total number of residues in the substantially equivalent sequence is about 0.35 or less). Such a sequence is said to have 65% sequence identity to the listed sequence. In one embodiment, a substantially equivalent, e.g., mutant, sequence of the invention varies from a listed sequence by no more than 30% (70% sequence identity); in a variation of this embodiment, by no more than 25% (75% sequence identity); and in a further variation of this embodiment, by no more than 20% (80% sequence identity) and in a further variation of this embodiment, by no more than 10% (90% sequence identity) and in a further variation of this embodiment, by no more that 5% (95% sequence identity). Substantially equivalent, e.g., mutant, amino acid sequences according to the invention preferably have at least 80% sequence identity with a listed amino acid sequence, more preferably at least 85% sequence identity, more preferably at least 90% sequence identity, more preferably at least 95% sequence identity, more preferably at least 98% sequence identity, and most preferably at least 99% sequence identity. Substantially equivalent nucleotide sequence of the invention can have lower percent sequence identities, taking into account, for example, the redundancy or degeneracy of the genetic code. Preferably, the nucleotide sequence has at least about 65% identity, more preferably at least about 75% identity, more preferably at least about 80% sequence identity, more preferably at least 85% sequence identity, more preferably at least 90% sequence identity, more preferably at least about 95% sequence identity, more preferably at least 98% sequence identity, and most preferably at least 99% sequence identity. For the purposes of the present invention, sequences having substantially equivalent biological activity and substantially equivalent expression characteristics are considered substantially equivalent. For the purposes of determining equivalence, truncation of the mature sequence (e.g., via a mutation which creates a spurious stop codon) should be disregarded. Sequence identity may be determined, e.g., using the Jotun Hein method (Hein, J. (1990) Methods Enzymol. 183:626-645). Identity between sequences can also be determined by other methods known in the art, e.g. by varying hybridization conditions.




The term “totipotent” refers to the capability of a cell to differentiate into all of the cell types of an adult organism.




The term “transformation” means introducing DNA into a suitable host cell so that the DNA is replicable, either as an extrachromosomal element, or by chromosomal integration. The term “transfection” refers to the taking up of an expression vector by a suitable host cell, whether or not any coding sequences are in fact expressed. The term “infection” refers to the introduction of nucleic acids into a suitable host cell by use of a virus or viral vector.




As used herein, an “uptake modulating fragment,” UMF, means a series of nucleotides which mediate the uptake of a linked DNA fragment into a cell. UMFs can be readily identified using known UMFs as a target sequence or target motif with the computer-based systems described below. The presence and activity of a UMF can be confirmed by attaching the suspected UMF to a marker sequence. The resulting nucleic acid molecule is then incubated with an appropriate host under appropriate conditions and the uptake of the marker sequence is determined. As described above, a UMF will increase the frequency of uptake of a linked marker sequence.




Each of the above terms is meant to encompass all that is described for each, unless the context dictates otherwise.




3.2 Nucleic Acids of the Invention




Nucleotide sequences of the invention are set forth in the Sequence Listing.




The isolated polynucleotides of the invention include a polynucleotide comprising the nucleotide sequences of SEQ ID NO: 1-948; a polynucleotide encoding any one of the peptide sequences of SEQ ID NO: 1-948; and a polynucleotide comprising the nucleotide sequence encoding the mature protein coding sequence of the polynucleotides of any one of SEQ ID NO: 1-948. The polynucleotides of the present invention also include, but are not limited to, a polynucleotide that hybridizes under stringent conditions to (a) the complement of any of the nucleotides sequences of SEQ ID NO: 1-948; (b) nucleotide sequences encoding any one of the amino acid sequences set forth in the Sequence Listing; (c) a polynucleotide which is an allelic variant of any polynucleotide recited above; (d) a polynucleotide which encodes a species homolog of any of the proteins recited above; or (c) a polynucleotide that encodes a polypeptide comprising a specific domain or truncation of the polypeptides of SEQ ID NO: 1-948. Domains of interest may depend on the nature of the encoded polypeptide; e.g., domains in receptor-like polypeptides include ligand-binding, extracellular, transmembrane, or cytoplasmic domains, or combinations thereof; domains in immunoglobulin-like proteins include the variable immunoglobulin-like domains; domains in enzyme-like polypeptides include catalytic and substrate binding domains; and domains in ligand polypeptides include receptor-binding domains.




The polynucleotides of the invention include naturally occurring or wholly or partially synthetic DNA, e.g., cDNA and genomic DNA, and RNA, e.g., mRNA. The polynucleotides may include all of the coding region of the cDNA or may represent a portion of the coding region of the cDNA.




The present invention also provides genes corresponding to the cDNA sequences disclosed herein. The corresponding genes can be isolated in accordance with known methods using the sequence information disclosed herein Such methods include the preparation of probes or primers from the disclosed sequence information for identification and/or amplification of genes in appropriate genomic libraries or other sources of genomic materials. Further 5′ and 3′ sequence can be obtained using methods known in the art. For example, full length cDNA or genomic DNA that corresponds to any of the polynucleotides of SEQ ID NO: 1-948 can be obtained by screening appropriate cDNA or genomic DNA libraries under suitable hybridization conditions using any of the polynucleotides of SEQ ID NO: 1-948 or a portion thereof as a probe. Alternatively, the polynucleotides of SEQ ID NO: 1-948 may be used as the basis for suitable primer(s) that allow identification and/or amplification of genes in appropriate genomic DNA or cDNA libraries.




The nucleic acid sequences of the invention can be assembled from ESTs and sequences (including cDNA and genomic sequences) obtained from one or more public databases, such as dbEST, gbpri, and UniGene. The EST sequences can provide identifying sequence information, representative fragment or segment information, or novel segment information for the full-length gene.




The polynucleotides of the invention also provide polynucleotides including nucleotide sequences that are substantially equivalent to the polynucleotides recited above. Polynucleotides according to the invention can have, e.g., at least about 65%, at least about 70%, at least about 75%, at least about 80%, 81%, 82%, 83%, 84%, more typically at least about 85%, 86%, 87%, 88%, 89%, more typically at least about 90%, 91%, 92%, 93%, 94%, and even more typically at least about 95%, 96%, 97%, 98%, 99% sequence identity to a polynucleotide recited above.




Included within the scope of the nucleic acid sequences of the invention are nucleic acid sequence fragments that hybridize under stringent conditions to any of the nucleotide sequences of SEQ ID NO: 1-948, or complements thereof, which fragment is greater than about 5 nucleotides, preferably 7 nucleotides, more preferably greater than 9 nucleotides and most preferably greater than 17 nucleotides. Fragments of, e.g. 15, 17, or 20 nucleotides or more that are selective for (i.e. specifically hybridize to any one of the polynucleotides of the invention) are contemplated. Probes capable of specifically hybridizing to a polynucleotide can differentiate polynucleotide sequences of the invention from other polynucleotide sequences in the same family of genes or can differentiate human genes from genes of other species, and are preferably based on unique nucleotide sequences.




The sequences falling within the scope of the present invention are not limited to these specific sequences, but also include allelic and species variations thereof. Allelic and species variations can be routinely determined by comparing the sequence provided in SEQ ID NO: 1-948, a representative fragment thereof, or a nucleotide sequence at least 90% identical, preferably 95% identical, to SEQ ID NOs: 1-948 with a sequence from another isolate of the same species. Furthermore, to accommodate codon variability, the invention includes nucleic acid molecules coding for the same amino acid sequences as do the specific ORFs disclosed herein. In other words, in the coding region of an ORF, substitution of one codon for another codon that encodes the same amino acid is expressly contemplated.




The nearest neighbor or homology result for the nucleic acids of the present invention, including SEQ ID NOs: 1-948, can be obtained by searching a database using an algorithm or a program. Preferably, a BLAST which stands for Basic Local Alignment Search Tool is used to search for local sequence alignments (Altshul, S. F. J Mol. Evol. 36 290-300 (1993) and Altschul S. F. et al. J. Mol. Biol. 21:403-410 (1990)). Alternatively a FASTA version 3 search against Genpept, using Fastxy algorithm.




Species homologs (or orthologs) of the disclosed polynucleotides and proteins are also provided by the present invention. Species homologs may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source from the desired species.




The invention also encompasses allelic variants of the disclosed polynucleotides or proteins; that is, naturally-occurring alternative forms of the isolated polynucleotide which also encode proteins which are identical, homologous or related to that encoded by the polynucleotides.




The nucleic acid sequences of the invention are further directed to sequences which encode variants of the described nucleic acids. These amino acid sequence variants may be prepared by methods known in the art by introducing appropriate nucleotide changes into a native or variant polynucleotide. There are two variables in the construction of amino acid sequence variants: the location of the mutation and the nature of the mutation. Nucleic acids encoding the amino acid sequence variants are preferably constructed by mutating the polynucleotide to encode an amino acid sequence that does not occur in nature. These nucleic acid alterations can be made at sites that differ in the nucleic acids from different species (variable positions) or in highly conserved regions (constant regions). Sites at such locations will typically be modified in series, e.g., by substituting first with conservative choices (e.g., hydrophobic amino acid to a different hydrophobic amino acid) and then with more distant choices (e.g., hydrophobic amino acid to a charged amino acid), and then deletions or insertions may be made at the target site. Amino acid sequence deletions generally range from about 1 to 30 residues, preferably about 1 to 10 residues, and are typically contiguous. Amino acid insertions include amino- and/or carboxyl-terminal fusions ranging in length from one to one hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Intrasequence insertions may range generally from about 1 to 10 amino residues, preferably from 1 to 5 residues. Examples of terminal insertions include the heterologous signal sequences necessary for secretion or for intracellular targeting in different host cells and sequences such as FLAG or poly-histidine sequences useful for purifying the expressed protein.




In a preferred method, polynucleotides encoding the novel amino acid sequences are changed via site-directed mutagenesis. This method uses oligonucleotide sequences to alter a polynucleotide to encode the desired amino acid variant, as well as sufficient adjacent nucleotides on both sides of the changed amino acid to form a stable duplex on either side of the site of being changed. In general, the techniques of site-directed mutagenesis are well known to those of skill in the art and this technique is exemplified by publications such as, Edelman et al.,


DNA


2:183 (1983). A versatile and efficient method for producing site-specific changes in a polynucleotide sequence was published by Zoller and Smith,


Nucleic Acids Res


. 10:6487-6500 (1982). PCR may also be used to create amino acid sequence variants of the novel nucleic acids. When small amounts of template DNA are used as starting material, primer(s) that differs slightly in sequence from the corresponding region in the template DNA can generate the desired amino acid variant. PCR amplification results in a population of product DNA fragments that differ from the polynucleotide template encoding the polypeptide at the position specified by the primer. The product DNA fragments replace the corresponding region in the plasmid and this gives a polynucleotide encoding the desired amino acid variant.




A further technique for generating amino acid variants is the cassette mutagenesis technique described in Wells et al.,


Gene


34:315 (1985); and other mutagenesis techniques well known in the art, such as, for example, the techniques in Sambrook et al., supra, and


Current Protocols in Molecular Biology


, Ausubel et al. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be used in the practice of the invention for the cloning and expression of these novel nucleic acids. Such DNA sequences include those which are capable of hybridizing to the appropriate novel nucleic acid sequence under stringent conditions.




Polynucleotides encoding preferred polypeptide truncations of the invention can be used to generate polynucleotides encoding chimeric or fusion proteins comprising one or more domains of the invention and heterologous protein sequences.




The polynucleotides of the invention additionally include the complement of any of the polynucleotides recited above. The polynucleotide can be DNA (genomic, cDNA, amplified, or synthetic) or RNA. Methods and algorithms for obtaining such polynucleotides are well known to those of skill in the art and can include, for example, methods for determining hybridization conditions that can routinely isolate polynucleotides of the desired sequence identities.




In accordance with the invention, polynucleotide sequences comprising the mature protein coding sequences corresponding to any one of SEQ ID NO: 1-948, or functional equivalents thereof, may be used to generate recombinant DNA molecules that direct the expression of that nucleic acid, or a functional equivalent thereof, in appropriate host cells. Also included are the cDNA inserts of any of the clones identified herein.




A polynucleotide according to the invention can be joined to any of a variety of other nucleotide sequences by well-established recombinant DNA techniques (see Sambrook J et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, NY). Useful nucleotide sequences for joining to polynucleotides include an assortment of vectors, e.g., plasmids, cosmids, lambda phage derivatives, phagemids, and the like, that are well known in the art. Accordingly, the invention also provides a vector including a polynucleotide of the invention and a host cell containing the polynucleotide. In general, the vector contains an origin of replication functional in at least one organism, convenient restriction endonuclease sites, and a selectable marker for the host cell. Vectors according to the invention include expression vectors, replication-vectors, probe generation vectors, and sequencing vectors. A host cell according to the invention can be a prokaryotic or eukaryotic cell and can be a unicellular organism or part of a multicellular organism.




The present invention further provides recombinant constructs comprising a nucleic acid having any of the nucleotide sequences of SEQ ID NOs: 1-948 or a fragment thereof or any other polynucleotides of the invention. In one embodiment, the recombinant constructs of the present invention comprise a vector, such as a plasmid or viral vector, into which a nucleic acid having any of the nucleotide sequences of SEQ ID NOs: 1-948 or a fragment thereof is inserted, in a forward or reverse orientation. In the case of a vector comprising one of the ORFs of the present invention, the vector may further comprise regulatory sequences, including for example, a promoter, operably linked to the ORF. Large numbers of suitable vectors and promoters are known to those of skill in the art and are commercially available for generating the recombinant constructs of the present invention. The following vectors are provided by way of example. Bacterial: pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene); pTrc99A, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLneo, pSV2cat, pOG44, PXTI, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia).




The isolated polynucleotide of the invention may be operably linked to an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufmnan et al.,


Nucleic Acids Res


. 19, 4485-4490 (1991), in order to produce the protein recombinantly. Many suitable expression control sequences are known in the art. General methods of expressing recombinant proteins are also known and are exemplified in R. Kaufman,


Methods in Enzymology


185, 537-566 (1990). As defined herein “operably linked” means that the isolated polynucleotide of the invention and an expression control sequence are situated within a vector or cell in such a way that the protein is expressed by a host cell which has been transformed (transfected) with the ligated polynucleotide/expression control sequence.




Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lac, lacZ, T3, T7, gpt, lambda PR, and trc. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art. Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of


E. coli


and


S. cerevisiae


TRP 1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an amino terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product. Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include


E. coli, Bacillus subtilis, Salmonella typhimurium


and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.




As a representative but non-limiting example, usefull expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM 1 (Promega Biotech, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced or derepressed by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period. Cells are typically harvested by centrimugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.




Polynucleotides of the invention can also be used to induce immune responses. For example, as described in Fan et al.,


Nat. Biotech


. 17:870-872 (1999), incorporated herein by reference, nucleic acid sequences encoding a polypeptide may be used to generate antibodies against the encoded polypeptide following topical administration of naked plasmid DNA or following injection, and preferably intra-muscular injection of the DNA. The nucleic acid sequences are preferably inserted in a recombinant expression vector and may be in the form of naked DNA.




3.3 Antisense




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: 1-948, 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 coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of a protein of any of SEQ ID NO: 1-948 or antisense nucleic acids complementary to a nucleic acid sequence of SEQ ID NO: 1-948 are additionally provided.




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




Given the coding strand sequences encoding a nucleic acid disclosed herein (e.g., SEQ ID NO: 1-948, 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 an mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of an mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of an 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 nucleicacid (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.




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).




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 protein according to the invention 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.




In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual α-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. (1 987)


FEBS Lett


215: 327-330).




3.4 Ribozymes and PNA Moieties




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 mRNA transcripts to thereby inhibit translation of an mRNA. A ribozyme having specificity for a nucleic acid of the invention can be designed based upon the nucleotide sequence of a DNA disclosed herein (ie., SEQ ID NO: 1-948). For example, a derivative of 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 SECX-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, SECX 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.




Alternatively, gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region (e.g., promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene in target cells. See generally, Helene. (1991)


Anticancer Drug Des


. 6: 569-84; Helene. et al. (1992)


Ann. N.Y. Acad. Sci


. 660:27-36; and Maher (1992)


Bioassays


14: 807-15.




In various embodiments, the nucleic acids of the invention 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.




PNAs of the invention 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 the invention 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 irestriction 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).




In another embodiment, PNAs of the invention 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 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.




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. W088/09810) or the blood-brain barrier (see, e.g., PCT Publication No. W089/10134). 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.




3.5 Hosts




The present invention further provides host cells genetically engineered to contain the polynucleotides of the invention. For example, such host cells may contain nucleic acids of the invention introduced into the host cell using known transformation, transfection or infection methods. The present invention still further provides host cells genetically engineered to express the polynucleotides of the invention, wherein such polynucleotides are in operative association with a regulatory sequence heterologous to the host cell which drives expression of the polynucleotides in the cell.




Knowledge of nucleic acid sequences allows for modification of cells to permit, or increase, expression of endogenous polypeptide. Cells can be modified (e.g., by homologous recombination) to provide increased polypeptide expression by replacing, in whole or in part, the naturally occurring promoter with all or part of a heterologous promoter so that the cells express the polypeptide at higher levels. The heterologous promoter is inserted in such a manner that it is operatively linked to the encoding sequences. See, for example, PCT International Publication No. WO94/12650, PCT International Publication No. WO92/20808, and PCT International Publication No. WO91/09955. It is also contemplated that, in addition to heterologous promoter DNA, amplifiable marker DNA (e.g., ada, dhfr, and the multifunctional CAD gene which encodes carbamyl phosphate synthase, aspartate transcarbamylase, and dihydroorotase) and/or intron DNA may be inserted along with the heterologous promoter DNA. If linked to the coding sequence, amplification of the marker DNA by standard selection methods results in co-amplification of the desired protein coding sequences in the cells.




The host cell can be a higher eukaryotic host cell, such as a mammalian cell, a lower eukaryotic host cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the recombinant construct into the host cell can be effected by calcium phosphate transfection, DEAE, dextran mediated transfection, or electroporation Davis, L. et al.,


Basic Methods in Molecular Biology


(1986)). The host cells containing one of the polynucleotides of the invention, can be used in conventional manners to produce the gene product encoded by the isolated fragment (in the case of an ORF) or can be used to produce a heterologous protein under the control of the EMF.




Any host/vector system can be used to express one or more of the ORFs of the present invention. These include, but are not limited to, eukaryotic hosts such as HeLa cells, Cv-1 cell, COS cells, 293 cells, and Sf9 cells, as well as prokaryotic host such as


E. coli


and


B. subtilis


. The most preferred cells are those which do not normally express the particular polypeptide or protein or which expresses the polypeptide or protein at low natural level. Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., in Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y. (1989), the disclosure of which is hereby incorporated by reference.




Various mammalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23:175 (1981). Other cell lines capable of expressing a compatible vector are, for example, the C127, monkey COS cells, Chinese Hamster Ovary (CHO) cells, human kidney 293 cells, human epidermal A431 cells, human Colo205 cells, 3T3 cells, CV-1 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HeLa cells, mouse L cells, BHK, HL60, U937, HaK or Jurkat cells. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking nontranscribed sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements. Recombinant polypeptides and proteins produced in bacterial culture are usually isolated by initial extraction from cell pellets, followed by one or more salting-out, aqueous ion exchange or size exclusion chromatography steps. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for fmal purification steps. Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.




Alternatively, it may be possible to produce the protein in lower eukaryotes such as yeast or insects or in prokaryotes such as bacteria. Potentially suitable yeast strains include


Saccharomyces cerevisiae, Schizosaccharomyces pombe


, Kluyveromyces strains, Candida, or any yeast strain capable of expressing heterologous proteins. Potentially suitable bacterial strains include


Escherichia coli, Bacillus subtilis, Salmonella typhimurium


, or any bacterial strain capable of expressing heterologous proteins. If the protein is made in yeast or bacteria, it may be necessary to modify the protein produced therein, for example by phosphorylation or glycosylation of the appropriate sites, in order to obtain the functional protein. Such covalent attachments may be accomplished using known chemical or enzymatic methods.




In another embodiment of the present invention, cells and tissues may be engineered to express an endogenous gene comprising the polynucleotides of the invention under the control of inducible regulatory elements, in which case the regulatory sequences of the endogenous gene may be replaced by homologous recombination. As described herein, gene targeting can be used to replace a gene's existing regulatory region with a regulatory sequence isolated from a different gene or a novel regulatory sequence synthesized by genetic engineering methods. Such regulatory sequences may be comprised of promoters, enhancers, scaffold-attachment regions, negative regulatory elements, transcriptional initiation sites, regulatory protein binding sites or combinations of said sequences. Alternatively, sequences which affect the structure or stability of the RNA or protein produced may be replaced, removed, added, or otherwise modified by targeting. These sequence include polyadenylation signals, mRNA stability elements, splice sites, leader sequences for enhancing or modifying transport or secretion properties of the protein, or other sequences which alter or improve the fumction or stability of protein or RNA molecules.




The targeting event may be a simple insertion of the regulatory sequence, placing the gene under the control of the new regulatory sequence, e.g., inserting a new promoter or enhancer or both upstream of a gene. Alternatively, the targeting event may be a simple deletion of a regulatory element, such as the deletion of a tissue-specific negative regulatory element. Alternatively, the targeting event may replace an existing element; for example, a tissue-specific enhancer can be replaced by an enhancer that has broader or different cell-type specificity than the naturally occurring elements. Here, the naturally occurring sequences are deleted and new sequences are added. In all cases, the identification of the targeting event may be facilitated by the use of one or more selectable marker genes that are contiguous with the targeting DNA, allowing for the selection of cells in which the exogenous DNA has integrated into the host cell genome. The identification of the targeting event may also be facilitated by the use of one or more marker genes exhibiting the property of negative selection, such that the negatively selectable marker is linked to the exogenous DNA, but configured such that the negatively selectable marker flanks the targeting sequence, and such that a correct homologous recombination event with sequences in the host cell genome does not result in the stable integration of the negatively selectable marker. Markers useful for this purpose include the Herpes Simplex Virus thyrnidine kinase (TK) gene or the bacterial xanthine-guanine phosphoribosyl-transferase (gpt) gene.




The gene targeting or gene activation techniques which can be used in accordance with this aspect of the invention are more particularly described in U.S. Pat. No. 5,272,071 to Chappel; U.S. Pat. No. 5,578,461 to Sherwin et al.; International Application No. PCT/US92/09627 (WO93/09222) by Selden et al.; and International Application No. PCT/US90/06436 (WO91/06667) by Skoultchi et al., each of which is incorporated by reference herein in its entirety.




3.6 Polypeptides of the Invention




The isolated polypeptides of the invention include, but are not limited to, a polypeptide comprising: the amino acid sequences set forth as any one of SEQ ID NO: 1-948 or an amino acid sequence encoded by any one of the nucleotide sequences SEQ ID NOs: 1-948 or the corresponding fulll length or mature protein. Polypeptides of the invention also include polypeptides preferably with biological or immunological activity that are encoded by: (a) a polynucleotide having any one of the nucleotide sequences set forth in SEQ ID NOs: 1-948 or (b) polynucleotides encoding any one of the amino acid sequences set forth as SEQ ID NO: 1-948 or (c) polynucleotides that hybridize to the complement of the polynucleotides of either (a) or (b) under stringent hybridization conditions. The invention also provides biologically active or immunologically active variants of any of the amino acid sequences set forth as SEQ ID NO: 1-948 or the corresponding full length or mature protein; and “substantial equivalents” thereof (e.g., with at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, 86%, 87%, 88%, 89%, at least about 90%, 91%, 92%, 93%, 94%, typically at least about 95%, 96%, 97%, more typically at least about 98%, or most typically at least about 99% amino acid identity) that retain biological activity. Polypeptides encoded by allelic variants may have a similar, increased, or decreased activity compared to polypeptides comprising SEQ ID NO: 1-948.




Fragments of the proteins of the present invention which are capable of exhibiting biological activity are also encompassed by the present invention. Fragments of the protein may be in linear form or they may be cyclized using known methods, for example, as described in H. U. Saragovi, et al., Bio/Technology 10, 773-778 (1992) and in R. S. McDowell, et al., J. Amer. Chem. Soc. 114, 9245-9253 (1992), both of which are incorporated herein by reference. Such fragments may be fused to carrier molecules such as immunoglobulins for many purposes, including increasing the valency of protein binding sites.




The present invention also provides both full-length and mature forms (for example, without a signal sequence or precursor sequence) of the disclosed proteins. The protein coding sequence is identified in the sequence listing by translation of the disclosed nucleotide sequences. The mature form of such protein may be obtained by expression of a full-length polynuclcotide in a suitable mammalian cell or other host cell. The sequence of the mature form of the protein is also determinable from the amino acid sequence of the full-length form. Where proteins of the present invention are membrane bound, soluble forms of the proteins are also provided. In such forms, part or all of the regions causing the proteins to be membrane bound are deleted so that the proteins are fully secreted from the cell in which they are expressed.




Protein compositions of the present invention may further comprise an acceptable carrier, such as a hydrophilic, e.g., pharmaceutically acceptable, carrier.




The present invention further provides isolated polypeptides encoded by the nucleic acid fragments of the present invention or by degenerate variants of the nucleic acid fragments of the present invention. By “degenerate variant” is intended nucleotide fragments which differ from a nucleic acid fragment of the present invention (e.g., an ORF) by nucleotide sequence but, due to the degeneracy of the genetic code, encode an identical polypeptide sequence. Preferred nucleic acid fragments of the present invention are the ORFs that encode proteins.




A variety of methodologies kcnown in the art can be utilized to obtain any one of the isolated polypeptides or proteins of the present invention. At the simplest level, the amino acid sequence can be synthesized using commercially available peptide synthesizers. The synthetically-constructed protein sequences, by virtue of sharing primary, secondary or tertiary structural and/or conformational characteristics with proteins may possess biological properties in common therewith, including protein activity. This technique is particularly useful in producing small peptides and fragments of larger polypeptides. Fragments are useful, for example, in generating antibodies against the native polypeptide. Thus, they may be employed as biologically active or immunological substitutes for natural, purified proteins in screening of therapeutic compounds and in immunological processes for the development of antibodies.




The polypeptides and proteins of the present invention can alternatively be purified from cells which have been altered to express the desired polypeptide or protein. As used herein, a cell is said to be altered to express a desired polypeptide or protein when the cell, through genetic manipulation, is made to produce a polypeptide or protein which it normally does not produce or which the cell normally produces at a lower level. One skilled in the art can readily adapt procedures for introducing and expressing either recombinant or synthetic sequences into eukaryotic or prokaryotic cells in order to generate a cell which produces one of the polypeptides or proteins of the present invention.




The invention also relates to methods for producing a polypeptide comprising growing a culture of host cells of the invention in a suitable culture medium, and purifying the protein from the cells or the culture in which the cells are grown. For example, the methods of the invention include a process for producing a polypeptide in which a host cell containing a suitable expression vector that includes a polynucleotide of the invention is cultured under conditions that allow expression of the encoded polypeptide. The polypeptide can be recovered from the culture, conveniently from the culture medium, or from a lysate prepared from the host cells and further purified. Preferred embodiments include those in which the protein produced by such process is a full length or mature form of the protein.




In an alternative method, the polypeptide or protein is purified from bacterial cells which naturally produce the polypeptide or protein. One skilled in the art can readily follow known methods for isolating polypeptides and proteins in order to obtain one of the isolated polypeptides or proteins of the present invention. These include, but are not limited to, immunochromatography, HPLC, size-exclusion chromatography, ion-exchange chromatography, and immuno-affinity chromatography. See, e.g., Scopes,


Protein Purification: Principles and Practice


, Springer-Verlag (1994); Sambrook, et al., in Molecular Cloning:


A Laboratory Manual


; Ausubel et al.,


Current Protocols in Molecular Biology


. Polypeptide fragments that retain biological/immnunological activity include fragments comprising greater than about 100 amino acids, or greater than about 200 amino acids, and fragments that encode specific protein domains.




The purified polypeptides can be used in in vitro binding assays which are well known in the art to identify molecules which bind to the polypeptides. These molecules include but are not limited to, for e.g., small molecules, molecules from combinatorial libraries, antibodies or other proteins. The molecules identified in the binding assay are then tested for antagonist or agonist activity in in vivo tissue culture or animal models that are well known in the art. In brief, the molecules are titrated into a plurality of cell cultures or animals and then tested for either cell/animal death or prolonged survival of the animal/cells.




In addition, the peptides of the invention or molecules capable of binding to the peptides may be complexed with toxins, e.g., ricin or cholera, or with other compounds that are toxic to cells. The toxin-binding molecule complex is then targeted to a tumor or other cell by the specificity of the binding molecule for SEQ ID NO: 1-948.




The protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a nucleotide sequence encoding the protein.




The proteins provided herein also include proteins characterized by amino acid sequences similar to those of purified proteins but into which modification are naturally provided or deliberately engineered. For example, modifications, in the peptide or DNA sequence, can be made by those skilled in the art using known techniques. Modifications of interest in the protein sequences may include the alteration, substitution, replacement, insertion or deletion of a selected amino acid residue in the coding sequence. For example, one or more of the cysteine residues may be deleted or replaced with another amino acid to alter the conformation of the molecule. Techniques for such alteration, substitution, replacement, insertion or deletion are well known to those skilled in the art (see, e.g., U.S. Pat. No. 4,518,584). Preferably, such alteration, substitution, replacement, insertion or deletion retains the desired activity of the protein. Regions of the protein that are important for the protein function can be determined by various methods known in the art including the alanine-scanning method which involved systematic substitution of single or strings of amino acids with alanine, followed by testing the resulting alanine-containing variant for biological activity. This type of analysis determines the importance of the substituted amino acid(s) in biological activity. Regions of the protein that are important for protein function may be determined by the eMATRIX program.




Other fragments and derivatives of the sequences of proteins which would be expected to retain protein activity in whole or in part and are useful for screening or other immunological methodologies may also be easily made by those skilled in the art given the disclosures herein. Such modifications are encompassed by the present invention.




The protein may also be produced by operably linking the isolated polynucleotide of the invention to suitable control sequences in one or more insect expression vectors, and employing an insect expression system. Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, e.g., Invitrogen, San Diego, Calif., U.S.A. (the MaxBa™ kit), and such methods are well known in the art, as described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987), incorporated herein by reference. As used herein, an insect cell capable of expressing a polynucleotide of the present invention is “transformed.”




The protein of the invention may be prepared by culturing transformed host cells under culture conditions suitable to express the recombinant protein. The resulting expressed protein may then be purified from such culture (i.e., from culture medium or cell extracts) using known purification processes, such as gel filtration and ion exchange chromatography. The purification of the protein may also include an affinity column containing agents which will bind to the protein; one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl™ or Cibacrom blue 3GA Sepharose™; one or more steps involving hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or immunoaffinity chromatography.




Alternatively, the protein of the invention may also be expressed in a form which will facilitate purification. For example, it may be expressed as a fulsion protein, such as those of maltose binding protein (MBP), glutathione-S-transferase (GST) or thioredoxin (TRX), or as a His tag. Kits for expression and purification of such fusion proteins are commercially available from New England BioLab (Beverly, Mass.), Pharmacia (Piscataway, N.J.) and Invitrogen, respectively. The protein can also be tagged with an epitope and subsequently purified by using a specific antibody directed to such epitope. One such epitope (“FLAG®”) is commercially available from Kodak (New Haven, Conn.).




Finally, one or more reverse-phase high performance liquid chromatography (RP-BPLC) steps employing hydrophobic RP-HPLC media, e.g. silica gel having pendant methyl or other aliphatic groups, can be employed to further purify the protein. Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a substantially homogeneous isolated recombinant protein. The protein thus purified is substantially free of other mammalian proteins and is defined in accordance with the present invention as an “isolated protein.”




The polypeptides of the invention include analogs (variants). This embraces fragments, as well as peptides in which one or more amino acids has been deleted, inserted, or substituted. Also, analogs of the polypeptides of the invention embrace fusions of the polypeptides or modifications of the polypeptides of the invention, wherein the polypeptide or analog is fused to another moiety or moieties, e.g., targeting moiety or another therapeutic agent. Such analogs may exhibit improved properties such as activity and/or stability. Examples of moieties which may be fused to the polypeptide or an analog include, for example, targeting moieties which provide for the delivery of polypeptide to pancreatic cells, e.g., antibodies to pancreatic cells, antibodies to immune cells such as T-cells, monocytes, dendritic cells, granulocytes, etc., as well as receptor and ligands expressed on pancreatic or immune cells. Other moieties which may be fused to the polypeptide include therapeutic agents which are used for treatment, for example, immunosuppressive drugs such as cyclosporin, SK506, azathioprine, CD3 antibodies and steroids. Also, polypeptides may be fused to immune modulators, and other cytokines such as alpha or beta interferon.




3.6.1 Determining Polypeptide and Polynucleotide Identity and Similarity




Preferred identity and/or similarity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in computer programs including, but are not limited to, the GCG program package, including GAP (Devereux, J., et al., Nucleic Acids Research 12(1):387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, BLASTX, FASTA (Altschul, S. F. et al., J. Molec. Biol. 215:403-410 (1990), PSI-BLAST (Altschul S. F. et al., Nucleic Acids Res. vol. 25, pp. 3389-3402, herein incorporated by reference), eMatrix software (Wu et al., J. Comp. Biol., Vol. 6, pp. 219-235 (1999), herein incorporated by reference), eMotif software (Nevill-Manning et al, ISMB-97, Vol. 4, pp. 202-209, herein incorporated by reference), pFam software (Sornhammer et al., Nucleic Acids Res., Vol. 26(1), pp. 320-322 (1998), herein incorporated by reference) and the Kyte-Doolittle hydrophobocity prediction algorithm (J. Mol Biol, 157, pp. 105-31 (1982), incorporated herein by reference). The BLAST programs are publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul, S., et al. NCB NLM NIH Bethesda, Md. 20894; Altschul, S., et al., J. Mol. Biol. 215:403-410 (1990).




3.7 Chimeric and Fusion Proteins




The invention also provides chimeric or fusion proteins. As used herein, a “chimeric protein” or “fusion protein” comprises a polypeptide of the invention operatively linked to another polypeptide. Within a fusion protein the polypeptide according to the invention can correspond to all or a portion of a protein according to the invention. In one embodiment, a fusion protein comprises at least one biologically active portion of a protein according to the invention. In another embodiment, a fusion protein comprises at least two biologically active portions of a protein according to the invention. Within the fusion protein, the term “operatively linked” is intended to indicate that the polypeptide according to the invention and the other polypeptide are fused in-frame to each other. The polypeptide can be fused to the N-terminus or C-terminus, or to the middle.




For example, in one embodiment a fusion protein comprises a polypeptide according to the invention operably linked to the extracellular domain of a second protein.




In another embodiment, the fusion protein is a GST-fusion protein in which the polypeptide sequences of the invention are fused to the C-terminus of the GST (i.e., glutathione S-transferase) sequences.




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




A chimeric or fiusion 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.) C


URRENT


P


ROTOCOLS IN


M


OLECULAR


B


IOLOGY


, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the protein of the invention.




3.8 Gene Therapy




Mutations in the polynucleotides of the invention gene may result in loss of normal function of the encoded protein. The invention thus provides gene therapy to restore normal activity of the polypeptides of the invention; or to treat disease states involving polypeptides of the invention. Delivery of a functional gene encoding polypeptides of the invention to appropriate cells is effected ex vivo, in situ, or in vivo by use of vectors, and more particularly viral vectors (e.g., adenovirus, adeno-associated virus, or a retrovirus), or ex vivo by use of physical DNA transfer methods (e.g., liposomes or chemical treatments). See, for example, Anderson, Nature, supplement to vol. 392, no. 6679, pp.25-20 (1998). For additional reviews of gene therapy technology see Friedmann, Science, 244: 1275-1281 (1989); Verma, Scientific American: 68-84 (1990); and Miller, Nature, 357: 455460(1992). Introduction of any one of the nucleotides of the present invention or a gene encoding the polypeptides of the present invention can also be accomplished with extrachromosomal substrates (transient expression) or artificial chromosomes (stable expression). Cells may also be cultured ex vivo in the presence of proteins of the present invention in order to proliferate or to produce a desired effect on or activity in such cells. Treated cells can then be introduced in vivo for therapeutic purposes. Alternatively, it is contemplated that in other human disease states, preventing the expression of or inhibiting the activity of polypeptides of the invention will be useful in treating the disease states. It is contemplated that antisense therapy or gene therapy could be applied to negatively regulate the expression of polypeptides of the invention.




Other methods inhibiting expression of a protein include the introduction of antisense molecules to the nucleic acids of the present invention, their complements, or their translated RNA sequences, by methods known in the art. Further, the polypeptides of the present invention can be inhibited by using targeted deletion methods, or the insertion of a negative regulatory element such as a silencer, which is tissue specific.




The present invention still further provides cells genetically engineered in vivo to express the polynucleotides of the invention, wherein such polynucleotides are in operative association with a regulatory sequence heterologous to the host cell which drives expression of the polynucleotides in the cell. These methods can be used to increase or decrease the expression of the polynucleotides of the present invention.




Knowledge of DNA sequences provided by the invention allows for modification of cells to permit, increase, or decrease, expression of endogenous polypeptide. Cells can be modified (e.g., by homologous recombination) to provide increased polypeptide expression by replacing, in whole or in part, the naturally occurring promoter with all or part of a heterologous promoter so that the cells express the protein at higher levels. The heterologous promoter is inserted in such a manner that it is operatively linked to the desired protein encoding sequences. See, for example, PCT International Publication No. WO 94/12650, PCT International Publication No. WO 92/20808, and PCT International Publication No. WO 91/09955. It is also contemplated that, in addition to heterologous promoter DNA, amplifiable marker DNA (e.g., ada, dhfr, and the multifunctional CAD gene which encodes carbamyl phosphate synthase, aspartate transcarbamylase, and dihydroorotase) and/or intron DNA may be inserted along with the heterologous promoter DNA. If linked to the desired protein coding sequence, amplification of the marker DNA by standard selection methods results in co-amplification of the desired protein coding sequences in the cells.




In another embodiment of the present invention, cells and tissues may be engineered to express an endogenous gene comprising the polynucleotides of the invention under the control of inducible regulatory elements, in which case the regulatory sequences of the endogenous gene may be replaced by homologous recombination. As described herein, gene targeting can be used to replace a gene's existing regulatory region with a regulatory sequence isolated from a different gene or a novel regulatory sequence synthesized by genetic engineering methods. Such regulatory sequences may be comprised of promoters, enhancers, scaffold-attachment regions, negative regulatory elements, transcriptional initiation sites, regulatory protein binding sites or combinations of said sequences. Alternatively, sequences which affect the structure or stability of the RNA or protein produced may be replaced, removed, added, or otherwise modified by targeting. These sequences include polyadenylation signals, mRNA stability elements, splice sites, leader sequences for enhancing or modifying transport or secretion properties of the protein, or other sequences which alter or improve the function or stability of protein or RNA molecules.




The targeting event may be a simple insertion of the regulatory sequence, placing the gene under the control of the new regulatory sequence, e.g., inserting a new promoter or enhancer or both upstream of a gene. Alternatively, the targeting event may be a simple deletion of a regulatory element, such as the deletion of a tissue-specific negative regulatory element. Alternatively, the targeting event may replace an existing element; for example, a tissue-specific enhancer can be replaced by an enhancer that has broader or different cell-type specificity than the naturally occurring elements. Here, the naturally occurring sequences are deleted and new sequences are added. In all cases, the identification of the targeting event may be facilitated by the use of one or more selectable marker genes that are contiguous with the targeting DNA, allowing for the selection of cells in which the exogenous DNA has integrated into the cell genome. The identification of the targeting event may also be facilitated by the use of one or more marker gencs exhibiting the property of negative selection, such that the negatively selectable marker is linked to the exogenous DNA, but configured such that the negatively selectable marker flanks the targeting sequence, and such that a correct homologous recombination event with sequences in the host cell genome does not result in the stable integration of the negatively selectable marker. Markers useful for this purpose include the Herpes Simplex Virus thyrnidine kinase (TK) gene or the bacterial xanthine-guanine phosphoribosyl-transferase (gpt) gene.




The gene targeting or gene activation techniques which can be used in accordance with this aspect of the invention are more particularly described in U.S. Pat. No. 5,272,071 to Chappel; U.S. Pat. No. 5,578,461 to Sherwin et al.; International Application No. PCT/US92/09627 (WO93/09222) by Selden et al.; and International Application No. PCT/US90/06436 (WO91/06667) by Skoultchi et al., each of which is incorporated by reference herein in its entirety.




3.9 Transgenic Animals




In preferred methods to determine biological functions of the polypeptides of the invention in vivo, one or more genes provided by the invention are either over expressed or inactivated in the germ line of animals using homologous recombination [Capecchi, Science 244:1288-1292 (1989)]. Animals in which the gene is over expressed, under the regulatory control of exogenous or endogenous promoter elements, are known as transgenic animals. Animals in which an endogenous gene has been inactivated by homologous recombination are referred to as “knockout” animals. Knockout animals, preferably non-human mammals, can be prepared as described in U.S. Pat. No. 5,557,032, incorporated herein by reference. Transgenic animals are useful to determine the roles polypeptides of the invention play in biological processes, and preferably in disease states. Transgenic animals are useful as model systems to identify compounds that modulate lipid metabolism. Transgenic animals, preferably non-human mammals, are produced using methods as described in U.S. Pat. No. 5,489,743 and PCT Publication No. WO94/28122, incorporated herein by reference.




Transgenic animals can be prepared wherein all or part of a promoter of the polynucleotides of the invention is either activated or inactivated to alter the level of expression of the polypeptides of the invention. Inactivation can be carried out using homologous recombination methods described above. Activation can be achieved by supplementing or even replacing the homologous promoter to provide for increased protein expression. The homologous promoter can be supplemented by insertion of one or more heterologous enhancer elements known to confer promoter activation in a particular tissue.




The polynucleotides of the present invention also make possible the development, through, e.g., homologous recombination or knock out strategies, of animals that fail to express polypeptides of the invention or that express a variant polypeptide. Such animals are useful as models for studying the in vivo activities of polypeptide as well as for studying modulators of the polypeptides of the invention.




In preferred methods to determine biological functions of the polypeptides of the invention in vivo, one or more genes provided by the invention are either over expressed or inactivated in the germ line of animals using homologous recombination [Capecchi, Science 244:1288-1292 (1989)]. Animals in which the gene is over expressed, under the regulatory control of exogenous or endogenous promoter elements, are known as transgenic animals. Animals in which an endogenous gene has been inactivated by homologous recombination are referred to as “knockout” animals. Knockout animals, preferably non-human mammals, can be prepared as described in U.S. Pat. No. 5,557,032, incorporated herein by reference. Transgenic animals are useful to determine the roles polypeptides of the invention play in biological processes, and preferably in disease states. Transgenic animals are useful as model systems to identify compounds that modulate lipid metabolism. Transgenic animals, preferably non-human mammals, are produced using methods as described in U.S. Pat. No. 5,489,743 and PCT Publication No. WO94/28122, incorporated herein by reference.




Transgenic animals can be prepared wherein all or part of the polynucleotides of the invention promoter is either activated or inactivated to alter the level of expression of the polypeptides of the invention. Inactivation can be carried out using homologous recombination methods described above. Activation can be achieved by supplementing or even replacing the homologous promoter to provide for increased protein expression. The homologous promoter can be supplemented by insertion of one or more heterologous enhancer elements known to confer promoter activation in a particular tissue.




3.10 Uses and Biological Activity




The polynucleotides and proteins of the present invention are expected to exhibit one or more of the uses or biological activities (including those associated with assays cited herein) identified herein. Uses or activities described for proteins of the present invention may be provided by administration or use of such proteins or of polynuclcotides encoding such proteins (such as, for example, in gene therapies or vectors suitable for introduction of DNA). The mechanism underlying the particular condition or pathology will dictate whether the polypeptides of the invention, the polynucleotides of the invention or modulators (activators or inhibitors) thereof would be beneficial to the subject in need of treatment. Thus, “therapeutic compositions of the invention” include compositions comprising isolated polynucleotides (including recombinant DNA molecules, cloned genes and degenerate variants thereof) or polypeptides of the invention (including full length protein, mature protein and truncations or domains thereof), or compounds and other substances that modulate the overall activity of the target gene products, either at the level of target gene/protein expression or target protein activity. Such modulators include polypeptides, analogs, (variants), including fragments and fusion proteins, antibodies and other binding proteins; chemical compounds that directly or indirectly activate or inhibit the polypeptides of the invention (identified, e.g., via drug screening assays as described herein); antisense polynucleotides and polynucleotides suitable for triple helix formation; and in particular antibodies or other binding partners that specifically recogrize one or more epitopes of the polypeptides of the invention.




The polypeptides of the present invention may likewise be involved in cellular activation or in one of the other physiological pathways described herein.




3.10.1 Research Uses and Utilities




The polynucleotides provided by the present invention can be used by the research community for various purposes. The polynucleotides can be used to express recombinant protein for analysis, characterization or therapeutic use; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in disease states); as molecular weight markers on gels; as chromosome markers or tags (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic fingerprinting; as a probe to “subtract-out” known sequences in the process of discovering other novel polynucleotides; for selecting and making oligomers for attachment to a “gene chip” or other support, including for examination of expression patterns; to raise anti-protein antibodies using DNA immunization techniques; and as an antigen to raise anti-DNA antibodies or elicit another immune response. Where the polynucleotide encodes a protein which binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction), the polynucleotide can also be used in interaction trap assays (such as, for example, that described in Gyuris et al., Cell 75:791-803 (1993)) to identify polynucleotides encoding the other protein with which binding occurs or to identify inhibitors of the binding interaction.




The polypeptides provided by the present invention can similarly be used in assays to determine biological activity, including in a panel of multiple proteins for high-throughput screening; to raise antibodies or to elicit another immune response; as a reagent (including the labeled reagent) in assays designed to quantitatively determine levels of the protein (or its receptor) in biological fluids; as markers for tissues in which the corresponding polypeptide is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in a disease state); and, of course, to isolate correlative receptors or ligands. Proteins involved in these binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction.




Any or all of these research utilities are capable of being developed into reagent grade or kit format for corrjercialization as research products.




Methods for performing the uses listed above are well known to those skilled in the art. References disclosing such methods include without limitation “Molecular Cloning: A Laboratory Manual”, 2d ed., Cold Spring Harbor Laboratory Press, in Sambrook, J., E. F. Fritsch and T. Maniatis eds., 1989, and “Methods in Enzymology: Guide to Molecular Cloning Techniques”, Academic Press, Berger, S. L. and A. R Kimmel eds., 1987.




3.10.2 Nutritional Uses




Polynucleotides and polypeptides of the present invention can also be used as nutritional sources or supplements. Such uses include without limitation use as a protein or amino acid supplement, use as a carbon source, use as a nitrogen source and use as a source of carbohydrate. In such cases the polypeptidc or polynucleotide of the invention can be added to the feed of a particular organism or can be administered as a separate solid or liquid preparation, such as in the form of powder, pills, solutions, suspensions or capsules. In the case of microorganisms, the polypeptide or polynucleotide of the invention can be added to the medium in or on which the microorganism is cultured.




3.10.3 Cytokine and Cell Proliferation/Differentiation Activity




A polypeptide of the present invention may exhibit activity relating to 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. A polynucleotide of the invention can encode a polypeptide exhibiting such attributes. 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 therapeutic compositions 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, DA1G, T10, B9, B9/11, BaF3, MC9/G, M+(preB M+), 2E8, RB5, DA1, 123, T1165, HT2, CTLL2, TF-1, Mo7e, CMK, HUVEC, and Caco. Therapeutic compositions of the invention can be used in the following:




Assays for T-cell or thymocyte proliferation 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 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Bertagnolli et al., J. Immunol. 145:1706-1712, 1990; Bertagnolli et al., Cellular Imrnunology 133:327-341, 1991; Bertagnolli, et al., I. Immunol. 149:3778-3783, 1992; Bowman et al., I. Immunol. 152:1756-1761, 1994.




Assays for cytokine production and/or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described in: Polyclonal T cell stimulation, Kruisbeek, A. M. and Shevach, E. M. In Current Protocols in Immunology. J. E. e.a. Coligan eds. Vol 1 pp. 3.12.1-3.12.14, John Wiley and Sons, Toronto. 1994; and Measurement of mouse and human interleukin-y, Schreiber, R. D. In Current Protocols in Immunology. J. E. e.a. Coligan eds. Vol 1 pp. 6.8.1-6.8.8, John Wiley and Sons, Toronto. 1994.




Assays for proliferation and differentiation of hematopoietic and lymphopoietic cells include, without limitation, those described in: Measurement of Human and Murine Interleukin 2 and Interleukin 4, Bottomly, K., Davis, L. S. and Lipsky, P. E. In Current Protocols in Immunology. J. E. e.a. Coligan 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; Measurement of mouse and human interleukin 6—Nordan, R. In Current Protocols in Immunology. J. E. Coligan eds. Vol 1 pp. 6.6.1-6.6.5, John Wilcy and Sons, Toronto. 1991; Smith et al., Proc. Natl. Aced. Sci. U.S.A. 83:1857-1861, 1986; Measurement of human Interleukin 11—Bennett, F., Giannotti, J., Clark, S. C. and Turner, K. J. In Current Protocols in Immunology. J. E. Coligan eds. Vol 1 pp. 6.15.1 John Wiley and Sons, Toronto. 1991; Measurement of mouse and human Interlcukin 9—Ciarletta, A., Giannotti, J., Clark, S. C. and Turner, K. J. In Current Protocols in Imnnunology. J. E. Coligan eds. Vol I pp. 6.13.1, John Wiley and Sons, Toronto. 1991.




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: A 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 3, In Vitro assays for Mouse Lymphocyte Function; Chapter 6, Cytokines and their cellular receptors; Chapter 7, Immunologic studies in Humans); 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.




3.10.4 Stem Cell Growth Factor Activity




A polypeptide of the present invention may exhibit stem cell growth factor activity and be involved in the proliferation, differentiation and survival of pluripotent and totipotent stem cells including primordial germ cells, embryonic stem cells, hematopoietic stem cells and/or germ line stem cells. Administration of the polypeptide of the invention to stem cells in vivo or ex vivo is expected to maintain and expand cell populations in a totipotential or pluripotential state which would be useful for re-engineering damaged or diseased tissues, transplantation, manufacture of bio-pharmaceuticals and the development of bio-sensors. The ability to produce large quantities of human cells has important working applications for the production of human proteins which currently must be obtained from non-human sources or donors, implantation of cells to treat diseases such as Parkinson's, Alzheimer's and other neurodegenerative diseases; tissues for grafting such as bone marrow, skin, cartilage, tendons, bone, muscle (including cardiac muscle), blood vessels, cornea, neural cells, gastrointestinal cells and others; and organs for transplantation such as kidney, liver, pancreas (including islet cells), heart and lung.




It is contemplated that multiple different exogenous growth factors and/or cytokines may be administered in combination with the polypeptide of the invention to achieve the desired effect, including any of the growth factors listed herein, other stem cell maintenance factors, and specifically including stem cell factor (SCF), leukemia inhibitory factor (LIF), Flt-3 ligand (Flt-3L), any of the interleukins, recombinant soluble IL-6 receptor fused to IL-6, macrophagc inflammatory protein 1-alpha (MIP-1-alpha), G-CSF, GM-CSF, thrombopoietin (TPO), platelet factor 4 (PF-4), platelet-derived growth factor (PDGF), neural growth factors and basic fibroblast growth factor (bFGF).




Since totipotent stem cells can give rise to virtually any mature cell type, expansion of these cells in culture will facilitate the production of large quantities of mature cells. Techniques for culturing stem cells are known in the art and administration of polypeptides of the invention, optionally with other growth factors and/or cytokines, is expected to enhance the survival and proliferation of the stem cell populations. This can be accomplished by direct administration of the polypeptide of the invention to the culture medium. Alternatively, stroma cells transfected with a polynucleotide that encodes for the polypeptide of the invention can be used as a feeder layer for the stem cell populations in culture or in vivo. Stromal support cells for feeder layers may include embryonic bone marrow fibroblasts, bone marrow stromal cells, fetal liver cells, or cultured embryonic fibroblasts (see U.S. Pat. No. 5,690,926).




Stem cells themselves can be transfected with a polynucleotide of the invention to induce autocrine expression of the polypeptide of the invention. This will allow for generation of undifferentiated totipotential/pluripotential stem cell lines that are useful as is or that can then be differentiated into the desired mature cell types. These stable cell lines can also serve as a source of undifferentiated totipotential/pluripotential mRNA to create cDNA libraries and templates for polymerase chain reaction experiments. These studies would allow for the isolation and identification of differentially expressed genes in stem cell populations that regulate stem cell proliferation and/or maintenance.




Expansion and maintenance of totipotent stem cell populations will be useful in the treatment of many pathological conditions. For example, polypeptides of the present invention may be used to manipulate stem cells in culture to give rise to neuroepithelial cells that can be used to augment or replace cells damaged by illness, autoimmune disease, accidental damage or genetic disorders. The polypeptide of the invention may be useful for inducing the proliferation of neural cells and for the 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. In addition, the expanded stem cell populations can also be genetically altered for gene therapy purposes and to decrease host rejection of replacement tissues after grafting or implantation.




Expression of the polypeptide of the invention and its effect on stem cells can also be manipulated to achieve controlled differentiation of the stem cells into more differentiated cell types. A broadly applicable method of obtaining pure populations of a specific differentiated cell type from undifferentiated stem cell populations involves the use of a cell-type specific promoter driving a selectable marker. The selectable marker allows only cells of the desired type to survive. For example, stem cells can be induced to differentiate into cardiomyocytes (Wobus et al., Differentiation, 48: 173-182, (1991); Klug et al., J. Clin. Invest., 98(1): 216-224, (1998)) or skeletal muscle cells (Browder, L. W. In:


Principles of Tissue Engineering eds


. Lanza et al., Acadernic Press (1997)). Alternatively, directed differentiation of stem cells can be accomplished by culturing the stem cells in the presence of a differentiation factor such as retinoic acid and an antagonist of the polypeptide of the invention which would inhibit the effects of endogenous stem cell factor activity and allow differentiation to proceed.




In vitro cultures of stem cells can be used to determine if the polypeptide of the invention exhibits stem cell growth factor activity. Stem cells are isolated from any one of various cell sources (including hematopoietic stem cells and embryonic stem cells) and cultured on a feeder layer, as described by Thompson et al. Proc. Natl. Acad. Sci, U.S.A., 92: 7844-7848 (1995), in the presence of the polypeptide of the invention alone or in combination with other growth factors or cytokines. The ability of the polypeptide of the invention to induce stem cells proliferation is determined by colony formation on semi-solid support e.g. as described by Bernstein et al., Blood, 77: 2316-2321 (1991).




3.10.5 Hematopoiesis Regulating Activity




A polypeptide of the present invention may be involved in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell disorders. 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 erythwid 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.




Therapeutic compositions of the invention can be used in the following:




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




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.




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 formring assays, Freshney, M. G. In Culture of Hematopoietic Cells. R. L. 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; Primitive hematopoietic colony forming cells with high proliferative potential, McNiece, I. K. and Briddell, R. A. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. Vol pp. 23-39, Wiley-Liss, Inc., New York, N.Y. 1994; Neben et al., Experimental Hematology 22:353-359, 1994; Cobblestone area forming cell assay, Ploemacher, R. E. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. Vol pp. 1-21, Wiley-Liss, Inc., New York, N.Y. 1994; Long term bone marrow cultures in the presence of stromal cells, Spooncer, E., Dexter, M. and Allen, T. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. Vol pp. 163-179, Wiley-Liss, Inc., New York, N.Y. 1994; Long term culture initiating cell assay, Sutherland, H. J. In Culture of Hematopoietic Cells. R I. Freshney, et al. eds. Vol pp. 139-162, Wiley-Liss, Inc., New York, N.Y. 1994.




3.10.6 Tissue Growth Activity




A polypeptide of the present invention also may be involved in bone, cartilage, tendon, ligament and/or nerve tissue growth or regeneration, as well as in wound healing and tissue repair and replacement, and in healing of bums, incisions and ulcers.




A polypeptide 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. Compositions of a polypeptide, antibody, binding partner, or other modulator 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.




A polypeptide of this invention may also be involved in attracting bone-forming cells, stimulating growth of bone-formning cells, or inducing differentiation of progenitors of bone-forming cells. Treatment of osteoporosis, osteoarthritis, bone degenerative disorders, or periodontal disease, 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 may also be possible using the composition of the invention.




Another category of tissue regeneration activity that may involve the polypeptide of the present invention is tendon/ligament formation. Induction of 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 environment to attract tendon- or ligament-forrning cells, stimulate growth of tendon- or ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/liganent 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 tendinitis, carpal tunnel syndrome and other tendon or ligament defects. The compositions may also include an appropriate matrix and/or sequestering agent as a carrier as is well known in the art.




The compositions 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 composition 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 composition of the invention.




Compositions 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.




Compositions of the present invention may also be involved in the 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 may allow normal tissue to regenerate. A polypeptide of the present invention may also exhibit angiogenic activity.




A composition 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.




A composition 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.




Therapeutic compositions of the invention can be used in the following:




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).




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




3.10.7 Immune Stimulating or Suppressing Activity




A polypeptide 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 polynucleotide of the invention can encode a polypeptide exhibiting such activities. A protein may be 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., HI) as well as bacterial or fungal infections, or may result from autoimmune disorders. Morc specifically, infectious diseases causes by viral, bacterial, ftngal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpes viruses, mycobacteria, Leishmania spp., malaria spp. and various fungal infections such as candidiasis. Of course, in this regard, proteins 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.




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 mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease. Such a protein (or antagonists thereof, including antibodies) of the present invention may also to be useful in the treatment of allergic reactions and conditions (e.g., anaphylaxis, serum sickness, drug reactions, food allergies, insect venom allergies, mastocytosis, allergic rhinitis, hypersensitivity pneumonitis, urticaria, angioedema, eczema, atopic dermatitis, allergic contact dermatitis, erythema multiforme, Stevens-Johnson syndrome, allergic conjunctivitis, atopic keratoconjunctivitis, venereal keratoconjunctivitis, giant papillary conjunctivitis and contact allergies), such as asthma (particularly allergic asthma) or other respiratory problems. Other conditions, in which inrnune suppression is desired (including, for example, organ transplantation), may also be treatable using a protein (or antagonists thereof) of the present invention. The therapeutic effects of the polypeptides or antagonists thereof on allergic reactions can be evaluated by in vivo animals models such as the cumulative contact enhancement test (Lastbom et al., Toxicology 125: 59-66, 1998), skin prick test (Hoffmann et al., Allergy 54: 446-54, 1999), guinea pig skin sensitization test (Vohr et al., Arch. Toxocol. 73: 501-9), and murine local lymph node assay (Kimber et al., J. Toxicol. Environ. Health 53: 563-79).




Using the proteins of the invention it may also be possible to modulate 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 anergy 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 reexposure to specific antigen in the absence of the tolerizing agent.




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, will be 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 therapeutic composition of the invention may prevent cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant. Moreover, a 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 a combination of B lymphocyte antigens.




The efficacy of particular therapeutic compositions 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 therapeutic compositions of the invention on the development of that disease.




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 autoantibodies 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 stimulation of T cells can be used to inhibit T cell activation and prevent production of autoantibodies 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 erythmatosis 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).




Upregulation of an antigen function (e.g., 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 may be useful in cases of viral infection, including systemic viral diseases such as influenza, the common cold, and encephalitis.




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.




A polypeptide of the present invention may provide the necessary stimulation 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 mounts 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 alpha chain protein and β


2


microglobulin protein or an MHC class II alpha chain protein and an MHC class II beta 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.




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, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); 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., I. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Bowman et al., J. Virology 61:1992-1998; Bertagnolli et al., Cellular Immunology 133:327-341, 1991; Brown et al., J. Immunol. 153:3079-3092, 1994.




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 Assays for B cell function: In vitro antibody production, Mond, J. J. and Brunswick, M. In Current Protocols in Immunology. J. E. e.a. Coligan eds. Vol 1 pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto. 1994.




Mixed lymphocyte reaction (MLR) assays (which will identify, among others, proteins that generate predominantly Th1 and CTL responses) 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 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); 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.




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., Journal of Experimental Medicine 173:549-559, 1991; Macatonia et al., Journal of Immunology 154:5071-5079, 1995; Porgador et al., Journal of Experimental Medicine 182:255-260, 1995; Nair et al., Journal of Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., Journal of Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., Journal of Clinical Investigation 94:797-807, 1994; and Inaba et al., Journal of Experimental Medicine 172:631-640, 1990.




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 Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, Journal of Immunology 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., International Journal of Oncology 1:639-648, 1992.




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., Cellular Immunology 155:111-122, 1994; Galy et al., Blood 85:2770-2778, 1995; Toki et al., Proc. Nat. Acad Sci. USA 88:7548-7551, 1991.




3.10.8 Activin/Inhibin Activity




A polypeptide of the present invention may also exhibit activin- or inhibin-related activities. A polynucleotide of the invention may encode a polypeptide exhibiting such characteristics. 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 polypeptide of the present invention, alone or in heterodimers with a member of the inhibin family, may be 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 polypeptide of the invention, as a homodimer or as a heterodimer with other protein subunits of the inhibin group, may be 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 polypeptide 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, but not limited to, cows, sheep and pigs.




The activity of a polypeptide of the invention may, among other means, be measured by the following methods.




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.




3.10.9 Cremotactic/Chemokinetic Activity




A polypeptide of the present invention may be involved in chemotactic or chemokinetic activity for mammalian cells, including, for example, monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells. A polynucleotide of the invention can encode a polypeptide exhibiting such attributes. Chemotactic and chemokinetic receptor activation can be used to mobilize or attract a desired cell population to a desired site of action. Chemotactic or chemokinetic compositions (e.g. proteins, antibodies, binding partners, or modulators of the invention) 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.




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.




Therapeutic compositions of the invention can be used in the following:




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, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Marguiles, E. M. Shevach, W. Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 6.12, Measurernent 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; Gruber et al. J. of Immunol. 152:5860-5867, 1994; Johnston et al. J. of Immunol. 153:1762-1768, 1994.




3.10.10 Hemostatic and Thrombolytic Activity




A polypeptide of the invention may also be involved in hemostatis or thrombolysis or thrombosis. A polynucleotide of the invention can encode a polypeptide exhibiting such attributes. Compositions may 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 composition 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).




Therapeutic compositions of the invention can be used in the following:




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.




3.10.11 Cancer Diagnosis and Therapy




Polypeptides of the invention may be involved in cancer cell generation, proliferation or metastasis. Detection of the presence or amount of polynucleotides or polypeptides of the invention may be useful for the diagnosis and/or prognosis of one or more types of cancer. For example, the presence or increased expression of a polynucleotide/polypeptide of the invention may indicate a hereditary risk of cancer, a precancerous condition, or an ongoing malignancy. Conversely, a defect in the gene or absence of the polypeptide may be associated with a cancer condition. Identification of single nucleotide polymorphisms associated with cancer or a predisposition to cancer may also be useful for diagnosis or prognosis.




Cancer treatments promote tumor regression by inhibiting tumor cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumor growth) and/or prohibiting metastasis by reducing tumor cell motility or invasiveness. Therapeutic compositions of the invention may be effective in adult and pediatric oncology including in solid phase tumors/malignancies, locally advanced tumors, human soft tissue sarcomas, metastatic cancer, including lymphatic metastases, blood cell malignancies including multiple myeloma, acute and chronic leukemnias, and lymphomas, head and neck cancers including mouth cancer, larynx cancer and thyroid cancer, lung cancers including small cell carcinoma and non-small cell cancers, breast cancers including small cell carcinoma and ductal carcinoma, gastrointestinal cancers including esophageal cancer, stomach cancer, colon cancer, colorectal cancer and polyps associated with colorectal neoplasia, pancreatic cancers, liver cancer, urologic cancers including bladder cancer and prostate cancer, malignancies of the female genital tract including ovarian carcinoma, uterine (including endometrial) cancers, and solid tumor in the ovarian follicle, kidney cancers including renal cell carcinoma, brain cancers including intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, metastatic tumor cell invasion in the central nervous system, bone cancers including osteomas, skin cancers including malignant melanoma, tumor progression of human skin keratinocytes, squamous cell carcinoma, basal cell carcinoma, hemangiopericytoma and Karposi's sarcoma.




Polypeptides, polynucleotides, or modulators of polypeptides of the invention (including inhibitors and stimulators of the biological activity of the polypeptide of the invention) may be administered to treat cancer. Therapeutic compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumor size, slowing rate of tumor growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.




The composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail. An anti-cancer cocktail is a mixture of the polypeptide or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. The use of anti-cancer cocktails as a cancer treatment is routine. Anti-cancer drugs that are well known in the art and can be used as a treatment in combination with the polypeptide or modulator of the invention include: Actinomycin D, Aminoglutethimide, Asparaginase, Bleomycin, Busulfan, Carboplatin, Carmustine, Chlorambucil, Cisplatin (cis-DDP), Cyclophosphamide, Cytarabine HCl (Cytosine arabinoside), Dacarbazine, Dactinomycin, Daunorubicin HCl, Doxorubicin HCl, Estramustine phosphate sodium, Etoposide (V16-213), Floxuridine, 5-Fluorouracil (5-Fu), Flutamide, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alpha-2a, Interferon Alpha-2b, Leuprolide acetate (LHRH-releasing factor analog), Lomustine, Mechiorethamnine HCl (nitrogen mustard), Melphalan, Mercaptopurine, Mesna, Methotrexate (MTX), Mitomycin, Mitoxantrone HCl, Octreotide, Plicamycin, Procarbazine HCl, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Vincristine sulfate, Ainsacrinc, Azacitidine, Hexamethylnelamine, Interleukin-2, Mitoguazone, Pentostatin, Semustine, Teniposide, and Vindesine sulfate.




In addition, therapeutic compositions of the invention may be used for prophylactic treatment of cancer. There are hereditary conditions and/or environmental situations (e.g. exposure to carcinogens) known in the art that predispose an individual to developing cancers. Under these circumstances, it may be beneficial to treat these individuals with therapeutically effective doses of the polypeptide of the invention to reduce the risk of developing cancers.




In vitro models can be used to determine the effective doses of the polypeptide of the invention as a potential cancer treatment. These in vitro models include proliferation assays of cultured tumor cells, growth of cultured tumor cells in soft agar (see Freshney, (1987) Culture of Animal Cells: A Manual of Basic Technique, Wily-Liss, New York, N.Y. Ch 18 and Ch 21), tumor systems in nude mice as described in Giovanella et al., J. Natl. Can. Inst., 52: 921-30 (1974), mobility and invasive potential of tumor cells in Boyden Chamber assays as described in Pilkington et al., Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et al., Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li et al., Clin. Exp. Metastasis, 17:423-9 (1999), respectively. Suitable tumor cells lines are available, e.g. from American Type Tissue Culture Collection catalogs.




3.10.12 Receptor/Ligand Activity




A polypeptide of the present invention may also demonstrate activity as receptor, receptor ligand or inhibitor or agonist of receptor/ligand interactions. A polynucleotide of the invention can encode a polypeptide exhibiting such characteristics. 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.




The activity of a polypeptide of the invention may, among other means, be measured by the following methods:




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.




By way of example, the polypeptides of the invention may be used as a receptor for a ligand(s) thereby transmitting the biological activity of that ligand(s). Ligands may be identified through binding assays, affinity chromatography, dihybrid screening assays, BIAcore assays, gel overlay assays, or other methods known in the art.




Studies characterizing drugs or proteins as agonist or antagonist or partial agonists or a partial antagonist require the use of other proteins as competing ligands. The polypeptides of the present invention or ligand(s) thereof may be labeled by being coupled to radioisotopes, colorimetric molecules or a toxin molecules by conventional methods. (“Guide to Protein Purification” Murray P. Deutscher (ed) Methods in Enzymology Vol. 182 (1990) Academic Press, Inc. San Diego). Examples of radioisotopes include, but are not limited to, tritium and carbon-14. Examples of calorimetric molecules include, but are not limited to, fluorescent molecules such as fluorescamine, or rhodamine or other colorimetric molecules. Examples of toxins include, but are not limited, to ricin.




3.10.13 Drug Screening




This invention is particularly useful for screening chemical compounds by using the novel polypeptides or binding fragments thereof in any of a variety of drug screening techniques. The polypeptides or fragments employed in such a test may either be free in solution, affixed to a solid support, borne on a cell surface or located intracellularly. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant nucleic acids expressing the polypeptide or a fragment thereof. Drugs are screened against such transformed cells in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays. One may measure, for example, the formation of complexes between polypeptides of the invention or fragments and the agent being tested or examine the diminution in complex formation between the novel polypeptides and an appropriate cell line, which are well known in the art.




Sources for test compounds that may be screened for ability to bind to or modulate (i.e., increase or decrease) the activity of polypeptides of the invention include (1) inorganic and organic chemical libraries, (2) natural product libraries, and (3) combinatorial libraries comprised of either random or mimetic peptides, oligonucleotides or organic molecules.




Chemical libraries may be readily synthesized or purchased from a number of commercial sources, and may include structural analogs of known compounds or compounds that are identified as “hits” or “leads” via natural product screening.




The sources of natural product libraries are microorganisms (including bacteria and fungi), animals, plants or other vegetation, or marine organisms, and libraries of mixtures for screening may be created by: (1) fermentation and extraction of broths from soil, plant or marine microorganisms or (2) extraction of the organisms themselves. Natural product libraries include polyketides, non-ribosomal peptides, and (non-naturally occurring) variants thereof. For a review, see


Science


282:63-68 (1998).




Combinatorial libraries are composed of large numbers of peptides, oligonucleotides or organic compounds and can be readily prepared by traditional automated synthesis methods, PCR, cloning or proprietary synthetic methods. Of particular interest are peptide and oligonucleotide combinatorial libraries. Still other libraries of interest include peptide, protein, peptidomimetic, multiparallel synthetic collection, recombinatorial, and polypeptide libraries. For a review of combinatorial chemistry and libraries created therefrom, see Myers,


Curr. Opin. Biotechnol


. 8:701-707 (1997). For reviews and examples of peptidomimetic libraries, see Al-Obeidi et al.,


Mol. Biotechnol


, 9(3):205-23 (1998); Hrubyet al.,


Curr Opin Chem Biol,


1(1):114-19(1997); Domer et al.,


Bioorg Med Chem


, 4(5):709-15 (1996) (alkylated dipeptides).




Identification of modulators through use of the various libraries described herein permits modification of the candidate “hit” (or “lead”) to optimize the capacity of the “hit” to bind a polypeptide of the invention. The molecules identified in the binding assay are then tested for antagonist or agonist activity in in vivo tissue culture or animal models that are well known in the art. In brief, the molecules are titrated into a plurality of cell cultures or animals and then tested for either cell/animal death or prolonged survival of the animal/cells.




The binding molecules thus identified may be complexed with toxins, e.g., ricin or cholera, or with other compounds that are toxic to cells such as radioisotopes. The toxin-binding molecule complex is then targeted to a tumor or other cell by the specificity of the binding molecule for a polypeptide of the invention. Alternatively, the binding molecules may be complexed with imaging agents for targeting and imaging purposes.




3.10.14 Assay for Receptor Activity




The invention also provides methods to detect specific binding of a polypeptide e.g. a ligand or a receptor. The art provides numerous assays particularly useful for identifying previously unknown binding partners for receptor polypeptides of the invention. For example, expression cloning using mammalian or bacterial cells, or dihybrid screening assays can be used to identify polynucleotides encoding binding partners. As another example, affinity chromatography with the appropriate immobilized polypeptide of the invention can be used to isolate polypeptides that recognize and bind polypeptides of the invention. There are a number of different libraries used for the identification of compounds, and in particular small molecules, that modulate (i.e., increase or decrease) biological activity of a polypeptide of the invention. Ligands for receptor polypeptides of the invention can also be identified by adding exogenous ligands, or cocktails of ligands to two cells populations that are genetically identical except for the expression of the receptor of the invention: one cell population expresses the receptor of the invention whereas the other does not. The response of the two cell populations to the addition of ligands(s) are then compared. Alternatively, an expression library can be co-expressed with the polypeptide of the invention in cells and assayed for an autocrine response to identify potential ligand(s). As still another example, BIAcore assays, gel overlay assays, or other methods known in the art can be used to identify binding partner polypeptides, including, (1) organic and inorganic chemical libraries, (2) natural product libraries, and (3) combinatorial libraries comprised of random peptides, oligonuclcotides or organic molecules.




The role of downstream intracellular signaling molecules in the signaling cascade of the polypeptide of the invention can be determined. For example, a chimeric protein in which the cytoplasmic domain of the polypeptide of the invention is fused to the extracellular portion of a protein, whose ligand has been identified, is produced in a host cell. The cell is then incubated with the ligand specific for the extracellular portion of the chimeric protein, thereby activating the chimeric receptor. Known downstream proteins involved in intracellular signaling can then be assayed for expected modifications i.e. phosphorylation. Other methods known to those in the art can also be used to identify signaling molecules involved in receptor activity.




3.10.15 Anti-inflammatory Activity




Compositions 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. Compositions with such activities can be used to treat inflammatory conditions including chronic or acute conditions), including without limitation intimation 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. Compositions of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material. Compositions of this invention may be utilized to prevent or treat conditions such as, but not limited to, sepsis, acute pancreatitis, endotoxin shock, cytokine induced shock, rheumatoid arthritis, chronic inflammatory arthritis, pancreatic cell damage from diabetes mellitus type 1, graft versus host disease, inflammatory bowel disease, inflamation associated with pulmonary disease, other autoimmune disease or inflammatory disease, an antiproliferative agent such as for acute or chronic mylegenous leukemia or in the prevention of premature labor secondary to intrauterine infections.




3.10.16 Leukemias




Leukemias and related disorders may be treated or prevented by administration of a therapeutic that promotes or inhibits function of the polynucleotides and/or polypeptides of the invention. Such leukemias and related disorders include but are not limited to acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, myeloblastic, promyclocytic, myelomonocytic, monocytic, erythroleukemia, chronic leukemia, chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia (for a review of such disorders, see Fishman et al., 1985, Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia).




3.10.17 Nervous System Disorders




Nervous system disorders, involving cell types which can be tested for efficacy of intervention with compounds that modulate the activity of the polynucleotides and/or polypeptides of the invention, and which can be treated upon thus observing an indication of therapeutic utility, include but are not limited to nervous system injuries, and diseases or disorders which result in either a disconnection of axons, a diminution or degeneration of neurons, or demyelination. Nervous system lesions which may be treated in a patient (including human and non-human mammalian patients) according to the invention include but are not limited to the following lesions of either the central (including spinal cord, brain) or peripheral nervous systems:




(i) traumatic lesions, including lesions caused by physical injury or associated with surgery, for example, lesions which sever a portion of the nervous system, or compression injuries;




(ii) ischemnic lesions, in which a lack of oxygen in a portion of the nervous system results in neuronal injury or death, including cerebral infarction or ischemia, or spinal cord infarction or ischemia;




(iii) infectious lesions, in which a portion of the nervous system is destroyed or injured as a result of infection, for example, by an abscess or associated with infection by human immunodeficiency virus, herpes zoster, or herpes simplex virus or with Lymc disease, tuberculosis, syphilis;




(iv) degenerative lesions, in which a portion of the nervous system is destroyed or injured as a result of a degenerative process including but not limited to degeneration associated with Parkinson's disease, Alzheimer's disease, Huntington's chorea, or amyotrophic lateral sclerosis;




(v) lesions associated with nutritional diseases or disorders, in which a portion of the nervous system is destroyed or injured by a nutritional disorder or disorder of metabolism including but not limited to, vitamin B12 deficiency, folic acid deficiency, Wemicke disease, tobacco-alcohol amblyopia, Marchiafava-Bignami disease (primary degeneration of the corpus callosum), and alcoholic cerebellar degeneration;




(vi) neurological lesions associated with systemic diseases including but not limited to diabetes (diabetic neuropathy, Bell's palsy), systemic lupus erythematosus, carcinoma, or sarcoidosis;




(vii) lesions caused by toxic substances including alcohol, lead, or particular neurotoxins; and




(viii) demyelinated lesions in which a portion of the nervous system is destroyed or injured by a demyelinating disease including but not limited to multiple sclerosis, human immunodeficiency virus-associated myelopathy, transverse myelopathy or various etiologies, progressive multifocal leukoencephalopathy, and central pontine myelinolysis.




Therapeutics which are useful according to the invention for treatment of a nervous system disorder may be selected by testing for biological activity in promoting the survival or differentiation of neurons. For example, and not by way of limitation, therapeutics which elicit any of the following effects may be useful according to the invention:




(i) increased survival time of neurons in culture;




(ii) increased sprouting of neurons in culture or in vivo;




(iii) increased production of a neuron-associated molecule in culture or in vivo, e.g., choline acetyltransferase or acetylcholinesterase with respect to motor neurons; or




(iv) decreased symptoms of neuron dysfimction in vivo.




Such effects may be measured by any method known in the art. In preferred, non-limiting embodiments, increased survival of neurons may be measured by the method set forth in Arakawa et al. (1990, J. Neurosci. 10:3507-3515); increased sprouting of neurons may be detected by methods set forth in Pestronk et al. (1980, Exp. Neurol. 70:65-82) or Brown et al. (1981, Ann. Rev. Neurosci. 4:1742); increased production of neuron-associated molecules may be measured by bioassay, enzymatic assay, antibody binding, Northern blot assay, etc., depending on the molecule to be measured; and motor neuron dysfunction may be measured by assessing the physical manifestation of motor neuron disorder, e.g., weakness, motor neuron conduction velocity, or functional disability.




In specific embodiments, motor neuron disorders that may be treated according to the invention include but are not limited to disorders such as infarction, infection, exposure to toxin, trauma, surgical damage, degenerative disease or malignancy that may affect motor neurons as well as other components of the nervous system, as well as disorders that selectively affect neurons such as amyotrophic lateral sclerosis, and including but not limited to progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, infantile and juvenile muscular atrophy, progressive bulbar paralysis of childhood (Fazio-Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary Motorsensory Neuropathy (Charcot-Marie-Tooth Disease).




3.10.18 Other Activities




A polypeptide 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, fuingi 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 pigrnentation, 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, co-factors 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.




3.10.19 Identification of Polymorphisms




The demonstration of polymorphisms makes possible the identification of such polymorphisms in human subjects and the pharmacogenetic use of this information for diagnosis and treatment. Such polymorphisms may be associated with, e.g., differential predisposition or susceptibility to various disease states (such as disorders involving inflammation or immune response) or a differential response to drug admninistration, and this genetic information can be used to tailor preventive or therapeutic treatment appropriately. For example, the existence of a polymorphism associated with a predisposition to inflammation or autoimmune disease makes possible the diagnosis of this condition in humans by identifying the presence of the polymorphism.




Polymorphisms can be identified in a variety of ways known in the art which all generally involve obtaining a sample from a patient, analyzing DNA from the sample, optionally involving isolation or amplification of the DNA, and identifying the presence of the polymorphism in the DNA. For example, PCR may be used to amplify an appropriate fragment of genomic DNA which may then be sequenced. Alternatively, the DNA may be subjected to allele-specific oligonucleotide hybridization (in which appropriate oligonucleotides are hybridized to the DNA under conditions permitting detection of a single base mismatch) or to a single nucleotide extension assay (in which an oligonucleotide that hybridizes immediately adjacent to the position of the polymorphism is extended with one or more labeled nucleotides). In addition, traditional restriction fragment length polymorphism analysis (using restriction enzymes that provide differential digestion of the genomic DNA depending on the presence or absence of the polymorphism) may be performed. Arrays with nucleotide sequences of the present invention can be used to detect polymorphisms. The array can comprise modified nucleotide sequences of the present invention in order to detect the nucleotide sequences of the present invention. In the alternative, any one of the nucleotide sequences of the present invention can be placed on the array to detect changes from those sequences.




Alternatively a polymorphism resulting in a change in the amino acid sequence could also be detected by detecting a corresponding change in amino acid sequence of the protein, e.g., by an antibody specific to the variant sequence. 3.10.20 Arthritis and Inflammation




The immunosuppressive effects of the compositions of the invention against rheumatoid arthritis is determined in an experimental animal model system. The experimental model system is adjuvant induced arthritis in rats, and the protocol is described by J. Holoshitz, et at., 1983, Science, 219:56, or by B. Waksman et al., 1963, Int. Arch. Allergy Appl. Immunol., 23:129. Induction of the disease can be caused by a single injection, generally intradermally, of a suspension of killed Mycobacterium tuberculosis in complete Freund's adjuvant (CFA). The route of injection can vary, but rats may be injected at the base of the tail with an adjuvant mixture. The polypeptide is administered in phosphate buffered solution (PBS) at a dose of about 1-5 mg/kg. The control consists of administering PBS only.




The procedure for testing the effects of the test compound would consist of intradermally injecting killed Mycobacterium tuberculosis in CFA followed by immediately administering the test compound and subsequent treatment every other day until day 24. At 14, 15, 18, 20, 22, and 24 days after injection of Mycobacterium CFA, an overall arthritis score may be obtained as described by J. Holoskitz above. An analysis of the data would reveal that the test compound would have a dramatic affect on the swelling of the joints as measured by a decrease of the arthritis score.




3.11 Therapeutic Methods




The compositions (including polypeptide fragments, analogs, variants and antibodies or other binding partners or modulators including antisense polynucleotides) of the invention have numerous applications in a variety of therapeutic methods. Examples of therapeutic applications include, but are not limited to, those exemplified herein.




3.11.1 Example




One embodiment of the invention is the administration of an effective amount of the polypeptides or other composition of the invention to individuals affected by a disease or disorder that can be modulated by regulating the peptides of the invention. While the mode of administration is not particularly important, parenteral administration is preferred. An exemplary mode of administration is to deliver an intravenous bolus. The dosage of the polypeptides or other composition of the invention will normally be determined by the prescribing physician. It is to be expected that the dosage will vary according to the age, weight, condition and response of the individual patient. Typically, the amount of polypeptide administered per dose will be in the range of about 0.01 μg/kg to 100 mg/kg of body weight, with the preferred dose being about 0.1 μg/kg to 10 mg/kg of patient body weight. For parenteral administration, polypeptides of the invention will be formulated in an injectable form combined with a pharmaceutically acceptable parenteral vehicle. Such vehicles are well known in the art and examples include water, saline, Ringer's solution, dextrose solution, and solutions consisting of small amounts of the human serum albumin. The vehicle may contain minor amounts of additives that maintain the isotonicity and stability of the polypeptide or other active ingredient. The preparation of such solutions is within the slill of the art.




3.12 Pharmaceutical Formulations and Routes of Administration




A protein or other composition of the present invention (from whatever source derived, including without limitation from recombinant and non-recombinant sources and including antibodies and other binding partners of the polypeptides of the invention) may be administered to a patient in need, by itself, or in pharmaceutical compositions where it is mixed with suitable carriers or excipient(s) at doses to treat or ameliorate a variety of disorders. Such a composition may optionally contain (in addition to protein or other active ingredient and a carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration. The pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IFN, TNF0, TNF1, TNF2, G-CSF, Meg-CSF, thrombopoietin, stem cell factor, and erythropoietin. In further compositions, proteins of the invention may be combined with other agents beneficial to the treatment of the disease or disorder in question. These agents include various growth factors such as epidermal growth factor (EGF), platelet-derived growth factor (PDGF), transforming growth factors (TGF-α and TGF-β), insulin-like growth factor (IGF), as well as cytokines described herein.




The pharmaceutical composition may further contain other agents which either enhance the activity of the protein or other active ingredient or complement its activity or use in treatment. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect with protein or other active ingredient of the invention, or to minimize side effects. Conversely, protein or other active ingredient of the present invention may be included in formulations of the particular clotting factor, cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to minimize side effects of the clotting factor, cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent (such as IL-1Ra, IL-1 Hy1, IL-1 Hy2, anti-TNF, corticosteroids, immunosuppressive agents). A protein of the present invention may be active in multimers (e.g., heterodimers or homodimers) or complexes with itself or other proteins. As a result, pharmaceutical compositions of the invention may comprise a protein of the invention in such multimeric or complexed form.




As an alternative to being included in a pharmaceutical composition of the invention including a first protein, a second protein or a therapeutic agent may be concurrently administered with the first protein (e.g., at the same time, or at differing times provided that therapeutic concentrations of the combination of agents is achieved at the treatment site). Techniques for formulation and administration of the compounds of the instant application may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. A therapeutically effective dose further refers to that amount of the compound sufficient to result in amelioration of symptoms, e.g., treatment, healing, prevention or amelioration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions. When applied to an individual active ingredient, administered alone, a therapeutically effective dose refers to that ingredient alone. When applied to a combination, a therapeutically effective dose refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.




In practicing the method of treatment or use of the present invention, a therapeutically effective amount of protein or other active ingredient of the present invention is administered to a mammal having a condition to be treated. Protein or other active ingredient of the present invention may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors. When co-administered with one or more cytokines, lymphokines or other hematopoiefic factors, protein or other active ingredient of the present invention may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering protein or other active ingredient of the present invention in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors.




3.12.1 Routes of Administration




Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. Administration of protein or other active ingredient of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, topical application or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Intravenous administration to the patient is preferred.




Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into a arthritic joints or in fibrotic tissue, often in a depot or sustained release formulation. In order to prevent the scarring process frequently occurring as complication of glaucoma surgery, the compounds may be administered topically, for example, as eye drops. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with a specific antibody, targeting, for example, arthritic or fibrotic tissue. The liposomes will be targeted to and taken up selectively by the afflicted tissue.




The polypeptides of the invention are administered by any route that delivers an effective dosage to the desired site of action. The determination of a suitable route of administration and an effective dosage for a particular indication is within the level of skill in the art. Preferably for wound treatment, one administers the therapeutic compound directly to the site. Suitable dosage ranges for the polypeptides of the invention can be extrapolated from these dosages or from similar studies in appropriate animal models. Dosages can then be adjusted as necessary by the clinician to provide maximal therapeutic benefit.




3.12.2 Compositions/Formulations




Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. These pharmaceutical compositions may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen. When a therapeutically effective amount of protein or other active ingredient of the present invention is administered orally, protein or other active ingredient of the present invention will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet forrm, the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder contain from about 5 to 95% protein or other active ingredient of the present invention, and preferably from about 25 to 90% protein or other active ingredient of the present invention. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may bather contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of protein or other active ingredient of the present invention, and preferably from about 1 to 50% protein or other active ingredient of the present invention.




When a therapeutically effective amount of protein or other active ingredient of the present invention is administered by intravenous, cutaneous or subcutaneous injection, protein or other active ingredient of the present invention will be in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable protein or other active ingredient solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to protein or other active ingredient of the present invention, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For tarsmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.




For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained from a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.




Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.




For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.




Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.




The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides. In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.




A pharmaceutical carrier for the hydrophobic compounds of the invention is a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The co-solvent system may be the VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD:5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermnore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose. Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various types of sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein or other active ingredient stabilization may be employed.




The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Many of the active ingredients of the invention may be provided as salts with pharmaceutically compatible counter ions. Such pharmaceutically acceptable base addition salts are those salts which retain the biological effectiveness and properties of the free acids and which are obtained by reaction with inorganic or organic bases such as sodium hydroxide, magnesium hydroxide, ammonia, trialkylamine, dialkylamine, monoalkylamine, dibasic amino acids, sodium acetate, potassium benzoate, triethanol amine and the like.




The pharmaceutical composition of the invention may be in the form of a complex of the protein(s) or other active ingredient(s) of present invention along with protein or peptide antigens. The protein and/or peptide antigen will deliver a stimulatory signal to both B and T lymphocytes. B lymphocytes will respond to antigen through their surface immunoglobulin receptor. T lymphocytes will respond to antigen through the T cell receptor (TCR) following presentation of the antigen by MHC proteins. MHC.and structurally related proteins including those encoded by class I and class II MHC genes on host cells will serve to present the peptide antigen(s) to T lymphocytes. The antigen components could also be supplied as purified MHC-peptide complexes alone or with co-stimulatory molecules that can directly signal T cells. Alternatively antibodies able to bind surface immunoglobulin and other molecules on B cells as well as antibodies able to bind the TCR and other molecules on T cells can be combined with the pharmaceutical composition of the invention




The pharmaceutical composition of the invention may be in the form of a liposome in which protein of the present invention is combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids which exist in aggregated form as micelles, insoluble monolayers, liquid crystals, or lamellar layers in aqueous solution. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithins, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. Nos. 4,235,871; 4,501,728; 4,837,028; and 4,737,323, all of which are incorporated herein by reference.




The amount of protein or other active ingredient of the present invention in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of protein or other active ingredient of the present invention with which to treat each individual patient. Initially, the attending physician will administer low doses of protein or other active ingredient of the present invention and observe the patient's response. Larger doses of protein or other active ingredient of the present invention may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further. It is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.01 μg to about 100 mg (preferably about 0.1 μg to about 10 mg, more preferably about 0.1 μg to about 1 mg) of protein or other active ingredient of the present invention per kg body weight. For compositions of the present invention which are useful for bone, cartilage, tendon or ligament regeneration, the therapeutic method includes administering the composition topically, systematically, or locally as an implant or device. When administered, the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form. Further, the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage or tissue damage. Topical administration may be suitable for wound healing and tissue repair. Therapeutically useful agents other than a protein or other active ingredient of the invention which may also optionally be included in the composition as described above, may alternatively or additionally, be administered simultaneously or sequentially with the composition in the methods of the invention. Preferably for bone and/or cartilage formation, the composition would include a matrix capable of delivering the protein-containing or other active ingredient-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and optimally capable of being resorbed into the body. Such matrices may be formed of materials presently in use for other implanted medical applications.




The choice of matrix material is based on biocompatibility, biodegradability, mechanical properties, cosmetic appearance and interface properties. The particular application of the compositions will define the appropriate formulation. Potential matrices for the compositions may be biodegradable and chemically defined calcium sulfate, tricalcium phosphate, hydroxyapatite, polylactic acid, polyglycolic acid and polyanhydrides. Other potential materials are biodegradable and biologically well-defined, such as bone or dermal collagen. Further matrices are comprised of pure proteins or extracellular matrix components. Other potential matrices are nonbiodegradable and chemically defined, such as sintered hydroxyapatite, bioglass, aluminates, or other ceramics. Matrices may be comprised of combinations of any of the above mentioned types of material, such as polylactic acid and hydroxyapatite or collagen and tricalcium phosphate. The bioceramics may be altered in composition, such as in calcium-aluminate-phosphate and processing to alter pore size, particle size, particle shape, and biodegradability. Presently preferred is a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns. In some applications, it will be useful to utilize a sequestering agent, such as carboxymethyl cellulose or autologous blood clot, to prevent the protein compositions from disassociating from the matrix.




A preferred family of sequestering agents is cellulosic materials such as alkylcelluloses (including hydroxyalkylcelluloses), including methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropyl-methylcellulose, and carboxymethylcellulose, the most preferred being cationic salts of carboxymethylcellulose (CMC). Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer and poly(vinyl alcohol). The amount of sequestering agent useful herein is 0.5-20 wt %, preferably 1-10 wt % based on total formulation weight, which represents the amount necessary to prevent desorption of the protein from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the protein the opportunity to assist the osteogenic activity of the progenitor cells. In further compositions, proteins or other active ingredients of the invention may be combined with other agents beneficial to the treatment of the bone and/or cartilage defect, wound, or tissue in question. These agents include various growth factors such as epidermal growth factor (EGF), platelet derived growth factor (PDGF), transforming growth factors (TGF-α and TGF-β), and insulin-like growth factor (IGF).




The therapeutic compositions are also presently valuable for veterinary applications. Particularly domestic animals and thoroughbred horses, in addition to humans, are desired patients for such treatment with proteins or other active ingredients of the present invention. The dosage regimen of a protein-containing pharmaceutical composition to be used in tissue regeneration will be determined by the attending physician considering various factors which modify the action of the proteins, e.g., amount of tissue weight desired to be formed, the site of damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue (e.g., bone), the patient's age, sex, and diet, the severity of any infection, time of administration and other clinical factors. The dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition. For example, the addition of other known growth factors, such as IGF I (insulin like growth factor I), to the final composition, may also effect the dosage. Progress can be monitored by periodic assessment of tissue/bone growth and/or repair, for example, X-rays, histomorphometric determinations and tetracycline labeling.




Polynucleotides of the present invention can also be used for gene therapy. Such polynucleotides can be introduced either in vivo or ex vivo into cells for expression in a mammalian subject. Polynucleotides of the invention may also be administered by other known methods for introduction of nucleic acid into a cell or organism (including, without limitation, in the form of viral vectors or naked DNA). Cells may also be cultured ex vivo in the presence of proteins of the present invention in order to proliferate or to produce a desired effect on or activity in such cells. Treated cells can then be introduced in vivo for therapeutic purposes.




3.12.3 Effective Dosage




Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from appropriate in vitro assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that can be used to more accurately determine useful doses in humans. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC


50


as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the protein's biological activity). Such information can be used to more accurately determine useful doses in humans.




A therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms or a prolongation of survival in a patient. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD


50


(the dose lethal to 50% of the population) and the ED


50


(the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD


50


and ED


50


. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studiescan be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED


50


with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. See, e.g., Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p.1. Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the desired effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.




Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.




An exemplary dosage regimen for polypeptides or other compositions of the invention will be in the range of about 0.01 μg/kg to 100 mg/kg of body weight daily, with the preferred dose being about 0.1 μg/kg to 25 mg/kg of patient body weight daily, varying in adults and children. Dosing may be once daily, or equivalent doses may be delivered at longer or shorter intervals.




The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's age and weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.




3.12.4 Packaging




The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.




3.13 Antibodies




Also included in the invention are antibodies to proteins, or fragments of proteins of the invention. The term “antibody” as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen-binding site that specifically binds (immunoreacts with) an antigen. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, F


ab


, F


ab′


and F


(ab′)2


fragments, and an F


ab


expression library. In general, an antibody molecule obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule. Certain classes have subclasses as well, such as IgG


1


, IgG


2


, and others. Furthermore, in humans, the light chain may be a kappa chain or a lambda chain. Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species.




An isolated related protein of the invention may be intended to serve as an antigen, or a portion or fragment thereof, and additionally can be used as an immunogen to generate antibodies that inmmunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation. The full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens. An antigenic peptide fragment comprises at least 6 mino acid residues of the amino acid sequence of the full length protein, such as an amino acid sequence shown in SEQ ID NO: 1-948, and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope. Preferably, the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues. Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions.




In certain embodiments of the invention, at least one epitope encompassed by the antigenic peptide is a region of alpha-2-macroglobulin-like protein that is located on the surface of the protein, e.g., a hydrophilic region. A hydrophobicity analysis of the human related protein sequence will indicate which regions of a related protein are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production. As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981


, Proc. Nat. Acad. Sci. USA


78: 3824-3828; Kyte and Doolittle 1982


, J. Mol. Biol


. 157: 105-142, each of which is incorporated herein by reference in its entirety. Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.




A protein of the invention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.




The term “specific for” indicates that the variable regions of the antibodies of the invention recognize and bind polypeptides of the invention exclusively (i.e., able to distinguish the polypeptide of the invention from other similar polypeptides despite sequence identity, homology, or similarity found in the family of polypeptides), but may also interact with other proteins (for example,


S. aureus


protein A or other antibodies in ELISA techniques) through interactions with sequences outside the variable region of the antibodies, and in particular, in the constant region of the molecule. Screening assays to determine binding specificity of an antibody of the invention are well known and routinely practiced in the art. For a comprehensive discussion of such assays, see Harlow et al. (Eds), Antibodies A Laboratory Manual; Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1988), Chapter 6. Antibodies that recognize and bind fragments of the polypeptides of the invention are also contemplated, provided that the antibodies are first and foremost specific for, as defined above, full-length polypeptides of the invention. As with antibodies that are specific for full length polypeptides of the invention, antibodies of the invention that recognize fragments are those which can distinguish polypeptides from the same family of polypeptides despite inherent sequence identity, homology, or similarity found in the family of proteins.




Antibodies of the invention are useful for, for example, therapeutic purposes (by modulating activity of a polypeptide of the invention), diagnostic purposes to detect or quantitate a polypeptide of the invention, as well as purification of a polypeptide of the invention. Kits comprising an antibody of the invention for any of the purposes described herein are also comprehended. In general, a kit of the invention also includes a control antigen for which the antibody is immunospecific. The invention further provides a hybridoma that produces an antibody according to the invention. Antibodies of the invention are useful for detection and/or purification of the polypeptides of the invention.




Monoclonal antibodies binding to the protein of the invention may be useful diagnostic agents for the immunodetection of the protein. Neutralizing monoclonal antibodies binding to the protein may also be useful therapeutics for both conditions associated with the protein and also in the treatment of some forms of cancer where abnormal expression of the protein is involved. In the case of cancerous cells or leukemic cells, neutralizing monoclonal antibodies against the protein may be useful in detecting and preventing the metastatic spread of the cancerous cells, which may be mediated by the protein.




The labeled antibodies of the present invention can be used for in vitro, in vivo, and in situ assays to identify cells or tissues in which a fragment of the polypeptide of interest is expressed. The antibodies may also be used directly in therapies or other diagnostics. The present invention further provides the above-described antibodies immobilized on a solid support. Examples of such solid supports include plastics such as polycarbonate, complex carbohydrates such as agarose and Sepharose®, acrylic resins and such as polyacrylamide and latex beads. Techniques for coupling antibodies to such solid supports are well known in the art (Weir, D. M. et al., “Handbook of Experimental Immunology” 4th Ed., Blackwell Scientific Publications, Oxford, England, Chapter 10 (1986); Jacoby, W. D. et al., Meth. Enzym. 34 Academic Press, N.Y. (1974)). The immobilized antibodies of the present invention can be used for in vitro, in vivo, and in situ assays as well as for immuno-affinity purification of the proteins of the present invention.




Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies directed against a protein of the invention, or against derivatives, fragments, analogs homologs or orthologs thereof (see, for example, Antibodies: A Laboratory Manual, Harlow E, and Lane D, 1988, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., incorporated herein by reference). Some of these antibodies are discussed below.




3.13.1 Polyclonal Antibodies




For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by one or more injections with the native protein, a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the immunogenic protein, or a recombinantly expressed immunogenic protein. Furthermore, the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. 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.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents. Additional examples of adjuvants that can be employed include MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).




The polyclonal antibody molecules directed against the immunogenic protein can be isolated from the manmmal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protein A or protein G, which provide primarily the IgG faction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, may be immobilized on a column to purify the immune specific antibody by immunoaffinity chromatography. Purification of inmmunoglobulins is discussed, for example, by D. Wilkinson (The Scientist, published by The Scientist, Inc., Philadelphia Pa., Vol. 14, No. 8 (Apr. 17, 2000), pp. 25-28).




3.13.2 Monoclonal Antibodies




The term “monoclonal antibody” (MAb) or “monoclonal antibody composition”, as used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a unique light chain gene product and a unique heavy chain gene product. In particular, the complementarity determining regions (CDRs) of the monoclonal antibody are identical in all the molecules of the population. MAbs thus contain an antigen-binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it.




Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein,


Nature


, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes can be immunized in vitro.




The immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof. Generally, either peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding,


Monoclonal Antibodies: Principles and Practice


, Academic Press, (1986) pp. 59-103). Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (“HAT medium”), which substances prevent the growth of HGPRT-deficient cells.




Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, Calif. and the American Type Culture Collection, Manassas, Va. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor,


J. Immunol


., 133:3001 (1984); Brodeur et al.,


Monoclonal Antibody Production Techniques and Applications


, Marcel Dekker, Inc., New York, (1987) pp. 51-63).




The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard,


Anal. Biochem


., 107:220 (1980). Preferably, antibodies having a high degree of specificity and a high binding affinity for the target antigen are isolated.




After the desired hybridoma cells are identified, the clones can be subcloned by limiting dilution procedures and grown by standard methods. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal.




The monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.




The monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Pat. No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison,


Nature


368, 812-13 (1994)) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.




3.13.3 Humanized Antibodies




The antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the administered immunoglobulin. Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′)


2


or other antigen-binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin. Humanization can be performed following the method of Winter and co-workers (Jones et al.,


Nature


, 321:522-525 (1986); Riechmann et al.,


Nature


, 332:323-327 (1988); Verhoeyen et al.,


Science


, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. (See also U.S. Pat. No. 5,225,539). In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies can also comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al., 1986; Riechmann et al., 1988; and Presta,


Curr. Op. Struct. Biol


., 2:593-596 (1992)).




3.13.4 Human Antibodies




Fully human antibodies relate to antibody molecules in which essentially the entire sequences of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed “human antibodies”, or “fully human antibodies” herein. Human monoclonal antibodies can be prepared by 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: M


ONOCLONAL


A


NTIBODIES AND


C


ANCER


T


HERAPY


, 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: M


ONOCLONAL


A


NTIBODIES AND


C


ANCER


T


HERAPY


, Alan R. Liss, Inc., pp. 77-96).




In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter,


J. Mol. Biol


., 227:381 (1991); Marks et al.,


J. Mol. Biol


., 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (


Bio/Technology


10, 779-783 (1992)); Lonberg et al. (


Nature


368 856-859 (1994)); Morrison (Nature 368, 812-13 (1994)); Fishwild et al, (


Nature Biotechnology


14, 845-51 (1996)); Neuberger (


Nature Biotechnology


14, 826 (1996)); and Lonberg and Huszar (


Intern. Rev. Immunol


. 13 65-93 (1995)).




Human antibodies may additionally be produced using taansgenic nonhuman animals that are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication WO94/02602). The endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain immunoglobulins are inserted into the host's genome. The human genes are incorporated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The preferred embodiment of such a nonhuman animal is a mouse, and is termed the Xenomouse™ as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells that secrete fully human immunoglobulins. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.




An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Pat. No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent rearrangement of the locus and to prevent formation of a transcript of a rearranged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker.




A method for producing an antibody of interest, such as a human antibody, is disclosed in U.S. Pat. No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell. The hybrid cell expresses an antibody containing the heavy chain and the light chain.




In a further improvement on this procedure, a method for identifying a clinically relevant epitope on an imnmunogen, and a correlative method for selecting an antibody that binds immunospecifically to the relevant epitope with high affinity, are disclosed in PCT publication WO 99/53049.




3.13.5 Fab Fragments and Single Chain Antibodies




According to the invention, techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S. Pat. No. 4,946,778). In addition, methods can be adapted for the construction of F


ab


expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal F


ab


fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof. Antibody fragments that contain the idiotypes to a protein antigen 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 F


ab


fragment generated by reducing the disulfide bridges of an F


(ab′)2


fragment; (iii) an F


ab


fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) F


v


fragments.




3.13.6 Bispecific Antibodies




Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for an antigenic protein of the invention. The second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit.




Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-exprcssion of two immunoglobulin heavyhain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello,


Nature


, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al., 1991


EMBO J


., 10:3655-3659.




Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al.,


Methods in Enzymology


, 121:210 (1986).




According to another approach described in WO 96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers that are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.




Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab′)


2


bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al.,


Science


229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab′)


2


fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab′ fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab′-TNB derivatives is then reconverted to the Fab′-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab′-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.




Additionally, Fab′ fragments can be directly recovered from


E. coli


and chemically coupled to form bispecific antibodies. Shalaby et al.,


J. Exp. Med


. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab′)


2


molecule. Each Fab′ fragment was separately secreted from


E. coli


and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.




Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al.,


J. Immunol


. 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab′ portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The “diabody” technology described by Hollinger et al.,


Proc. Natl. Acad. Sci. USA


90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (V


H


) connected to a lighthain variable domain (V


L


) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the V


H


and V


L


domains of one fragment are forced to pair with the complementary V


L


and V


H


domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al.,


J. Immunol


. 152:5368 (1994).




Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al.,


J. Immunol


. 147:60 (1991).




Exemplary bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention. Alternatively, an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or Fc receptors for IgG (Fc R), such as Fc RI (CD64), Fe RII (CD32) and Fc RIII (CD16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen. Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA. Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor TF).




3.13.7 Heteroconjugate Antibodies




Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360; WO 92/200373; EP 03089). It is contemplated that the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Pat. No. 4,676,980.




3.13.8 Effector Function Engineering




It can be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) can be introduced into the Fe region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med., 176: 1191-1195 (1992) and Shopes, J. Imunol., 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design, 3: 219-230 (1989).




3.13.9 Immunoconjugates




The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).




Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include


212


Bi,


131


I,


131


In,


90


y, and


186


Re.




Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (pazidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.




In another embodiment, the antibody can be conjugated to a “receptor” (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a “ligand” (e.g., avidin) that is in turn conjugated to a cytotoxic agent.




3.14 Computer Readable Sequences




In one application of this embodiment, a nucleotide sequence of the present invention can be recorded on computer readable media. As used herein, “computer readable media” refers to any medium which can be read and accessed directly by a computer. Such media include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage medium, and magnetic tape; optical storage media such as CD-ROM; electrical storage media such as RAM and ROM; and hybrids of these categories such as magnetic/optical storage media. A skilled artisan can readily appreciate how any of the presently known computer readable mediums can be used to create a manufacture comprising computer readable medium having recorded thereon a nucleotide sequence of the present invention. As used herein, “recorded” refers to a process for storing information on computer readable medium. A skilled artisan can readily adopt any of the presently known methods for recording information on computer readable medium to generate manufactures comprising the nucleotide sequence information of the present invention.




A variety of data storage structures are available to a skilled artisan for creating a computer readable medium having recorded thereon a nucleotide sequence of the present invention. The choice of the data storage structure will generally be based on the means chosen to access the stored information. In addition, a variety of data processor programs and formats can be used to store the nucleotide sequence information of the present invention on computer readable medium. The sequence information can be represented in a word processing text file, formatted in commercially-available software such as WordPerfect and Microsoft Word, or represented in the form of an ASCII file, stored in a database application, such as DB2, Sybase, Oracle, or the like. A skilled artisan can readily adapt any number of data processor structuring formats (e.g. text file or database) in order to obtain computer readable medium having recorded thereon the nucleotide sequence information of the present invention.




By providing any of the nucleotide sequences SEQ ID NOs: 1-948 or a representative fragment thereof; or a nucleotide sequence at least 95% identical to any of the nucleotide sequences of SEQ ID NOs: 1-948 in computer readable form, a skilled artisan can routinely access the sequence information for a variety of purposes. Computer software is publicly available which allows a skilled artisan to access sequence information provided in a computer readable medium. The examples which follow demonstrate how software which implements the BLAST (Altschul et al., J. Mol. Biol. 215:403-410 (1990)) and BLAZE (Brutlag et al., Comp. Chem. 17:203-207 (1993)) search algorithms on a Sybase system is used to identify open reading frames (ORFs) within a nucleic acid sequence. Such ORFs may be protein encoding fragments and may be useful in producing commercially important proteins such as enzymes used in fermentation reactions and in the production of commercially useful metabolites.




As used herein, “a computer-based system” refers to the hardware means, software means, and data storage means used to analyze the nucleotide sequence information of the present invention. The minimum hardware means of the computer-based systems of the present invention comprises a central processing unit (CPU), input means, output means, and data storage means. A skilled artisan can readily appreciate that any one of the currently available computer-based systems are suitable for use in the present invention. As stated above, the computer-based systems of the present invention comprise a data storage means having stored therein a nucleotide sequence of the present invention and the necessary hardware means and software means for supporting and implementing a search means. As used herein, “data storage means” refers to memory which can store nucleotide sequence information of the present invention, or a memory access means which can access manufactures having recorded thereon the nucleotide sequence information of the present invention.




As used herein, “search means” refers to one or more programs which are implemented on the computer-based system to compare a target sequence or target structural motif with the sequence information stored within the data storage means. Scarch means are used to identify fragments or regions of a known sequence which match a particular target sequence or target motif A variety of known algorithms are disclosed publicly and a variety of commercially available software for conducting search means are and can be used in the computer-based systems of the present invention. Examples of such software includes, but is not limited to, Smnith-Waterman, MacPattern (EMBL), BLASTN and BLASTA (NPOLYPEPTIDEIA). A skilled artisan can readily recognize that any one of the available algorithms or implementing software packages for conducting homology searches can be adapted for use in the present computer-based systems. As used herein, a “target sequence” can be any nucleic acid or amino acid sequence of six or more nucleotides or two or more amino acids. A skilled artisan can readily recognize that the longer a target sequence is, the less likely a target sequence will be present as a random occurrence in the database. The most preferred sequence length of a target sequence is from about 10 to 300 amino acids, more preferably from about 30 to 100 nucleotide residues. However, it is well recognized that searches for commercially important fragments, such as sequence fragments involved in gene expression and protein processing, may be of shorter length.




As used herein, “a target structural motif,” or “target motif,” refers to any rationally selected sequence or combination of sequences in which the sequence(s) are chosen based on a three-dimensional configuration which is formed upon the folding of the target motif. There are a variety of target motifs known in the art. Protein target motifs include, but are not limited to, enzyme active sites and signal sequences. Nucleic acid target motifs include, but are not limited to, promoter sequences, hairpin structures and inducible expression elements (protein binding sequences).




3.15 Triple Helix Formation




In addition, the fragments of the present invention, as broadly described, can be used to control gene expression through triple helix formation or antisense DNA or RNA, both of which methods are based on the binding of a polynucleotide sequence to DNA or RNA. Polynucleotides suitable for use in these methods are preferably 20 to 40 bases in length and are designed to be complementary to a region of the gene involved in transcription (triple helix—see Lee et al., Nucl. Acids Res. 6:3073 (1979); Cooney et al., Science 15241:456 (1988); and Dervan et al., Science 251:1360 (1991)) or to the mRNA itself (antisense—Olmno, J. Neurochem. 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)). Triple helix-formation optimally results in a shut-off of RNA transcription from DNA, while antisense RNA hybridization blocks translation of an mRNA molecule into polypeptide. Both techniques have been demonstrated to be effective in model systems. Information contained in the sequences of the present invention is necessary for the design of an antisense or triple helix oligonucleotide.




3.16 Diagnostic Assays and Kits




The present invention further provides methods to identify the presence or expression of one of the ORFs of the present invention, or homolog thereof, in a test sample, using a nucleic acid probe or antibodies of the present invention, optionally conjugated or otherwise associated with a suitable label.




In general, methods for detecting a polynucleodide of the invention can comprise contacting a sample with a compound that binds to and forms a complex with the polynucleotide for a period sufficient to form the complex, and detecting the complex, so that if a complex is detected, a polynucleotide of the invention is detected in the sample. Such methods can also comprise contacting a sample under stringent hybridization conditions with nucleic acid primers that anneal to a polynucleotide of the invention under such conditions, and amplifying annealed polynucleotides, so that if a polynucleotide is amplified, a polynucleotide of the invention is detected in the sample.




In general, methods for detecting a polypeptide of the invention can comprise contacting a sample with a compound that binds to and forms a complex with the polypeptide for a period sufficient to form the complex, and detecting the complex, so that if a complex is detected, a polypeptide of the invention is detected in the sample.




In detail, such methods comprise incubating a test sample with one or more of the antibodies or one or more of the nucleic acid probes of the present invention and assaying for binding of the nucleic acid probes or antibodies to components within the test sample.




Conditions for incubating a nucleic acid probe or antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the nucleic acid probe or antibody used in the assay. One skilled in the art will recognize that any one of the commonly available hybridization, amplification or immunological assay formats can readily be adapted to employ the nucleic acid probes or antibodies of the present invention. Examples of such assays can be found in Chard, T., An Introduction to Radioimmunoassay and Related Techniques, Elsevier Science Publishers, Amsterdam, The Netherlands (1986); Bullock, G. R. et al., Techniques in Immunocytochemistry, Academic Press, Orlando, Fla. Vol. 1 (1982), Vol. 2 (1983), Vol. 3 (1985); Tijssen, P., Practice and Theory of immunoassays: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1985). The test samples of the present invention include cells, protein or membrane extracts of cells, or biological fluids such as sputum, blood, serum, plasma, or urine. The test sample used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed. Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can be readily be adapted in order to obtain a sample which is compatible with the system utilized.




In another embodiment of the present invention, kits are provided which contain the necessary reagents to carry out the assays of the present invention. Specifically, the invention provides a compartment kit to receive, in close confinement, one or more containers which comprises: (a) a first container comprising one of the probes or antibodies of the present invention; and (b) one or more other containers comprising one or more of the following: wash reagents, reagents capable of detecting presence of a bound probe or antibody.




In detail, a compartment kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers or strips of plastic or paper. Such containers allows one to efficiently transfer reagents from one compartment to another compartment such that the samples and reagents are not cross-contaminated, and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another. Such containers will include a container which will accept the test sample, a container which contains the antibodies used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents used to detect the bound antibody or probe. Types of detection reagents include labeled nucleic acid probes, labeled secondary antibodies, or in the alternative, if the primary antibody is labeled, the enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody. One skilled in the art will readily recognize that the disclosed probes and antibodies of the present invention can be readily incorporated into one of the established kit formats which are well known in the art.




3.17 Medical Imaging




The novel polypeptides and binding partners of the invention are useful in medical imaging of sites expressing the molecules of the invention (e.g., where the polypeptide of the invention is involved in the immune response, for imaging sites of inflammation or infection). See, e.g., Kunkel et al., U.S. Pat. No. 5,413,778. Such methods involve chemical attachment of a labeling or imaging agent, administration of the labeled polypeptide to a subject in a pharmaceutically acceptable carrier, and imaging the labeled polypeptide in vivo at the target site.




3.18 Screening Assays




Using the isolated proteins and polynucleotides of the invention, the present invention further provides methods of obtaining and identifying agents which bind to a polypeptide encoded by an ORF corresponding to any of the nucleotide sequences set forth in SEQ ID NOs: 1-948, or bind to a specific domain of the polypeptide encoded by the nucleic acid. In detail, said method comprises the steps of:




(a) contacting an agent with an isolated protein encoded by an ORF of the present invention, or nucleic acid of the invention; and




(b) determining whether the agent binds to said protein or said nucleic acid.




In general, therefore, such methods for identifying compounds that bind to a polynucleotide of the invention can comprise contacting a compound with a polynucleotide of the invention for a time sufficient to form a polynucleotide/compound complex, and detecting the complex, so that if a polynucleotide/compound complex is detected, a compound that binds to a polynucleotide of the invention is identified.




Likewise, in general, therefore, such methods for identifying compounds that bind to a polypeptide of the invention can comprise contacting a compound with a polypeptide of the invention for a time sufficient to form a polypeptide/compound complex, and detecting the complex, so that if a polypeptide/compound complex is detected, a compound that binds to a polynucleotide of the invention is identified.




Methods for identifying compounds that bind to a polypeptide of the invention can also comprise contacting a compound with a polypeptide of the invention in a cell for a time sufficient to form a polypeptide/compound complex, wherein the complex drives expression of a receptor gene sequence in the cell, and detecting the complex by detecting reporter gene sequence expression, so that if a polypeptide/compound complex is detected, a compound that binds a polypeptide of the invention is identified.




Compounds identified via such methods can include compounds which modulate the activity of a polypeptide of the invention (that is, increase or decrease its activity, relative to activity observed in the absence of the compound). Alternatively, compounds identified via such methods can include compounds which modulate the expression of a polynucleotide of the invention (that is, increase or decrease expression relative to expression levels observed in the absence of the compound). Compounds, such as compounds identified via the methods of the invention, can be tested using standard assays well known to those of skill in the art for their ability to modulate activity/expression.




The agents screened in the above assay can be, but are not limited to, peptides, carbohydrates, vitamin derivatives, or other pharmaceutical agents. The agents can be selected and screened at random or rationally selected or designed using protein modeling techniques.




For random screening, agents such as peptides, carbohydrates, pharmaceutical agents and the like are selected at random and are assayed for their ability to bind to the protein encoded by the ORF of the present invention. Alternatively, agents may be rationally selected or designed. As used herein, an agent is said to be “rationally selected or designed” when the agent is chosen based on the configuration of the particular protein. For example, one skilled in the art can readily adapt currently available procedures to generate peptides, pharmaceutical agents and the like, capable of binding to a specific peptide sequence, in order to generate rationally designed antipeptide peptides, for example see Hurby et al., Application of Synthetic Peptides: Antisense Peptides,” In Synthetic Peptides, A User's Guide, W. H. Freeman, NY (1992), pp. 289-307, and Kaspczak et al., Biochemistry 28:9230-8 (1989), or pharmaceutical agents, or the like.




In addition to the foregoing, one class of agents of the present invention, as broadly described, can be used to control gene expression through binding to one of the ORFs or EMFs of the present invention. As described above, such agents can be randomly screened or rationally designed/selected. Targeting the ORF or EMF allows a skilled artisan to design sequence specific or element specific agents, modulating the expression of either a single ORF or multiple ORFs which rely on the same EMF for expression control. One class of DNA binding agents are agents which contain base residues which hybridize or form a triple helix formation by binding to DNA or RNA. Such agents can be based on the classic phosphodiester, ribonucleic acid backbone, or can be a variety of sulfliydryl or polymeric derivatives which have base attachment capacity.




Agents suitable for use in these methods preferably contain 20 to 40 bases and are designed to be complementary to a region of the gene involved in transcription (triple helix—see Lee et al., Nucl. Acids Res. 6:3073 (1979); Cooney et al., Science 241:456 (1988); and Dervan et al., Science 251:1360 (1991)) or to the mRNA itself (antisense—Okano, J. Neurochem. 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)). Triple helix-formation optimally results in a shut-off of RNA transcription from DNA, while antisense RNA hybridization blocks translation of an mRNA molecule into polypeptide. Both techniques have been demonstrated to be effective in model systems. Information contained in the sequences of the present invention is necessary for the design of an antisense or triple helix oligonucleotide and other DNA binding agents.




Agents which bind to a protein encoded by one of the ORFs of the present invention can be used as a diagnostic agent. Agents which bind to a protein encoded by one of the ORFs of the present invention can be formulated using known techniques to generate a pharmaceutical composition.




3.19 Use of Nucleic Acids as Probes




Another aspect of the subject invention is to provide for polypeptide-specific nucleic acid hybridization probes capable of hybridizing with naturally occurring nucleotide sequences. The hybridization probes of the subject invention may be derived from any of the nucleotide sequences SEQ ID NOs: 1-948. Because the corresponding gene is only expressed in a limited number of tissues, a hybridization probe derived from of any of the nucleotide sequences SEQ ID NOs: 1-948 can be used as an indicator of the presence of RNA of cell type of such a tissue in a sample.




Any suitable hybridization technique can be employed, such as, for example, in situ hybridization. PCR as described in U.S. Pat. Nos. 4,683,195 and 4,965,188 provides additional uses for oligonucleotides based upon the nucleotide sequences. Such probes used in PCR may be of recombinant origin, may be chemically synthesized, or a mixture of both. The probe will comprise a discrete nucleotide sequence for the detection of identical sequences or a degenerate pool of possible sequences for identification of closely related genomic sequences.




Other means for producing specific hybridization probes for nucleic acids include the cloning of nucleic acid sequences into vectors for the production of mRNA probes. Such vectors are known in the art and are commercially available and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerase as T7 or SP6 RNA polymerase and the appropriate radioactively labeled nucleotides. The nucleotide sequences may be used to construct hybridization probes for mapping their respective genomic sequences. The nucleotide sequence provided herein may be mapped to a chromosome or specific regions of a chromosome using well known genetic and/or chromosomal mapping techniques. These techniques include in situ hybridization, linkage analysis against known chromosomal markers, hybridization screening with libraries or flow-sorted chromosomal preparations specific to known chromosomes, and the like. The technique of fluorescent in situ hybridization of chromosome spreads has been described, among other places, in Verma et al (1988) Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York N.Y.




Fluorescent in situ hybridization of chromosomal preparations and other physical chromosome mapping techniques may be correlated with additional genetic map data. Examples of genetic map data can be found in the 1994 Genome Issue of Science (265:1981f). Correlation between the location of a nucleic acid on a physical chromosomal map and a specific disease (or predisposition to a specific disease) may help delimit the region of DNA associated with that genetic disease. The nucleotide sequences of the subject invention may be used to detect differences in gene sequences between normal, carrier or affected individuals.




3.20 Preparation of Support Bound Oligonucleotides




Oligonucleotides, i.e., small nucleic acid segments, may be readily prepared by, for example, directly synthesizing the oligonucleotide by chemical means, as is commonly practiced using an automated oligonucleotide synthesizer.




Support bound oligonucleotides may be prepared by any of the methods known to those of skill in the art using any suitable support such as glass, polystyrene or Teflon. One strategy is to precisely spot oligonucleotides synthesized by standard synthesizers. Inmobilization can be achieved using passive adsorption (Inouye & Hondo, (1990) J. Clin. Microbiol. 28(6) 1469-72); using UV light (Nagata et al., 1985; Dahlen et al., 1987; Morrissey & Collins, (1989) Mol. Cell Probes 3(2) 189-207) or by covalent binding of base modified DNA (Keller et al., 1988; 1989); all references being specifically incorporated herein.




Another strategy that may be employed is the use of the strong biotin-streptavidin interaction as a linker. For example, Broude et a. (1994) Proc. Natl. Acad. Sci. USA 91(8) 3072-6, describe the use of biotinylated probes, although these are duplex probes, that are immobilized on streptavidin-coated magnetic beads. Streptavidin-coated beads may be purchased from Dynal, Oslo. Of course, this same linking chemistry is applicable to coating any surface with streptavidin. Biotinylated probes may be purchased from various sources, such as, e.g., Operon Technologies (Alameda, Calif.).




Nunc Laboratories (Naperville, Ill.) is also selling suitable material that could be used. Nunc Laboratories have developed a method by which DNA can be covalently bound to the microwell surface termed Covalink NH. CovaLink NH is a polystyrene surface grafted with secondary amino groups (>NH) that serve as bridge-heads for further covalent coupling. CovaLink Modules may be purchased from Nunc Laboratories. DNA molecules may be bound to CovaLink exclusively at the 5′-end by a phosphoramidate bond, allowing immobilization of more than 1 pmol of DNA (Rasmussen et al., (1991) Anal. Biochem. 198(1) 138-42).




The use of CovaLink NH strips for covalent binding of DNA molecules at the 5′-end has been described (Rasmussen et al., (1991). In this technology, a phosphoramidate bond is employed (Chu et al., (1983) Nucleic Acids Res. 11(8) 6513-29). This is beneficial as immobilization using only a single covalent bond is preferred. The phosphoramnidate bond joins the DNA to the CovaLink NH secondary amino groups that are positioned at the end of spacer arms covalently grafted onto the polystyrene surface through a 2 nm long spacer arm. To link an oligonucleotide to CovaLink NH via an phosphoramidate bond, the oligonucleotide terminus must have a 5′-end phosphate group. It is, perhaps, even possible for biotin to be covalently bound to CovaLink and then streptavidin used to bind the probes.




More specifically, the linkage method includes dissolving DNA in water (7.5 ng/ul) and denaturing for 10 min. at 95° C. and cooling on ice for 10 min. Ice-cold 0.1 M 1-methylimidazole, pH 7.0 (1-MeIm


7


), is then added to a final concentration of 10 mM 1-MeIm


7


. A ss DNA solution is then dispensed into CovaLink NH strips (75 ul/well) standing on ice.




Carbodiimide 0.2 M 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (EDC), dissolved in 10 mM 1-MeIm


7


, is made fresh and 25 ul added per well. The strips are incubated for 5 hours at 50° C. After incubation the strips are washed using, e.g., Nunc-Immuno Wash; first the wells are washed 3 times, then they are soaked with washing solution for 5 min., and finally they are washed 3 times (where in the washing solution is 0.4 N NaOH, 0.25% SDS heated to 50° C.).




It is contemplated that a further suitable method for use with the present invention is that described in PCT Patent Application WO 90/03382 (Southern & Maskos), incorporated herein by reference. This method of preparing an oligonucleotide bound to a support involves attaching a nucleoside 3′-reagent through the phosphate group by a covalent phosphodiester link to aliphatic hydroxyl groups carried by the support. The oligonucleotide is then synthesized on the supported nucleoside and protecting groups removed from the synthetic oligonucleotide chain under standard conditions that do not cleave the oligonucleotide from the support. Suitable reagents include nucleoside phosphoramidite and nucleoside hydrogen phosphorate.




An on-chip strategy for the preparation of DNA probe for the preparation of DNA probe arrays may be employed. For example, addressable laser-activated photodeprotection may be employed in the chemical synthesis of oligonucleotides directly on a glass surface, as described by Fodor et al. (1991) Science 251(4995) 767-73, incorporated herein by reference. Probes may also be immobilized on nylon supports as described by Van Ness et al. (1991) Nucleic Acids Res. 19(12) 3345-50; or linked to Teflon using the method of Duncan & Cavalier (1 988) Anal. Biochem. 169(1) 104-8; all references being specifically incorporated herein.




To link an oligonucleotide to a nylon support, as described by Van Ness et al. (1991), requires activation of the nylon surface via alkylation and selective activation of the 5′-amine of oligonucleotides with cyanuric chloride.




One particular way to prepare support bound oligonucleotides is to utilize the light-generated synthesis described by Pease et al., (1994) PNAS USA 91(11) 5022-6, incorporated herein by reference). These authors used current photolithographic techniques to generate arrays of immobilized oligonucleotide probes (DNA chips). These methods, in which light is used to direct the synthesis of oligonucleotide probes in high-density, miniaturized arrays, utilize photolabile 5′-protected N-acyl-deoxynucleoside phosphoramidites, surface linker chemistry and versatile combinatorial synthesis strategies. A matrix of 256 spatially defined oligonucleotide probes may be generated in this manner.




3.21 Preparation of Nucleic Acid Fragments




The nucleic acids may be obtained from any appropriate source, such as cDNAs, genomic DNA, chromosomal DNA, iicrodissected chromosome bands, cosmid or YAC inserts, and RNA, including mRNA without any amplification steps. For example, Sambrook et al. (1989) describes three protocols for the isolation of high molecular weight DNA from mammalian cells (p. 9.14-9.23).




DNA fragments may be prepared as clones in M13, plasmid or lambda vectors and/or prepared directly from genomic DNA or cDNA by PCR or other amplification methods. Samples may be prepared or dispensed in multiwell plates. About 100-1000 ng of DNA samples may be prepared in 2-500 ml of final volume.




The nucleic acids would then be fragmented by any of the methods known to those of slill in the art including, for example, using restriction enzymes as described at 9.24-9.28 of Sambrook et al. (1989), shearing by ultrasound and NaOH treatment.




Low pressure shearing is also appropriate, as described by Schriefer et al. (1990) Nucleic Acids Res. 18(24) 7455-6, incorporated herein by reference). In this method, DNA samples are passed through a small French pressure cell at a variety of low to intermediate pressures. A lever device allows controlled application of low to intermediate pressures to the cell. The results of these studies indicate that low-pressure shearing is a useful alternative to sonic and enzymatic DNA iragmentation methods.




One particularly suitable way for fragmenting DNA is contemplated to be that using the two base recognition endonuclease, CviJI, described by Fitzgerald et al. (1992) Nucleic Acids Res. 20(14) 3753-62. These authors described an approach for the rapid fragmentation and fractionation of DNA into particular sizes that they contemplated to be suitable for shotgun cloning and sequencing.




The restriction endonuclease CviJI normally cleaves the recognition sequence PuGCPy between the G and C to leave blunt ends. Atypical reaction conditions, which alter the specificity of this enzyme (CviJI**), yield a quasi-random distribution of DNA fragments form the small molecule pUC19 (2688 base pairs). Fitzgerald et al. (1992) quantitatively evaluated the randomness of this fragmentation strategy, using a CviJI** digest of pUC19 that was size fractionated by a rapid gel filtration method and directly ligated, without end repair, to a lac Z minus M13 cloning vector. Sequence analysis of 76 clones showed that CviJI** restricts pyGCPy and PuGCPu, in addition to PuGCPy sites, and that new sequence data is accumulated at a rate consistent with random fragmentation.




As reported in the literature, advantages of this approach compared to sonication and agarose gel fractionation include: smaller amounts of DNA are required (0.2-0.5 ug instead of 2-5 ug); and fewer steps are involved (no preligation, end repair, chemical extraction, or agarose gel electrophoresis and elution are needed




Irrespective of the manner in which the nucleic acid fragments are obtained or prepared, it is important to denature the DNA to give single stranded pieces available for hybridization. This is achieved by incubating the DNA solution for 2-5 minutes at 80-90° C. The solution is then cooled quickly to 2° C. to prevent renaturation of the DNA fragments before they are contacted with the chip. Phosphate groups must also be removed from genomic DNA by methods known in the art.




3.22 Preparation of DNA Arrays




Arrays may be prepared by spotting DNA samples on a support such as a nylon membrane. Spotting may be performed by using arrays of metal pins (the positions of which correspond to an array of wells in a microtiter plate) to repeated by transfer of about 20 nl of a DNA solution to a nylon membrane. By offset printing, a density of dots higher than the density of the wells is achieved. One to 25 dots may be accommodated in 1 mm


2


, depending on the type of label used. By avoiding spotting in some preselected number of rows and columns, separate subsets (subarrays) may be formed. Samples in one subarray may be the same genomic segment of DNA (or the same gene) from different individuals, or may be different, overlapped genomic clones. Each of the subarrays may represent replica spotting of the same samples. In one example, a selected gene segment may be amplified from 64 patients. For each patient, the amplified gene segment may be in one 96-well plate (all 96 wells containing the same sample). A plate for each of the 64 patients is prepared. By using a 96-pin device, all samples may be spotted on one 8×12 cm membrane. Subarrays may contain 64 samples, one from each patient. Where the 96 subarrays are identical, the dot span may be 1 mm


2


and there may be a 1 mm space between subarrays.




Another approach is to use membranes or plates (available from NUNC, Naperville, Ill.) which may be partitioned by physical spacers e.g. a plastic grid molded over the membrane, the grid being similar to the sort of membrane applied to the bottom of multiwell plates, or hydrophobic strips. A fixed physical spacer is not preferred for imaging by exposure to flat phosphor-storage screens or x-ray films.




The present invention is illustrated in the following examples. Upon consideration of the present disclosure, one of skill in the art will appreciate that many other embodiments and variations may be made in the scope of the present invention. Accordingly, it is intended that the broader aspects of the present invention not be limited to the disclosure of the following examples. The present invention is not to be limited in scope by the exemplified embodiments which are intended as illustrations of single aspects of the invention, and compositions and methods which are functionally equivalent are within the scope of the invention. Indeed, numerous modifications and variations in the practice of the invention are expected to occur to those skilled in the art upon consideration of the present preferred embodiments. Consequently, the only limitations which should be placed upon the scope of the invention are those which appear in the appended claims.




All references cited within the body of the instant specification are hereby incorporated by reference in their entirety.




4.0 EXAMPLES




4.1 Example 1




Novel Nucleic Acid Sequences Obtained from Various Libraries




A plurality of novel nucleic acids were obtained from cDNA libraries prepared from various human tissues and in some cases isolated from a genomic library derived from human chromosome using standard PCR, SBH sequence signature analysis and Sanger sequencing techniques. The inserts of the library were amplified with PCR using primers specific for the vector sequences which flank the inserts. Clones from cDNA libraries were spotted on nylon membrane filters and screened with oligonucleotide probes (e.g., 7-mers) to obtain signature sequences. The clones were clustered into groups of similar or identical sequences. Representative clones were selected for sequencing.




In some cases, the 5′ sequence of the amplified inserts was then deduced using a typical Sanger sequencing protocol. PCR products were purified and subjected to fluorescent dye terminator cycle sequencing. Single pass gel sequencing was done using a 377 Applied Biosystems (ABI) sequencer to obtain the novel nucleic acid sequences. In some cases RACE (Random Amplification of cDNA Ends) was performed to further extend the sequence in the 5′ direction.




4.2 Example 2




Novel Nucleic Acids




The novel nucleic acids of the present invention of the invention were assembled from sequences that were obtained from a cDNA library by methods described in Example 1 above, and in some cases sequences obtained from one or more public databases, The nucleic acids were assembled using an EST sequence as a seed. Then a recursive algorithm was used to extend the seed EST into an extended assemblage, by pulling additional sequences from different databases (i.e., Hyseq's database containing EST sequences, dbEST version 119, gb pri 119, and UniGene version 119) that belong to this assemblage. The algorithm terminated when there was no additional sequences from the above databases that would extend the assemblage. Inclusion of component sequences into the assemblage was based on a BLASTN hit to the extending assemblage with BLAST score greater than 300 and percent identity greater than 95%.




Using PHRAP (Univ. of Washington) or CAP4 (Paracel), a full length gene cDNA sequence and its corresponding protein sequence were generated from the assemblage. Any frame shifts and incorrect stop codons were conrected by hand editing. During editing, the sequence was checked using FASTY and/or BLAST against Genbank (i.e., dbEST version 121, gb pri 121, UniGene version 121, Genpept release 121). Other computer programs which may have been used in the editing process were phredPhrap and Consed (University of Washington) and ed-ready, ed-ext and cg-zip-2 (Hyseq, Inc.). The full-length nucleotide and amino acid sequences, including splice variants resulting from these procedures are shown in the Sequence Listing as SEQ ID NO: 1-948.




Table 1 shows the various tissue sources of SEQ ID NO: 1-948.




The homology for SEQ ID NO: 1-948 were obtained by a BLASTP version 2.0al 19MP-WashU search against Genpept release 120 and the amino acid version of Geneseq released on Oct. 26, 2000, using BLAST algorithm. The results showed homologues for SEQ ID NO: 1-948 from Genpept. The homologues with identifiable functions for SEQ ID NO: 1-948 are shown in Table 2 below.




Using eMatrix software package (Stanford University, Stanford, Calif.) (Wu et al., J. Comp. Biol., Vol. 6 pp. 219-235 (1999) herein incorporated by reference), all the sequences were examined to determine whether they had identifiable signature regions. Table 3 shows the signature region found in the indicated polypeptide sequences, the description of the signature, the eMatrix p-value(s) and the position(s) of the signature within the polypeptide sequence.




Using the pFam software program (Sonnhammer et al., Nucleic Acids Res., Vol. 26(1) pp. 320-322 (1998) herein incorporated by reference) all the polypeptide sequences were examined for domains with homology to certain peptide domains. Table 4 shows the name of the domain found, the description, the p-value and the pFam score for the identified domain within the sequence.




The nucleotide sequence within the sequences that codes for signal peptide sequences and their cleavage sites can be determined from using Neural Network SignalP V1.1 program (from Center for Biological Sequence Analysis, The Technical University of Denmark). The process for identifying prokaryotic and eukaryotic signal peptides and their cleavage sites are also disclosed by Henrik Nielson, Jacob Engelbrecht, Soren Brunak, and Gunnar von Heijne in the publication “Identification of prokaryotic and eukaryotic signal peptides and prediction of their cleavage sites” Protein Engineering, Vol. 10, no. 1, pp. 1-6 (1997), incorporated herein by reference. A maximum S score and a mean S score, as described in the Nielson et as reference, was obtained for the polypeptide sequences. Table 5 shows the position of the signal peptide in each of the polypeptides and the maximum score and mean score associated with that signal peptide.















TABLE 1











HYSEQ







TISSUE




LIBRARY/




LIBRARY







ORIGIN




RNA SOURCE




NAME




SEQ ID NOS:











adult brain




GIBCO




AB3001




16-17 19 40 66 92-94 97 124 131 134 163









186 188 208 213 231 268-270 284 288 295









297 299 311 315-325 340 373 387 396 407









429 469 489 495 498-499 533 542 545 562









568 587 589 618-619 643 664 687-688 694-









695 730 748 836 876 882 884 902 925-926









948






adult brain




GIBCO




ABD003




2 22-24 29 33 43 45 50-51 66 71 75 77 82









87-88 91-92 95 131 140 157 179 188-192 200









208 213 220 225 247 252 257 261 263-265









277 284 288 295 299 301 315-325 355-356









373 387-389 392 395-396 407 423 431 443-









444 450-451 457 459 468 476 489 495 499-









500 514 520-522 532-533 541-542 545-546









557-558 562 564 576-577 581-583 588-589









591 595 597 599 601 610 619 631-632 639









643-644 654-655 658-660 664 667 676 682









687-688 693 696 700 704 711 713-714 746









758 765-766 774-775 780 800 802 804 807









810 827 829 834 842 850 854-855 866 870-









871 878 892-893 897 899 910 916 920-921









929 931-932 934






adult brain




Clontech




ABR001




12 51 87 142 169 178 180 245 263 286 288









290 295 304 308 311-313 375 379-380 403









425 428 431 458 486 499 503 512 557-558









567-568 606 610 641 651 695 704 730 741









754 766 810 822 827 841 850 864 871 884









897 917 920 925-927 934 946






adult brain




Clontech




ABR006




2 14-15 22-23 29 32-33 49 66-68 83 99 111-









112 115 129 131 142 147 153 157 163 169









189-192 200 205 207 212-214 218 221 229









234 256-257 263 272 276 279 282 292-299









301 311 315 340-343 349 376-377 383-386









388 403 405 407 410 425 438 453-454 460









463 469 474 489 495 499-500 511 522 531-









532 539 541 545-546 551 556 563 565 571









579-583 591 594 606 626 628 631-632 643









647 651 678 684 691-692 700 717 721 726









730 732 741 744 754 757 769 772 774 782









788 793 810 820 827-828 853 867 869 875









879 897 913 921-922 925-926 933-934 939-









941 947






adult brain




Clontech




ABR008




1-2 9-10 13 16-18 23 27-28 30-32 37 39 42-









43 46 49-51 66 70 76 80 83 86-87 95-97 109









111-112 116-117 124 130-131 133-134 136-









137 141-142 146-147 152-157 160 162 169









171 179 184 189-192 195 200-201 206 211-









212 216-218 239 247-248 250 252 254-258









261-263 271-272 276 278 282 288 293-295









297 300 302 307 309 311 314-326 328 333









337-341 343 347 349 351-354 358 360-361









367 374 376-378 381 384 388-390 393 395-









396 400-403 405 407 409 411 414 418-420









422 427-429 433 438 440-441 445-447 450









453-455 458 460-461 463 466-470 474 476









486 491-493 496 498-500 507 511 514 520-









521 525 527-529 531-532 534-535 542 546









548-549 551-552 557-558 560 562 564-566









568 571-572 578-583 586-587 590-591 594









599 602 606 618-619 621 626 629 631-634









643-644 647 651 656-660 664 670 672 677









680 684 687-688 691-695 697 706 709-710









712-714 716-718 721-722 724-725 727-728









730 733 740-741 745 751-752 754 761 765









774 777-779 787 790 792-793 799 801-804









808 810 812 820 822 824 827 831-832 834









836 845 850 858-861 868-869 871-672 875-









876 883 887 891 897 900 904 907 910 913









917-920 925-927 929 931-934 938-941 946-









947






adult brain




Clontech




ABR011




51 133 810 892






adult brain




BioChain




ABR012




140 208 311 748 810






adult brain




Invitrogen




ABR013




51 245 311 316-325 436 717 810 936






adult brain




Invitrogen




ABR014




2 51 65 84 86 134 311 316-325 384 422 445









460 503 525 564 634 651 721 794 804 810









922






adult brain




Invitrogen




ABR015




37 134 263 272 277 294 311 443 467 500 514









582-583 619 651 694 850 871-872 883 888









936






adult brain




Invitrogen




ABR016




19 22 57 134 188 233 271 277 299 373 440









444 459 469 514 640 717 882 890 920






adult brain




Invitrogen




ABT004




1-2 18 28 51 55-57 67 87-88 115 119 137-









139 142 163 200 204 213 218 257 263 271









282 288 299 301 311 341 358 370 378 402









407 422-423 427 458 460 463 499 504 534-









535 551 557-558 571 586 605-606 610 618









627-628 640 643 680 687 691-692 697 701-









702 715 719-721 725 727 753-754 758 771









782 810 827 859 871-872 881 913 920 925-









926 938-941 944 946






cultured




Stratagene




ADP001




2 43 51 73 76 88 97 142 166 181 186 188






preadipocytes






208 257 262-263 267-270 282 311 316-325









383 386 427-429 459 463 465 493 507 514









522 545 552 572 643 651 667 700 721 740









754 758 778 795 872 881 883 888 947






adrenal gland




Clontech




ADR002




3-6 10-11 13 16 20-21 24 27-28 33 38 48-49









51 53-54 58 66-67 75 88 97 99 124-125 130









140 157-158 179 188 197-198 200 212-214









216 218 224 229 231 237 257 267 279 281-









282 288 302 311 326 362 376-377 381 383









396 398-403 429 443 453-454 456 459-460









474 489 515 526 531-532 540 545 550 559









564 568 577 581 586 589 599 605 610 613









631-632 643 648 651 667 670 672 681 684









699 703 706 708 717-718 734 736 751 779









785-786 795 813 817 837 871 876 887-888









897 904 907 916 921 924-926 948






adult heart




GIBCO




AHR001




1-2 5-6 14-18 20-21 23 28 32 37 41 45 51









53 55-56 62 66 69-70 80-81 85 87 91 97 107









120-121 124 134 140-141 156 163 165-166









172 188-192 195 197-198 200 208 213 216









221 229 231 235 261-265 267 271 276 284









288 302 305 308 311 316-325 328 333-334









337-338 347 368-369 373 376-377 379-380









389 396 420 440 445 453-454 459-460 465









468 478 483-484 489 491-493 495 501 504









507 514 524 529 533 539 541-543 545 549









552-553 564 566 568 574 577 581-583 587









589-591 596 599 602 605 608-609 618-619









623 625 629-632 643 645 647 651 664 672









676 678 683-684 707 714 716-717 732 735









740 743-744 751 754 757 765 775 778 784-









786 788 807-808 810 826 828-829 842 850









860 876 878-880 890 894 897 899 902 916









923-927 933 939-941






adult kidney




GIBCO




AKD001




1-2 5-6 13 16-17 19-23 26 28 33 38-39 43









45 48-51 55-57 60 66-67 69-73 79 82-83 87









90 94 96-97 100 103 126 131 134 140 148-









149 157 163 166 179 184 186 188-192 200-









203 213-216 220-221 224 226-229 232 235









245 252 257 261-263 268-270 272-274 276-









277 279 282 288 290 294 299 308 311 316-









325 332 335 339-340 358 360-363 373 375









379-380 386 388-389 392 395-396 402 413









421 423-424 428-429 431 436 440 444 450









454 457 459-460 468-469 476 489 492-493









499 504 511 513-514 520-521 524-526 531









533 538-542 544-547 552 564 567-568 574









577-578 582-583 590-591 595-596 598 602









607 610 613 618-619 622 631-632 639-642









644 647 651 654-655 658-659 664 667-669









673 678 680-682 684 687 689 693 696 706-









707 712 714-715 717-718 721 729-731 734-









736 740 744 748 754 760 771 774 782 784









789 795 807 809-810 819 825 834 836-837









842 850 859 870 872 876 878-879 884 887









890 895 897-899 902 905 910 919-921 925-









926 933 936 944






adult kidney




Invitrogen




AKT002




1 14-15 28 30 35-37 53-54 73 88 112 114









129 134 137 140 149 157 166 172 186-188









191-192 203-204 213 235 245 257 262-263









266 268-270 273-274 288-289 297 299 302









310-313 315-325 335 340 358 373 378-381









395 413 423 441 450 453 456 459-460 470









477 491-494 500 513 540 542 545 554 556









564 567 587 591 619 622 627 633 643 668-









669 677 684 689 693 701-702 704 714 729-









730 754 758 760 777 781 785-786 788-789









807 836-837 840 849-850 872 876 881 890









895 905-906 913 923 925-926 931-933 944






adult lung




GIBCO




ALG001




5-6 16 28 38 51 74 97 122 124 134 140 163









188-192 200 218 221 262-263 268-272 294









311 316-325 379-380 429 463 468 493 511









520-522 537-538 542 545 568-569 595 622









643-644 664 667 711 714 721 730 754 775









850 860 863 879 887 897 925-926 944






lymph node




Clontech




ALN001




43 98 131 140 163 188 221 245 277 299 311









491 515 546 564 593 603 610 615 630 682









694 707 717 800 831 850 878 880 936 939-









941 947






young liver




GIBCO




ALV001




3-4 17 20-21 32 43 55-56 70 100 134 137









163 172 174 179 186 188-192 200 213 216









219 221 229 232 252 275 301 311 315-325









378 381 392 441 459-460 497 499-500 514









524 526 533 539 550 568 571 588-589 595









619 622 631-632 642 658-659 664 677 680









693 700 707 713 719 743 754 757-758 766









807 834 863 867 876 884 887 904 907






adult liver




Invitrogen




ALV002




5-6 28 35-36 52 54 70 72 86-87 103 112 127









134 140 159 179 188 200-201 213 218-219









225 239-240 257 263 271 275 311 315 367









373 388 392 444 459-460 464 468 497-499









512 527 532 542 545 562 599 605 629 640









657 680 684 687-688 706 713 715 717-718









721 742 754 758 771 791-793 818 829 843









854-855 871 878-879 887 921 933-934






adult liver




Clontech




ALV003




159 179 189-192 201 219 257 349 392 568









664 753 796 887 934






adult ovary




Invitrogen




AOV001




2-7 10 13 18-22 25 27-28 30-31 33 38-39









41-43 45 48 50-51 53-56 62 66-67 69 72 74-









75 80 83 85-87 93 95 99-101 107 112-115









120 124-126 129 131 134-137 140 142-143









147-148 162-163 172 178 188 191-192 200









204 208 212-213 220-221 225 229 231 235









237 246-247 252-253 258 261-262 264-265









267-272 276-279 282 287-288 290 293-294









299 307-308 311 316-327 332 337-338 340-









342 349 360-362 373-374 379-381 386 388-









389 393 396 399 403 413-414 423 425 427









429 431 441 444-445 450 452 454-455 457









459-460 462 467-470 475 477 483-484 489









491-493 495-496 500 504-505 507 515 518-









519 522-523 527-529 531 533 537-540 542









545-546 548 551-552 555 564 568 570-571









577 579 581-583 589-596 599-600 605 610









613 615-616 619 623 625-627 630-636 639









641-644 647 649 651 654-655 664 668-669









672 676 678 680-682 684 687 694 701-704









706-707 715-717 721-722 727 729 731-732









734-735 738 740 743 748-749 753-754 758









764 771 775 777-782 784 800 802 807 821-









822 824 828 834 836-837 842 846-847 850









860-863 866 870-871 876-880 882 884 887









890-891 897 899 901 906 910 913 920-921









923-924 933 939-941 944 947






adult placenta




Clontech




APL001




34 68 102 263 444 493 520-521 534-535 689









706 754 797-798






placenta




Invitrogen




APL002




2 14-15 43 55-56 66-67 134 184 213 221 229









252 257 263 277 287 394 443 529 532 618









622 684 742 754 810 829 883 902






adult spleen




GIBCO




ASP001




2 14-15 20-22 29 38 43 48 51 53-56 65 67









72 74 84 87 131-132 134 137 140 172 188-









192 200 212 221 256 263 271 282 308 311









316-325 343 383 389 423 436 441 443 459-









460 467 469 495 499-500 505 514 520-522









524 529 537 539 545 552 585 619 631-632









639 643 664 673 707 723 735 742-744 758









771 799 810 817 836 850 878 925-926 934









936






testis




GIBCO




ATS001




1 3-4 14-16 28 31 45-46 66 85 90 95 97 103









112 128-130 134 140 163 166 188 191-192









199-200 213 226-228 261-265 267-271 284









302 311 316-325 327 379-380 391 413 421









428 444 454 457 459-460 467 491 493 495









500 505 519 525 529 532 534-535 545 552









556 566 568 575 596 599 613 616-617 647









649 651 680 684 703 707 716 719 721 727









734 738 740 744 748 758 765-766 774 777









782 802 810 817 827-828 834 842 846-847









850 862-863 871-872 878 880 892 901 916-









917 921






adult bladder




Invitrogen




BLD001




5-6 8 20-21 28 72 91 122 126 130 166 188









197 200 213-214 225 257 262 315-325 341









409 486 491 572 593 622 650 673 691-692









810 813 861 870 877 883 887 904






bone marrow




Clontech




BMD001




8 13-16 28 38 43 45-48 50-51 57 62-63 65









67 84-85 97 100 104 118 122-124 131 134









140 163 188 214 216 221 224 231 245 252









261-263 268-270 273-274 279 288 290 311









373 378 389-391 395 414 428 431 436 440-









441 443 451 455 459-460 465 469-470 475









495 497-498 502 507 514-519 529 537-538









542 546 550 552 556 560-561 563-564 568









576-577 580 587 589 596 601-602 610-613









619-620 626 642-643 647 651 664 666 668









676 678 681-682 684 696 704 706-707 715









727 730 732-735 740 748 753 758 761 764









771 775 780 794 800-801 830 834 836 842









850 863 871-872 878-879 882 884 888 897









900-901 904 910 921 923 929 934 947






bone marrow




Clontech




BMD002




1-2 5-6 10 13 16-21 27 31 38 42-43 46 57









65-66 76 80 84 87 97 99 110 112 118 131









134 137 140 145 161 163 165 172 195 206









208 221 229 231 237 244 247 252 256 267-









270 272 276 278-279 282 284 288 294 301









304 307 311 316-327 333-334 337-338 345-









347 352 360-361 368 373 376-378 381 383









388 414 436 441 443 450 452 454-455 457









469-470 483-484 486 490 498 516 519-521









524 530-531 539 542-543 545-546 551 553









555 559 564 571 576-577 580 585 591 594









602 604-605 607-608 610-612 619-621 625-









626 629 631-632 639-640 644 650-651 664-









665 684 687-688 693 699 703 714 723-724









727 733 735 740 742 745 748 750-752 754-









755 777-780 784 787 794-795 802 809 817









824 827 831-832 834 846-847 850-851 854-









855 861 867 875 878 883 886 891 894 897









900 902 910 914 919 921 925-926 929 936









939-941 944






bone marrow




Clontech




BMD004




65






bone marrow




Clontech




BMD007




65 76 84 245 516






adult colon




Invitrogen




CLN001




19-21 53 55-57 72 88 133-134 168 213 245









252 311-313 316-325 340 443 459 469 483-









484 486 497 515-516 597 606 622 643 667









676 706 718 742-743 753 766 829 833 872









887 902 923 929






Mixture of 16




Various




CTL016




52 137 189-192 316-325 529 591






tissues-




Vendors*






mRNAs*






Mixture of 16




Various




CTL021




65 84 169 189-192 311 316-325 406 676 727






tissues-




Vendors*





782 850






mRNAs*






Mixture of 16




Various




CTL028




65






tissues-mRNAs*




Vendors*






adult cervix




BioChain




CVX001




3-4 14-16 20-23 25 33 42-43 45 48-50 54 57









67 69 75 85 87 91 95-97 107 110 114 124









126-127 131 134 137 140 150 157 163 165









172 185-188 200 204 212-213 216 225 229









245 252 257 261-263 266-270 276 282 288









290 301-302 308 316-325 327 340 363-364









372-373 378 383 388-392 394 396 409 413-









414 421 428-429 438-440 443-444 454 456-









457 459 463 467 475 486 489 493 495 507









514-515 522 534-537 556 568 572 574 577









582-583 587 594 600 608 610 613 622 626









633 639 643 647-648 651 653 667 680 683









685-686 693 696 703-704 706 711 721 723-









725 727 730-731 734-735 742-743 748 754-









757 762 771 776 785-786 788 794 800 802









807 809-810 817 827 829 834-835 842 850









857 860 862-863 868 870 873 876-877 879-









880 884 887 891 897 904-905 910 916-917









921 925-926 933 937 947






diaphragm




BioChain




DIA002




305 311






endothelial




Stratagene




EDT001




1-2 7-8 14-16 19-22 24 28-29 32-33 41 43






cells






45 51 57 61 74 83 87-88 97 105 112 116-117









131 134 137 140 148 165 172 179 188-192









197-198 208 212-213 220-221 225 229 231









237 246 252 256-258 261-265 268-272 276-









277 279 281-282 284 286 288 294 297 299









302 307-308 311-313 326 334-335 340 355-









356 358 360-361 364 375 383 386 389 392









403 413 423-424 429 440 443 445 451 453









455-456 459-460 462-463 465-466 468-470









475 491 495 497-499 504 514 520-522 524-









526 528 532-536 539-540 546 551-552 554









556 564 566-567 571 574-577 581-583 587









591-593 597-599 601 607 615 618 622 625









633 639 641-644 651 667 677 680 684 691-









692 701-702 704 716-717 720-721 726 732-









733 735 743-744 754 758 765 785-786 795









802 806 809 819 826 828-830 832 834 836









846-847 850 867 871 877-878 890-891 897









902 907 921 923 925-926 944 946






esophagus




BioChain




ESO002




188






fetal brain




Clontech




FBR001




33 49 51 126 134 197-198 264-265 360-361









413 460 647 810 819 871






fetal brain




Clontech




FBR004




137 156 205 282 284 405 424 480 489 701-









702 820 921






fetal brain




Clontech




FBR006




2 9-10 18-19 22 28 30-32 37 39-40 42-43









46-47 49 57 66-67 76 80 83 96 109 112 116-









117 120 124 131 133-134 136 142-143 146









152 155 160 162 165 169 173 184 189-198









200-201 205 215-216 238 244 248 254-255









257-258 260-263 272-274 276-277 282 288









293-294 307 309 311 314-328 343 347 351-









352 354 357-358 360-361 373-375 378-381









390 392 400-401 403 405 407 410-411 413









420 424 429 445 450 452-453 458 460 463









467-469 472 474 477 479 483-484 491 499









507 520-521 525 527 529 531 533 538 545









551 562 564 566 571 574 579 581-583 587









591 599-600 604 606 611 626 629 631-632









638 643 651 654-655 657-660 672-673 676-









677 684 689 693-694 697 699 709 714-715









717 720-721 732-733 735 744 748 751-752









754 761 763 767 772 775 777-779 781 785-









786 790 792 802 804 808 810 820 824 826









838-840 850 858-860 864 866 872-873 881









891-892 901-902 904 910-911 913 917-918









920 925-926 933 939-941 946-947






fetal brain




Clontech




FBRs03




316-325 684






fetal brain




Invitrogen




FBT002




2-4 20-21 45 51 53 57 88 93 125-126 134









166 184 186 188 200 213 224 263 276-278









307 311 341 373 375 418-419 423 427 432









450 452 459-460 470 492 498-499 507 514









522 534-535 545 550-552 571 577 610 714









721 743 754 795 827 861 866 872-873 887









896 925-926 934 939-941 946 948






fetal heart




Invitrogen




FHR001




2-4 10 13 16-18 29 31-32 37-38 43 46 49 51









53 55-56 67-68 75 80 85 87 97 115 120 137









152 156 160-161 163 168-169 174 178 189-









192 196 200 216 220 225 252 262 276-277









282 288 301-302 305 311 315-325 333 343









351 357-358 360-361 368-372 378 424 436









440-441 445 453 460 469 478 483-484 495









520-521 527 533 538 541-543 546 556 564-









566 568 576 581 587 594-595 601-602 606









609 612 615 633 638 640 643 653-655 664-









665 672-673 677 684 691-693 697 704 707









709 717 735 738 744 746 748-749 751-752









754 761 777 779 781-782 785-786 797-798









820 824 826 829 834 838 841-847 850 875









877-878 893-894 897 901 910 913 925-927









936 946






fetal kidney




Clontech




FKD001




8 14-15 32 43 50 68 96 106 126 131 134 140









186 188 226-228 233 279 282 311 339 428









440 450 456 468 552 618 651 700 726 735









748 751 781 794 797-798 826 878 887 899






fetal kidney




Clontech




FKD002




50 83 96 131 134 143 163 172 193-194 201









203 215 263 273-274 311 316-325 339 360-









363 374 376-377 379-380 388 394 400-401









403 407 425 440 451 454 493 525 536-538









540-542 572 580 582-583 587 605-606 621









631-632 647 673 689 706 709 714 726 735









761 774 777 799 809 845-848 858 872 875









878-879 882 895 918 927






fetal kidney




Invitrogen




FKD007




66 214






fetal lung




Clontech




FLG001




65 179 213 223 340 360-361 491 564 577 591









627 646 650 712 715 744 758 939-941






fetal lung




Invitrogen




FLG003




49 54 75 97 137 148 152 188 197-199 213-









214 225 240 256 288 316-325 369 378 392









423 429 464 496 526 580-581 586 591 693









706 726-727 766 878 913 925-926 939-941






fetal lung




Clontech




FLG004




388 921






fetal liver-




Columbia




FLS001




1-23 25-39 41-43 45-46 49-65 76 83-84 87-






spleen




University





88 91 94 97 100 112 118 122 126 129-130









134 137 140 148 163 168 172 179 186 188-









189 191-192 197-198 200-201 213-214 216









221 225-229 231-232 235 242 252-253 256-









258 262-271 277 284 287-288 297 299 307-









309 311 315-326 330 355-356 360-362 370









373 378-380 388 392 394 396 400-402 413









426 428 436 440-441 443-447 450 454-455









457 459-460 463-465 467-469 475 477-478









489-509 511-514 519-521 525-527 529-535









537 539 542 545 551-552 555-556 559 561









567 569 571 576-577 580-583 586-587 589









591-592 595 598-602 605 607 610 612-613









618-619 623 625-626 631-632 638-640 642-









644 646-647 649 651-652 654-655 667 673









676 680-681 683-684 700 703-704 706 711









715-718 720-721 726 732-735 740 742-744









748 754 756-758 763-765 771 774-775 777-









782 785-786 790 793-795 797-798 806-808









815 818 824-826 829-830 834-835 837 841









846-847 849-850 856 860-861 866 870-872









876 878 881-883 888 894 897-898 902 905









907 910 919 924-926 929 933 942 947






fetal liver-




Columbia




FLS002




1 3-4 11-12 14-17 20-23 26-29 32-34 38 41






spleen




University





43 45-47 49 51-52 55-62 65-67 76 83-85 87-









88 90-91 95 97-99 104-105 112 114-117 126









130 133 150 163 165 172 178 186-187 193-









194 200-202 208 213 221 225 229 232-235









244-246 248-253 256-257 262-265 267-271









273-274 284 287 299 311 315 326 335 337-









338 343 355-356 358 375-378 381 392 394









400-402 414 416 426 428-429 440-442 444-









447 453-455 457 459 461 464-465 467 476-









477 483-484 489-490 492 495 497-500 504









506-507 509 511-514 519 522 524 526 532-









535 537 539-540 542 545 551-552 556 567









569 574 576-577 581 589-590 592 599 601-









602 605 607 610 612-613 619-620 625 627









629 631-632 638 640-641 646 648-649 654-









655 667 670 683-684 687-688 693 696 700









703-706 713 716-721 726 734-735 740 742









744 748 754 758 771 775 777-778 780-782









785-787 790-792 794-799 801-802 806 808









818 824 829 835 849-850 852-855 857 870-









871 876 882-884 886 888 890 894 897-898









900 902-903 907 919 921 923-926 929-930









933 938-942






fetal liver-




Columbia




FLS003




30 34 67 85 88 89 130 172 188-189 191-192






spleen




University





213 229 231 257 311 315-325 329 331 335









362 391 394 400-402 423 441 455 457 461









476 498 500 511 523 531-532 537 542 576









587 592 612-613 625 649 665-666 703 719









731 733 740 744 771 775 777-778 787 797-









798 819 824 826 850 854-856 861 863 870









879 884 897 923 931-933 947






fetal liver




Invitrogen




FLV001




2 7 19 28 35-37 47 52 54-56 66 95 134 139









179 188-192 200 213 218 263 272 288 294









305 311 315 349 378-381 388 392 403 426









443 454 459-460 469 496-499 514 527 529









532 534-535 555 586 605 640 644 658-659









673 680 687 698 713 715 720-721 723 726









754 758 778 795 817-818 829 853 861 868









870-871 897 903 933






fetal liver




Clontech




FLV002




52 189-192 219 297 308 335 364 378 427 828






fetal liver




Clontech




FLV004




2 19 28-29 37 39 49 52-53 55-56 62 65-66









76 87 124 134 137 139 142 179 188 195 208









216 219 244 252 263 268-270 272 277 287-









288 294 303 305 311 315-325 339 355-356









358 360-361 368 374 378 403 441 454-455









460 477 483-484 497 514 520-521 542 553









582-583 587 591 594 611-613 620-621 638-









639 654-655 658-659 681 684 687-688 709









721 730 738 744 752 754 781 793 802 813









818 826 832 836 854-855 876 878 893 897









900 910 924 933 944






fetal muscle




Invitrogen




FMS001




28 65 115 121 126 134 137 156 168 172-173









181 213-214 225 263 267 305 340-341 360-









361 440 459 516 534-535 543 564 586 606









609-610 623 650 676 683 754 766 853 871









886 894 930 934-935 948






fetal muscle




Invitrogen




FMS002




19-21 41 49 51 53 57 75 96 101 103 112 134









136 156 171 184 188 191-192 212 216 250









262 267 276 305 311 342 348-350 355-356









360-361 374 392 403 411 415 423 425 457









469 491 495 499 508 515 517 534-536 543









546 564 566 576 580 582-583 587 594 599









609 611 615 618 623 644 647 658-659 664









668-669 677 683 691-692 696 703 735 743









754 766 788 802 817 826 828 850 877-879









894 910 925-926 935






fetal skin




Invitrogen




FSK001




3-7 18 24 27 29 35-37 51 53 55-56 66-67 76









90 95 97 122 126 134 136-137 166-167 181









188-192 208 213-214 224-225 245 250 252









257 260 262 268-271 273-274 282 284 297









302 312-313 315-326 341 367 373 375 378









383 387-388 390 394 423 429 440-441 450









454-455 457 459 463-464 470 472 475 486









489 492 495 498-500 511 514 524 527 530-









532 534-535 541-542 545 550 552 555-556









565 571 586 589 591-592 602 604 606 610









613 618 622-623 626 631-632 640 651 654-









655 672-673 685-686 693 701-702 704 706









717-718 720 723 727-728 744 754 762 764-









766 768-771 795 809 814 821 824 827 843









853 868 870-874 887-888 890 897 902 907









925-926 928 930 933-934 939-941 944-945









947






fetal skin




Invitrogen




FSK002




2 5-6 19 29 34 51 57 59 88 97 101 124 131









134 143 163 166 172 189 191-192 196 212









216 222-223 231 250 257 263 268-272 282









284 287-288 294 297 299 302 304 310-311









316-325 328 333 340 352 360-361 365-367









372 379-380 388 390 400-401 403 410-411









440 449-450 454 457 463 470 478 491 495









500 505 515 520-521 524 532 534-535 541









555 560 562 564 572 576 581 592 595 599









611 622 626 630 636 640 642 650 664 677









683 691-693 696 699 701-702 708-709 715









721 723 728 735 744 747-748 750 754 766









779 782 799 803 807 813 820 824 826 834









846-847 867-868 872-874 878-879 890-891









897 901 904 907 910 912 916 918 925-926









933 944-945






fetal spleen




BioChain




FSP001




311 748






umbilical cord




BioChain




FUC001




1-2 29 32 46 67 83 87 94 134 136 140 148









160 163 166 172 181 186-192 197-198 208









213 216 225-231 237 252 261-265 267-270









279 282 288 295 302 308 311 316-326 339-









340 365 376-377 379-380 384 392-397 421









423 428 433 440 445 452 459 461 463-464









470 472 489 491 495 497 500 507 517-518









522 525-526 528 534-535 540 545-546 556-









558 564 566 568 571-572 577 592 599 601









605 610 618 623 644 651 661 668-669 673









678 680 685-686 696 706 709 718 735-736









748 754 769 772-777 782 792 797-799 802









807 809 815 817 824 850 854-855 870 876









881 888 891 897 899 901 913 921 928 930-









932






fetal brain




GIBCO




HFB001




2 12 16-17 19 23 27-28 32-33 39 41-45 49









87-89 94 97 100 107 112 130-131 134 142









157 163 172 188-192 200 216 224-225 231









237 242 246 252 258 261 263-265 271 273-









274 276-277 288 295 299 301 307 311 314-









326 328 341 355-356 373 375 387 389 392









395 424-425 431 438 445 450-452 457 459-









460 468-469 475 489 491 495 500 504 511









514 520-529 531 533 540-542 545 552 554









557-558 566 576-577 579-584 587 591 596









598-599 606 613 626 631-632 643 651 664









668 673 676 680 693-694 696 703-704 716-









717 721 727 735 738 740 744 748 757-758









769 774 778 780-781 810 827-828 830 850









869 871-872 876 878-879 884 890 892 897









899 904 906-907 913 916 918 920 924 928









934 938 946






macrophage




Invitrogen




HMP001




49 97 208 252 301 306 311 316-325 337-338









345-346 416 512 522 572 670 716 743 785-









786 802 888 919 923






infant brain




Columbia




IB2002




2-4 12 14-15 20-21 23-24 27 29 31-32 39 41







University





46 48-49 51 53 55-56 66 75-76 86-88 93 95









101 105 108-109 116-117 125 127 129 131









136 145 163 166 170 180-181 186 188-189









191-192 200-201 207-208 212-214 216 220









224 229 231 245 247 252 257 259 264-265









267 271 279 282 288 293 295 299-300 311









314-326 337-338 340 349 367 373 375 388









390-393 396 402 405 407 418-421 424 428-









429 431 433 436 450 452-453 457 459 463









468 489 495 496-500 507 511 522-524 526









528-530 532 541-542 545-546 552 557-558









562 564-566 571 577-583 587 589-591 599-









601 606 608 613-614 619 631-632 647 654-









655 658-659 667 676 684 691-693 696 700









704 711 718 721 723 725 740-741 743-744









748 754 775 777-778 780 788 792-793 795









802 805 808 819 826-829 834 836 838 861









863 869-870 875 879 881-882 884 887 890-









891 893 897 902 920-921 925-926 934 938-









941 946






infant brain




Columbia




IB2003




2 27 37 39 43 48-49 51 53 85-87 97 106 113







University





124 126-127 131 142 166 170 188 200-201









208 214-215 220 224 226-228 231 251 257









263 267 271-272 279 288 293 299 311 314-









326 337-339 349 360-361 367 386 392 397









400-402 407 410 418-419 424-425 427 429









452 454 460 475 489 495 497-500 507 522-









523 525 529 532 539 542 545-546 551-552









557-558 564-565 578 582-583 585 591 601









606 625-626 631-633 643-644 673 690-693









701-702 706 711 721 723 734 740-741 743-









744 748 751 754 761 778 788 795 802 808









819 826-827 829 837 843 869-871 875 878-









880 884 896-897 902 920 933-934 946






infant brain




Columbia




IBM002




32 43 66 340 387 541 562 693 712 751 795







University





829 871 920 925-927 929






infant brain




Columbia




IBS001




2 29 37 39 76 142 163 392 455 495 499 606







University





681 741 754 778






lung,




Stratagene




LFB001




2-4 22 28 32-33 47 51 79 120 129 134 140






fibroblast






163 172 188 208 220-221 231 252 257 263









276-277 284 307 375 378-380 396 423 428









440 450 459 463 486 491 493 495 499 539









571 591 601 607 613 615 618 625 639 651









684 716-717 721 727 735 748 782 828 850









870-871






lung tumor




Invitrogen




LGT002




2 5-8 13 16-17 29-31 35-39 43 46 57 67 72









76 78 81 85 87 90 94 97 100 110 119 122









130-131 134 137 140 146 149 167 172 174









179 188 197-198 201 213 216 218 220-221









223 231 245-246 251-252 256-257 262-263









267-270 277 284 288 296 299 301-302 311









316-325 340 354 373 379-380 388 392 395









400-401 410 413 421 431 436 441-443 445









451 455 457 460 463-464 467 469 475 478









489 491 493 497 499 504 507 514 518-519









524 529 534-535 537 542 545-546 548 552









555 559 568 578 581-583 592 597 602-603









605-607 613 615 619 621-622 636-637 642-









643 646-647 654-655 679-681 684 687-689









693 701-702 704 706 711 713 715-716 718









727 732-734 738 748 753-754 757-758 760









762 766 769 774 782 785-786 802 817 829









834 850 853 859-860 866-867 870-871 878-









879 887 890 899 902 904 910 917 923 925-









926 936-937






lymphocytes




ATCC




LPC001




2 16 19-21 25 31-32 49 53 55-56 63 67 85-









87 90 97 120 122 137 140 163 165 168 172









188 197-198 215-216 221 229 231 236-238









248 252 256 272-274 283-284 288 294 299









316-326 343 368 374 378 395 423 431 454-









455 467 469 476 478 491 495 498 505 512









515 517-518 520-522 524 526 529 531-532









537 539 542 545-546 551 556 571 577 580









589-590 592 596 601 622 631-632 640 642









654-655 664 666 668-669 673 684 703 708









716 718 721 723 727 733 735 743-744 754









758 765 771 775 777-779 783-786 797-799









810 816-817 828 834 845 859 861 863 870









878-879 881 884 887 890 897 904 907 910









912-913 918 923 929 939-942 945-947






leukocyte




GIBCO




LUC001




2-4 7-8 13-17 20-23 31-33 38 43 48-49 51









53-57 63 66-68 74-78 85-88 93 97 122 124









129 131-132 134 137 140 163 166 168 171-









172 175 188-192 197 200 208-213 216 221









223 231 236 242 252 257-258 261-263 268-









270 272 277 279 287-288 294 307 311 314









316-326 329 337-339 341 373-374 376-377









381 388-392 396 400-401 413-414 423 436









441 450 454-455 459 463 465 467 489 491-









493 495 498-499 504-505 507 514 518 520-









522 524 526 529 531 533 536-537 539-540









545 552-554 556 568 571 577 580 585 589-









590 596 599 602 605 607 610 612-613 615









618-619 621-622 625 638 640 642-644 664









667 677-678 684 690-693 696 700 703-704









707 713 715-718 721 727 734-735 738 740-









746 748 753-754 758 775 778 780 789 794









797-798 801-802 815 817 825 827 829 834









836 846-847 850 859-861 863-864 866-867









871-872 878 884 886-888 891 896-897 902









904 910 913 916 921 923-926 929-932 936









943






leukocyte




Clontech




LUC003




12 14-15 18 32 111 134 137 172 221 277 280









311 316-325 436 454 467 549 552 568 585









603 643 691-692 698-699 734 744 751 754









784 797-798 861 897 916 923






Melanoma from




Clontech




MEL004




3-4 16 20-21 43 46 48 97 103 147 163 188






cell line ATCC






191 213 216 221 231 241 245-246 260 262-






CRL #1424






263 316-325 381 407 431 504 525 527 542









556 568 577 589 596 607 613 676 693 714









735 737-739 744 758-760 775 822 850 863









878 887 897






mammary gland




Invitrogen




MMG001




1-8 14-18 20-21 25 28-29 37 39 43 49 51-57









60 66-67 72 75-76 87 95 97 103-104 106 112









115 119 122 127 130 134 137 139 142 150









166-168 172 175 184 186 188-189 191-192









200 213-214 222-224 226-229 240 252 257-









259 263 267 271 276 278 282 287-288 299









301-302 305 307-308 311-313 316-325 327-









328 332 340-341 358 360-362 369 373 378









381 383 388 390-392 397 402-403 409 415-









416 423 425 428-429 433 436 444 454-456









459-460 464 467 469 481 483-484 486 493









495 498-499 515 524-525 529-530 532-537









541-542 545 551-552 562 582-583 586-587









593 599-600 602 604-605 610 618-619 622









625-627 634 644 646-647 652 654-655 662









673 676 680 684 687-688 691-692 701-703









715 717 721 723 726 735 743 751 754 758









765-766 771 777-778 789 803 805 807 809-









811 821 827 829 850 860 887-888 892 896









898 901-902 905 911 913 917 925-926 930









936 939-942






induced neuron




Stratagene




NTD001




2 16 32 51 66 88 97 124 130 134 137 172






cells






188-189 191-192 231 252 257 260 277 291









373 424 431 454 460 489 495 523 525 582-









583 591 631-632 643 649 670 695 725-726









735 765 789 797-798 837 850 878 884 888









890 913 929 946






retinoid acid




Stratagene




NTR001




2 5-6 20-22 136-137 188-194 197-198 224






induced






311 375 381 410 457 462 475 495 531 546






neuronal cells






548 552 599 618 678 743 752 819 828 890









895 897 930 934 938 944 946






neuronal cells




Stratagene




NTU001




2 5-6 20-21 55-56 87 137 188-192 197-198









215-216 260 287 291 310-311 316-325 365









375 423 457 459 470 499 532 542 564 576









598-599 623 643 651-652 673 721 726 743









745 752 754 765 780 787 789 822 829 870









875 888 896 917 919 929






pituitary gland




Clontech




PIT004




41-42 83 85 97 134 193-194 204 208 213 224









257-258 263-265 285-286 308 311 360-361









413 443 445 491 514 529 532 639 644 647









682 701-702 716 781 822 829 836 850 933









939-941 947






placenta




Clontech




PLA003




16 31 34 49 66 80 87 97 101-102 134 158









165 172 179 184 188 197-198 209-210 218









220 229 235 249 256 267-270 277 287-288









302 307 332 360-361 365 388 394 414 441









444 454 457 460 493 498-500 505-506 509









529 531-532 550 559-560 564 572 587 601









625 630-632 638 672 682-684 689 706 708









726 733 735 744 754 761 784-786 793 863









875 897 924 929 937






prostate




Clontech




PRT001




7-8 51 85 87 97 100 122 134 139 214 216









221 231 257 271 276 335 337-338 392 400-









401 431 440 459 477 530 534-535 546 556









582-583 599 622 631-632 639 651 663-664









673 683 707 715 735 740 765 773-774 777









810 823 897 909 919 934 939-941 947






rectum




Invitrogen




REC001




18 54 66 134 137 169 188 200 213 225 251









263 288 311-313 316-325 340 388 423 429









441 454 459 514 532 542 610 626 646 651









657 715 719 723 728 735 740 758 766 785-









786 823 829 833 836 886 942






salivary gland




Clontech




SAL001




31 49 78 95 134 136-137 143 176 188 208









223 244 268-270 284 308 311 316-325 388-









389 391 436 441 459 476 514-515 520-521









532 543 568 589 596 610 619 684 691-692









713 718 727 736 754 777 824 836 864 867









878 883 897 901-902 916-917 933 938-941






salivary gland




Clontech




SALs03




460






skin fibroblast




ATCC




SFB001




379-380 850






skin fibroblast




ATCC




SFB002




742 850






skin fibroblast




ATCC




SFB003




87






small intestine




Clontech




SIN001




27-29 31 38 40 46 48 51 54 57-58 62 65 67









75 77 85 97 110 112 116-117 119 131-132









134 137 140 161 163 166 168 177 188 197-









198 208 213 220 224 229 246 257 261-262









264-265 276-277 288 295 297 299 311 316-









326 328-330 337-338 340 360-361 373 375









382 390-391 410 413 428-429 436 438 440









453-454 459 468 476-477 497 507 511 522









531 536 538 542 545-546 548 552 556 564









570-571 576 580-581 586-587 591 596 599









605 610 613 619 625-626 643-644 651-653









664-666 668-670 677 680 684 693 700-702









706-707 713-715 723-724 729-730 735 740









746 748 753-754 757-758 764 777-778 784-









786 818 822 824 826-829 833-837 842 862-









863 865-867 877-878 886 897 900-902 906









913 916 921 925-926 936 939-941






skeletal muscle




Clontech




SKM001




42 98 156 163 191-192 200 261 305 311 395









415 462 468 504 531 543 566 582-583 585









594 680 740 853 875 927 933 935






skeletal muscle




Clontech




SKM002




850






spinal cord




Clontech




SPC001




18 23 33 37 42 51 67 87 92 94 97 100 140









162 184 188 191-192 208 213 220 231 248









262 268-271 273-274 282 287-288 290 307









311 316-325 358 364 376-377 383 387 389-









390 402 412 422 444 455 476 483-484 489









504 522 534-535 556 562 587-588 591 597









603-604 618-619 643 651 667-670 677 693









703-704 717-718 727 746 757 773 808 810









827 834-835 837 850 871-872 875 904 910









931-932 939-941






adult spleen




Clontech




SPLc01




33 38 57 67 75 87 134 142 163 216 221 229









244 257 304 307 311 316-325 340 355-356









378 441 468 525 538 545 560 564 599 721









754 766 780 794 827 841 850 866






stomach




Clontech




STO001




18 65 88 163 188 208 213 261 272 277 286









294 336 373 396 412 459 514 553 602 610









647 651-652 671 673 714 774 790 831 833









842 850 876






thalamus




Clontech




THA002




2 87 96 103 106 189-192 208 252 258 295









308 311 367 376-377 383-384 445 455 459-









460 498 529 587 598 602 629 654-655 705-









706 715 717 723 754 775 810 817 822 864









867 881 892 927 930






thymus




Clontech




THM001




3-4 8 18 28 54 57 63 65 68 84 97 100 116-









117 122 134 142 151 169 171-172 188 195









197-198 201 213 221 237 245 261 287 311









316-325 360-361 376-377 423 441 444 459









489 491-493 495 498 504 507 514 527 532









534-536 539 553 556 568 571-572 590 595-









596 599 610 618 622 631-632 643 647 651









654-655 664 687-688 691-693 703 715 721









733-735 748 760 762 765 781 794 799 802









831 834 836 842 850 860-861 863 871 878









885 896-897 903 910 923 925-926 928 939-









941






thymus




Clontech




THMc02




2-4 17 20-22 37-38 42-43 46 63 65-68 76 88









95 103 118 120 124 134 137 140-141 143 163









165 171 179 182 189-194 198 200 212-215









221 226-228 231 244 257 262 266 276-277









287-288 297 299 307 316-325 341 352 358









360-361 373 376-377 379-381 389 391 394-









396 403 410-411 436 440 445 450 459 463-









464 469 478 491 495 500 507 511 519-521









530 532 539 542 550 555 560 563 576 581









587 595 601 610-611 613-614 618 622 625-









626 631-632 638 642-644 657 664 667 670









673 680 683 687 691-693 699 715-716 721









740 743-744 747-754 761 763-765 771 777









780-781 784-787 790 794 805 811 820 826









831 834 841 845 861 867-868 878 881 883









891 893-894 896-897 902 904 910 912-914









918 923 936-941 946-947






thyroid gland




Clontech




THR001




1-2 18-21 27 32 38 42 46 49 51 53-56 66 72









77-78 87-88 97 115 119 124 130-131 134 136









152 163-165 172 183 188-192 202 212-213









216 221 224 229 235 241-243 252 257-258









261 263-265 267 277 279 297 301 305 308









311 316-325 327 357 363 373 376-377 381









383 389 397 400-401 410 413-414 427-428









443-444 446-447 457 459 463 467-469 475









482 489 495 499-500 504 509 513 519-522









526 529 533 537-538 542 545-546 548 556









564 567-568 582-583 589 592 599 605 608









611 621 623 630 642-644 648 651-652 654-









655 664 672-676 684-686 691-694 700 706-









708 713 717-718 721-722 725 729 731 734-









735 740 748 753-754 760 764 766 771 774









777 781 792 797-800 802 805 826 828-829









834 842 850 861 863 868 876 879 897 899









901 910 913 929 937 939-941






trachea




Clontech




TRC001




20-21 38 112 161 163 188 263 267 327 413









420 457 459-460 471 514 540-541 552 572









574 622 639 654-655 676-677 691-692 707









725 743 748 765 777-778 862 868 897 905









908 944






uterus




Clontech




UTR001




51 67 126 130 133 140 188-192 229 267 329









373 440 491 514 599 685-686 693 713 716-









717 735 897 905 911 939-941











*The 16 tissue-mRNAs and their vendor source, are as follows: 1) Normal adult brain mRNA (Invitrogen), 2) normal adult kidney mRNA (Invitrogen), 3) normal adult liver mRNA (Invitrogen), 4) normal fetal brain mRNA (Invitrogen), 5) normal fetal kidney mRNA (Invitrogen), 6) normal fetal liver mRNA (Invitrogen), 7) normal fetal skin mRNA (Invitrogen), 8) human adrenal gland mRNA (Clontech), 9) human bone marrow mRNA (Clontech), 10)








# human leukemia lymphablastic mRNA (Clontech), 11) human thymus mRNA (Clontech), 12) human lymph node mRNA (Clontech), 13) human spinal cord mRNA (Clontech), 14) human thyroid mRNA (Clontech), 15) human esophagus mRNA (BioChain), 16) human conceptional umbilical cord mRNA (BioChain).























TABLE 2












SMITH-







SEQ




ACCESSION





WATERMAN




%






ID NO:




NUMBER




DESCRIPTION




SCORE




IDENTITY



























1




Z99162






Schizosaccharomyces pombe


putative transporter




134




30






2




U44839






Homo sapiens


UHX1 protein




3719




100






3




AF031939






Mus musculus


RalBP1-associated EH domain protein Reps1




3687




94






4




AF031939






Mus musculus


RalBP1-associated EH domain protein Reps1




1887




57






5




U69490






Mus musculus


p56lck-associated adapter protein Lad




173




37






6




U69490






Mus musculus


p56lck-associated adapter protein Lad




173




37






7




AL161538






Arabidopsis thaliana


disease resistance N like protein




398




33






8




W88660




Secreted protein encoded by gene 127 clone HSUBW09.




175




97






9




D50617






Saccharomyces cerevisiae


YFL025C




306




34






10




AB028070






Homo sapiens


activator of S phase Kinase




193




42






11




Y27676




Human secreted protein encoded by gene No. 110.




474




100






12




Y30721




Amino acid sequence of a human secreted protein.




355




98






13




AF257330






Homo sapiens


COBW-like protein




566




97






14




AF089812






Mus musculus


ubiquitin-conjugating enzyme HR6A




378




98






15




AF089812






Mus musculus


ubiquitin-conjugating enzyme HR6A




688




90






16




Y94959




Human secreted protein clone mc300_1 protein sequence SEO ID NO: 124.




204




97






17




AF212247






Homo sapiens


CDA08




2664




92






18




AF064868






Rattus norvegicus


brain-enriched guanylate kinase-associated




452




45








protein 1; BEGA1






19




W67863




Human secreted protein encoded by gene 57 clone HFEBF41.




551




98






20




AL132954






Arabidopsis thaliana


putative protein




201




25






21




AL132954






Arabidopsis thaliana


putative protein




361




34






22




Y48359




Human prostate cancer-associated protein 56.




403




98






23




AF202892






Mus musculus


Kif21a




5679




92






24




W75143




Human secreted protein encoded by gene 23 clone HBMCT32.




148




100






25




Y35921




Extended human secreted protein sequence, SEQ ID NO. 170.




548




99






26




Y27587




Human secreted protein encoded by gene No. 21.




448




100






27




AF190900






Homo sapiens


kelch-like protein C3IP1




1767




74






28




X12517






Homo sapiens


C protein (AA 1-159)




903




99






29




AY014403






Homo sapiens


kinesin-like protein RBKIN1




9290




99






30




AF240783






Mus musculus


ELKL motif kinase 2 short form




194




48






31




Y02661




Human secreted protein encoded by gene 12 clone HFTCU19.




858




81






32




AF271070






Homo sapiens


amino acid transporter system A1




2466




100






33




W75151




Human secreted protein encoded by gene 34 clone HTEGA81.




507




100






34




AF206329






Mus musculus


polydom protein




3886




81






35




AF162224






Mus musculus


angiopoietin-related protein 3




138




23






36




Y35996




Extended human secreted protein sequence, SEQ ID NO. 361.




503




97






37




AF022891






Drosophila melanogaster


Fuzzy




248




30






38




X76775






Homo sapiens


HLA-DMA




1091




100






39




Y16045






Arabidopsis thaliana


leucine-rich repeat protein




160




33






40




AC003077






Homo sapiens


60% similar to AB002297 (PID:g2224539)




2629




61






41




W74887




Human secreted protein encoded by gene 160 clone HCELB21.




203




100






42




AC005390






Homo sapiens


R31180_1




1550




66






43




M98450






Oryctolagus cuniculus


casein kinase-II beta




668




100






44




Y10840




Amino acid sequence of a human secreted protein.




349




100






45




AL050231






Drosophila melanogaster


alternatively spliced form




203




46






46




U58280






Mus musculus


second largest subunit of RNA polymerase I




4956




89






47




AJ224819






Homo sapiens


tumor suppressor




457




42






48




AF205935






Mus musculus


MGA protein




3477




77






49




AF095352






Homo sapiens


RAB-like protein 2B




718




99






50




L20321






Homo sapiens


protein serine/threonine kinase




394




41






51




Y13374






Homo sapiens


putative prenylated protein




397




87






52




AJ302031






Rattus norvegicus


putative alpha 1B-glycoprotein




519




40






53




AF145681






Drosophila melanogaster


BcDNA.LD23181




828




47






54




AJ132192






Mus musculus


HS1 binding protein 3




1454




75






55




AF145681






Drosophila melanogaster


BcDNA.LD23181




692




53






56




AB036800






Drosophila melanogaster


egg-derived tyrosine phosphatase




828




47






57




Y87327




Human signal peptide containing protein HSPP-104 SEQ ID NO: 104.




584




100






58




Y36237




Human secreted protein encoded by gene 14.




177




100






59




Y87310




Human signal peptide containing protein HSPP-87 SEQ ID NO: 87.




370




100






60




AF062476






Mus musculus


retinoic acid-responsive protein; STRA6




1437




74






61




Y38394




Human secreted protein encoded by gene No. 9.




213




100






62




AL139421






Homo sapiens


dJ717I23.1




3267




100








(novel protein similar to


Xenopus laevis


Sojo protein)






63




AL359782






Trypanosoma brucei


possible (hhv-6) u1102, variant a dna,




142




50








complete virion genome.






64




Y19561




Amino acid sequence of a human secreted protein.




514




100






65




L12690






Homo sapiens


neutrophil peptide-1




493




100






66




AF274057






Rattus norvegicus


GRIP-asaociated protein 1 long form




3814




92






67




AL162458






Homo sapiens


bA465L10.2




6467




99








(novel C2H2 type zinc finger protein similar to chicken FZF-1)






68




Y87100




Human secreted protein sequence SEQ ID NO: 139.




267




100






69




Y86320




Human secreted protein HPRBC80, SEQ ID NO: 235.




361




100






70




Z99162






Schizosaccharomyces pombe


putative transporter




169




27






71




S67057






Cricetulus migratorius


= Armenian hamsters, Peptide,




158




71








223 aa serum amyloid P, SAP, female protein, FP = pentraxin






72




Y36183




Human secreted protein #55.




449




96






73




U08813






Oryctolagus cuniculus


597 aa protein related to Na/glucose cotransporters




1231




85






74




AB009883






Nicotiana tabacum


KED




202




23






75




AX015323






Homo sapiens


hFATP1




3367




99






76




AB013361






Homo sapiens


DPM2




153




100






77




Y25732




Human secreted protein encoded from gene 22.




212




100






78




M37033






Homo sapiens


CD53 glycoprotein




116




95






79




Y36098




Extended human secreted protein sequence, SEQ ID NO. 483.




193




100






80




AB021644






Homo sapiens


gonadotropin inducible transcription repressor-4




886




60






81




U68267






Mus musculus


myosin binding protein H




1202




66






82




Y36332




Human secreted protein encoded by gene 109.




268




100






83




AF254956






Homo sapiens


candidate tumor suppressor protein




2030




99






84




L12690






Homo sapiens


neutrophil peptide-1




493




100






85




AF295378






Homo sapiens


MAGEF1




754




50






86




AF144627






Mus musculus


SLIT1




283




32






87




AF208536






Homo sapiens


nucleotide binding protein; NBP




1372




100






88




Y59657




Secreted protein 108-003-5-0-A8-FL.




689




100






89




Y27626




Human secreted protein encoded by gene No. 60.




352




100






90




AF035268






Homo sapiens


phosphatidylserine-specific phospholipase A1




498




41






91




Y12861




Human 5′ EST secreted protein SEQ ID NO: 451.




448




100






92




Y53049




Human secreted protein clone cj378_3 protein sequence SEQ ID NO: 104.




463




100






93




Y41354




Human secreted protein encoded by gene 47 clone HUFCJ30.




288




100






94




W74777




Human secreted protein encoded by gene 48 clone HFCAI74.




245




100






95




AB040610






Homo sapiens


glycoprotein beta-Gal 3′-sulfotransferase




388




47






96




D32215






Danio rerio


emx2 homeoprotein




1242




92






97




Y94959




Human secreted protein clone mc300_1 protein sequence SEQ ID NO: 124.




204




97






98




AF024496






Caenorhabditis elegans


contains similarity to


Plasmodium falciparum






390




30








glycophorin-binding protein homolog 2 (GB:X69769)






99




A09779






Homo sapiens


interferon-gamma receptor segment binding interferon-gamma




600




99






100




AE002030






Deinococcus radiodurans


thermoresistant gluconokinase




246




52






101




AB052620






Mus musculus


DDM36




4361




89






102




M69245






Homo sapiens


pregnancy-specific beta-1-glycoprotein




613




70






103




X92841






Homo sapiens


MHC class I chain-related protein A




588




100






104




AF136401






Rattus norvegicus


TRP2




410




88






105




AL031709






Homo sapiens


c316G12.4




561




93








(novel protein similar to API1 and API2








(apoptosis inhibitor 1 and 2 (MIHB, MIHC, IAP1, IAP2)))






106




Y38389




Human secreted protein encoded by gene No. 4.




152




90






107




Y27582




Human secreted protein encoded by gene No. 16.




320




100






108




AF130079






Homo sapiens


PRO2852




231




60






109




J02818






Oryctolagus cuniculus


cytochrome P-450p-2




893




46






110




Y07894




Human secreted protein fragment encoded from gene 43.




169




48






111




AF119297






Homo sapiens


neuroendocrine-specific protein-like protein 1




240




97






112




X56203






Plasmodium falciparum


liver stage antigen




254




23






113




Y76200




Human secreted protein encoded by gene 77.




262




100






114




Y36270




Human secreted protein encoded by gene 47.




359




100






115




AJ133120






Rattus norvegicus


Proline rich synapse associated protein 2




3938




93






116




AL031709






Homo sapiens


c316G12.4




299




84








(novel protein similar to API1 and API2








(apoptosis inhibitor 1 and 2 (MIHB, MIHC, IAP1, IAP2)))






117




AL031709






Homo sapiens


c316G12.4




582




100








(novel protein similar to API1 and AP12








(apoptosis inhibitor 1 and 2 (MIHB, MIHC, IAP1, IAP2)))






118




AB018542






Homo sapiens


CD98 light chain




1829




86






119




AB012692






Homo sapiens


CAC-1




132




41






120




AB021644






Homo sapiens


gonadotropin inducible transcription repressor-4




2050




57






121




AF110776






Homo sapiens


adrenal gland protein AD-003




542




53






122




AL391688






Homo sapiens


bA524D16A.2.1




683




42








(novel protein similar to mouse granuphilin-a)






123




AB042624






Homo sapiens


SIRP-b2




247




50






124




AF019980






Dictyostelium discoideum


ZipA




280




22






125




U58917






Homo sapiens


IL-17 receptor




266




30






126




AK021852






Homo sapiens


unnamed protein product




884




99






127




AY007378






Homo sapiens


G-protein beta subunit-like protein




1731




99






129




AF195522






Trypanosoma cruzi


B-cell mitogen precursor




345




39






130




Z81587






Caenorhabditis elegans


contains similarity to Pfam domain: PF01363




276




42








(FYVE zinc finger), Score = 65.2, E-value = 1.9e-17, N = 1






131




AF188700






Homo sapiens


actin filament associated protein




844




43






132




M77678






Mus musculus


NKR-P1 gene-40 protein




118




26






133




AF217226






Homo sapiens


zinc finger protein ZNF286




574




94






134




L06505






Homo sapiens


ribosomal protein L12




841




99






135




Y12902




Human 5′ EST secreted protein SEQ ID NO: 492.




134




100






136




L11672






Homo sapiens


zinc finger protein




3059




60






137




U47924






Homo sapiens


B-cell receptor associated protein




925




100






138




AC006271






Homo sapiens


BC319430_7




163




49






139




AE000799






Methanothermobacter thermoautotrophicus


O-linked GlcNAc transferase




151




37






141




Y13037




Human secreted protein encoded by 5′ EST SEQ ID NO: 51.




187




100






142




AJ3271735






Homo sapiens


sprouty (Drosophila) homolog 3




170




32






143




AF228917






Rattus norvegicus


small rec




454




56






144




U41552






Caenorhabditis elegans


Contains similarity to Pfam domain: PF00122




127




50








(E1-E2_ATPase), Score = 36.1, E-value = 8.1e-10, N = 4






145




AL049569






Homo sapiens


dJ37C10.3 (novel ATPase)




328




45






146




AF109888






Macaca mulatta


sodium-calcium exchanger circular exon 2 transcript




147




32






147




AF217227






Homo sapiens


zinc finger protein ZNF287




1146




43






148




B24426




Human PR01286 protein sequence SEQ ID NO: 199.




466




100






149




AB019120






Rattus norvegicus


seven transmembrane receptor




1015




37






150




AF062476






Mus musculus


retinoic acid-responsive protein; STRA6




190




72






151




X64223






Mus musculus


Fc-E receptor II (Fc-ERII/CD23)




138




34






152




X83543






Homo sapiens


APXL




412




41






153




AF217289






Homo sapiens


cadherin 20




4170




99






154




Y36310




Human secreted protein encoded by gene 87.




250




100






155




D50577






Mesocricetus auratus


carboxylesterase precursor




441




50






156




AB027004






Homo sapiens


protein phosphatase




229




41






157




AF305071






Mus musculus


calsenilin-like protein




1281




99






158




Y13126




Human secreted protein encoded by 5′ EST SEQ ID NO: 140.




160




96






159




AB016215






Cyprinus carpio


complement C3-Q2




132




35






160




AF294278






Homo sapiens


PR-domain-containing protein 16




6646




99






161




AF305210






Homo sapiens


concentrative Na+− nucleoside cotransporter hCNT3




3609




100






163




U10281






Sus scrofa


gastric mucin




165




22






164




M13095




Rattus sp. 0-44 protein




661




98






165




AY008763






Homo sapiens


sentrin/SUMO-specific protease




537




53






166




S72304




Mus ap. LMW G-protein




763




94






167




AF238315






Homo sapiens


HZFw1 protein




2251




99






168




M22414






Homo sapiens


ribonuclease inhibitor precursor




222




30






169




AL035702






Homo sapiens


dJ593C16.1




3010




58








(ras GTPase activating protein)






170




AL110500






Caenorhabditis elegans


Y87G2A.13




146




22






171




AB028860






Mus musculus


mDj10




189




37






172




M83653






Homo sapiens


cytoplasmic phosphotyrosyl protein phosphatase




648




100






173




AB041601






Mus musculus


unnamed protein product




255




61






174




M35012






Drosophila melanogaster


non-muscle myosin heavy chain




200




24






175




D88577






Mus musculus


Kupffer cell receptor




904




46






176




Y30847




Human secreted protein encoded from gene 37.




239




100






177




Y01390




Secreted protein encoded by gene 8 clone HTXDJ88.




301




100






178




M23725






Homo sapiens


M2-type pyruvate kinase




152




78






179




X72875






Homo sapiens


complement factor B




3527




100






180




Y08420






Homo sapiens


nicotinic acetylcholine receptor alpha7 subunit precursor




1794




100






181




M24766






Homo sapiens


alpha-2 type IV collagen




3756




99






183




AL110500






Caenorhabditis elegans


Y87G2A.13




180




25






184




AF286598






Homo sapiens


angiostatin binding protein 1




1447




62






185




Y13084




Human secreted protein encoded by 5′ EST SEQ ID NO: 98.




130




62






186




AF151067






Homo sapiens


HSPC233




892




58






187




AL133283






Homo sapiens


bA31M2.1




1629




100








(novel protein similar to the GLI family of zinc finger proteins)






188




J03799






Homo sapiens


laminin-binding protein




1331




94






189




M27132






Homo sapiens


ATP synthase beta subunit precursor




734




98






190




M27132






Homo sapiens


ATP synthase beta subunit precursor




206




100






191




M27132






Homo sapiens


ATP synthase beta subunit precursor




709




98






192




M27132






Homo sapiens


ATP synthase beta subunit precursor




1127




89






193




U76638






Homo sapiens


BRCA1-associated RING domain protein




4101




100






194




U76638






Homo sapiens


BRCA1-associated RING domain protein




394




100






195




S57688






Thermotoga maritima


EF-G




289




75






196




U09453






Cricetulus griseus


UDP-N-acetylglucosamine:




239




85








dolichyl phosphate N-acetylglucosamine 1-phosphate transferase






197




D86821






Streptomyces coelicolor


PrfB




151




40






198




Y36068




Extended human secreted protein sequence, SEQ ID NO. 453.




495




94






199




U38980






Homo sapiens


hPMSR6




408




92






200




Y08999






Homo sapiens


Sop2p-like protein




572




99






201




AJ401272






Canis familiaris


Band4.1-like5 protein




2233




95






202




U09608






Homo sapiens


cell surface protein




292




84






203




Y64747




Human 5′ EST related polypeptide SEQ ID NO: 908.




471




96






204




Y66754




Membrane-bound protein PR01187.




656




100






205




U52111






Homo sapiens


Ca2+/Calmodulin-dependent protein kinase I




1680




99






206




AF279265






Homo sapiens


putative anion transporter 1




186




39






207




X91911






Homo sapiens


rtvp-1




446




40






208




X56390






Canis familiaris


rac2




508




100






209




AK026888






Homo sapiens


unnamed protein product




1536




100






210




AK026888






Homo sapiens


unnamed protein product




927




98






211




AB044805






Homo sapiens


6-phosphofructo-2-kinase heart isoform




2452




100






212




AB013897






Homo sapiens


HKR1




3083




99






213




U37351






Mus musculus


Paneth cell enhanced expression PCEE




746




89






214




AJ278475






Homo sapiens


transport-secretion protein 2.1 (TTS-2.1)




2179




98






215




U58749






Caenorhabditis elegans


coded for by


C. elegans


cDNA yk8c7.5;




844




63








coded for by


C. elegans


cDNA yk47c5.5;








coded for by


C. elegans


cDNA yk76b5.5;








coded for by


C. elegans


CDNA yk8c7.3;








coded for by


C. elegans


CDNA yk47c5.3;








strong similarity to catalytic domains of ser/thr protein kinases






216




AF000195






Caenorhabditis elegans


Contains similarity to Pfam domain:




1072




47








PF00169 (PH), Score = 20.6, E-value = 1.9e-05, N = 1






217




AB016768






Mus musculus


thrombospondin type 1 domain




189




40






218




AB026256






Homo sapiens


organic anion transporter OATP-B




1928




99






219




X56692






Homo sapiens


C-reactive protein




327




100






220




AF064801






Homo sapiens


multiple membrane spanning receptor TRC8




603




29






221




AF191545






Homo sapiens


aminopeptidase




5048




99






222




X66171






Homo sapiens


CMRF-35 antigen




228




33






223




Z38061






Saccharomyces cerevisiae


mal5, stal, len: 1367, CAI: 0.3,




241




22








AMYH_YEAST P08640 GLUCOAMYLASE S1 (EC 3.2.1.3)






224




AC005306






Homo sapiens


R27216_1




1560




100






225




AF176532






Mus musculus


F-box protein FBX17




1059




81






226




AF097432






Homo sapiens


GROS1-L protein




2147




99






227




AF097432






Homo sapiens


GROS1-L protein




1900




96






226




AF097432






Homo sapiens


GROS1-L protein




3881




99






229




Z29094






Caenorhabditis elegans


contains similarity to Pfam domain:




388




32








PF01699 (Sodium/calcium exchanger protein),








Score = 268.7, E-value = 2.5e-77, N = 2






230




Y66669




Membrane-bound protein PRO839.




366




100






231




X75931






Bos taurus


Cleavage and Polyadenylation




4033




98








specificity factor (CPSF) 100 kD subunit






232




AF152562






Homo sapiens


angiopoietin-related protein 3




1210




99






233




L19686






Homo sapiens


macrophage migration inhibitory factor




564




94






234




AL008723






Homo sapiens


dj90G24.4 (SAAT1




3408




100








(low affinity sodium glucose cotransporter








(sodium:solute symporter family)))






235




U50927






Rattus norvegicus


zinc transporter ZnT-2




1040




85






236




AF099973






Mus musculus


schlafen2




982




53






237




U67557






Methanococcus jannaschii


cell division control protein 48 (cdc48),




1050




41








AAA family






238




AF207661






Homo sapiens


sodium bicarbonate cotransporter-like protein




5645




100






239




AF284337






Homo sapiens


SEBOX




1209




100






240




AF081669




synthetic construct VU91B calmodulin




109




42






241




U96166






Streptococcus cristatus


srpA




286




18






242




AE005024




Halobacterium sp. NRC-1 Vng0821c




130




37






243




AK023335






Homo sapiens


unnamed protein product




2344




99






244




AK024464






Homo sapiens


FLJ00057 protein




3033




99






245




U55376






Caenorhabditis elegans


coded for by


C. elegans


CDNA cm21e6;




752




40








coded for by


C. elegans


CDNA cm01e2;








similar to melibiose carrier protein (thiomethylgalactoeide permease II)






246




AK022660






Homo sapiens


unnamed protein product




1173




99






247




D16235






Bos taurus


PLC alpha




156




24






248




B08894




Human secreted protein sequence encoded by gene 4 SEQ ID NO: 51.




235




67






249




AJ289709




HERV-H/env62 envelope protein




754




39






250




AJ242540






Volvox carteri f. nagariensis


hydroxyproline-rich glycoprotein DZ-HRGP




209




63






251




Y66747




Membrane-bound protein PRO1158.




609




100






252




AL031532






Schizosaccharomyces pombe


ubiquitin conjugating enzyme




236




41






253




W74899




Human secreted protein encoded by gene 172 clone HODCW06.




197




100






254




D43633






Oryzias latipes


G protein-coupled seven-transmembrane receptor




462




36






255




D43633






Oryzias latipes


G protein-coupled seven-transmembrane receptor




823




42






256




AJ277750






Homo sapiens


UBASH3A protein




1035




44






257




Y12711






Homo sapiens


putative progesterone binding protein




187




42






258




W74939




Human secreted protein encoded by gene 49 clone HAGBI17.




211




100






259




D16432






Mus musculus


murine homologue of CD63/ME491




504




99






260




AB031051






Homo sapiens


organic anion transporter OATP-E




863




37






261




J03998






Plasmodium falciparum


glutamic acid-rich protein




139




29






262




U80953






Caenorhabditis elegans


weakly similar in serine repeat region to rat




336




32








thyroxine-binding globulin (PIR:A39567)








and to


D. melanogaster


ecdysone-inducible protein E75-C








(SP:E75C_DROME, P13055)






263




AF155662






Homo sapiens


putative 16.7 kDa protein




766




99






264




Z46237






Saccharomyces cerevisiae


putative protein




245




27






265




Z46237






Saccharomyces cerevisiae


putative protein




245




27






266




AF208795






Ictalurus punctatus


NCC receptor protein 1




286




50






267




AJ249901






Mus musculus


secreted modular calcium-binding protein 2




2332




95






268




X05908






Homo sapiens


lipocortin (AA 1-346)




967




100






269




X05908






Homo sapiens


lipocortin (AA 1-346)




1511




100






270




X05908






Homo sapiens


lipocortin (AA 1-346)




967




100






271




AC003038






Homo sapiens


R30923_1




2992




100






273




U37143






Homo sapiens


cytochrome P450 monooxygenase CYP2J2




942




44






274




U37143






Homo sapiens


cytochrome P450 monooxygenase CYP2J2




555




45






275




AF154933






Sus scrofa


complement component C3




374




39






276




Z54342






Caenorhabditis elegans


contains similarity to Pfam domain:




570




39








PF00328 (Histidine acid pliosphatase), Score = 511.8,








E-value = 2e-152, N = 1






277




M20259






Homo sapiens


thymosin beta-10




169




97






278




Y91386




Human secreted protein sequence encoded by gene 41 SEQ ID NO: 107.




558




100






279




AF151110






Mus musculus


COP1 protein




2268




99






280




AF117959






Homo sapiens


CDK4-binding protein p34SEI1




114




68






281




Y91370




Human secreted protein sequence encoded by gene 25 SEQ ID NO: 91.




293




100






282




AF079446






Dictyostelium discoideum


developmental protein DG1067




520




38






283




M60618






Homo sapiens


nuclear autoantigen




300




84






284




D83777






Homo sapiens


expressed ubiquitously with strong expression in brain




912




51






285




L77864






Homo sapiens


stat-like protein




290




96






286




Y02775




Human secreted protein encoded by gene 12 clone HFTCU19.




288




98






287




L21998






Homo sapiens


mucin




389




24






288




AF184275






Mus musculus


F-box protein FBX18




4409




92






289




AF053356






Homo sapiens


leucin rich neuronal protein




264




37






290




AC016829






Arabidopsis thaliana


putative O-linked GlcNAC transferase




377




27






291




Y73414




Human secreted protein clone yb101_1 protein sequence SEQ ID NO: 50.




472




100






292




W88615




Secreted protein encoded by gene 82 clone HNGBT31.




525




97






293




AJ278018






Homo sapiens


calsyntenin-2




5080




100






294




Y41460




Fragment of human secreted protein encoded by gene 42.




1034




100






295




D79995






Homo sapiens


similar to pig tubulin-tyrosine ligase.




415




42






296




AF092878






Homo sapiens


zinc RING finger protein SAG




323




92






297




AL137784






Homo sapiens


dJ199J3.1




561




100








(novel protein similar to ubiquitin carboxyl - terminal hydrolase 16








(EC 3.1.2.15))






298




Z70310






Caenorhabditis elegans


contains similarity to Pfam domain:




520




37








PF00013 (KH domain), Score = 42.8, E-value = 3 .7e-12, N = 1;








PF00023 (Ank repeat), Score = 428.2, E-value = 2.4e-125, N = 19






299




Y48600




Human breast tumour-associated protein 61.




288




98






300




AB007889






Homo sapiens


KIAA0429




386




46






301




U64601






Caenorhabditis elegans


Gene probably begins in the next cosmid




412




50






302




U27109






Homo sapiens


prepromultimerin




208




24






304




AF278532






Homo sapiens


beta-netrin




3347




99






305




X51957






Homo sapiens


muscle-specific enolase




555




94






306




Y07895




Human secreted protein fragment encoded from gene 44.




537




100






307




AE001045






Archaeoglobus fulgidus


proliferating-cell nucleolar antigen P120, putative




144




41






308




AB033882






Coturnix japonica


protein kinase C inhibitor




433




60






309




AC006284






Arabidopsis thaliana


putative ankyrin




198




44






310




AF093673






Cricetulus griseus


layilin




576




45






311




J02642






Homo sapiens


glyceraldehyde 3-phosphate dehydrogenase (EC 1.2.1.12)




1685




97






312




AB012692






Homo sapiens


CAC-1




560




94






313




AB012692






Homo sapiens


CAC-1




484




73






314




Y17571






Homo sapiens


aralar2




300




48






315




X15324






Homo sapiens


angiotensinogen




1925




97






316




X97321






Homo sapiens


HLA-C protein




1099




98






317




M24036






Homo sapiens


MHC HLA-B8 chain




1033




92






318




U04245






Homo sapiens


MHC class I antigen




940




95






319




M32318






Homo sapiens


HLA protein allele B57




982




88






320




U41057






Homo sapiens


HLA class I A locus antigen A*68new




1801




93






321




M32321






Homo sapiens


HLA protein allele A25




1163




96






322




AJ250917






Homo sapiens


human leucocyte antigen B




315




88






323




D64147






Homo sapiens


HLA-Cw*0602




1121




96






324




AB005048






Homo sapiens


A26null allele




977




85






325




M24043






Homo sapiens


MHC HLA-A1 chain




1123




97






326




AF190900






Homo sapiens


kelch-like protein C3IP1




2982




100






327




AB022023






Bos taurus


nonmuscle myosin heavy chain B




158




22






328




X85019






Homo sapiens


UDP-GalNAC:polypeptide N-acetylgalactosaminyl




1431




50








transferase






329




W58985






Homo sapiens


adult brain clone BV141_2 encoded protein.




201




100






330




Y14455




Human secreted protein encoded by gene 45 clone HCFBJ91.




284




100






331




M25757






Bos taurus


GTP:AMP phosphotransferase (EC 2.7.4.10)




127




73






332




AF037402






Bos taurus


butyrophilin




225




25






333




X99211






Drosophila melanogaster


ubiquitin-specific protease




846




68






334




Y36160




Human secreted protein #32.




500




100






335




W67869




Human secreted protein encoded by gene 63 clone HHGDB72.




454




91






336




X15334






Homo sapiens


creatine kinase B




162




71






337




AL109658






Homo sapiens


dJ776F14.1 (ortholog of mouse P47)




310




50






338




AL109658






Homo sapiens


dJ776F14.1 (ortholog of mouse P47)




797




55






339




AJ001309






Homo sapiens


DnaJ protein




659




100






340




AF096870






Homo sapiens


estrogen-responsive B box protein




465




28






341




L33243






Homo sapiens


polycystic kidney disease 1 protein




20117




99






342




Z83850






Homo sapiens


mouse NIK serine threonine protein kinase like; match:




1582




95








proteins P97820 CE02384






343




AF201084






Secale cereale


secalin precursor




149




29






344




L16685






Caenorhabditis elegans


homology with breakpoint cluster region protein;




549




41








putative






345




AL162458






Homo sapiens


bA465L10.4




2489




94








(matrix metalloproteinase 9 (gelatinase B, 92 kD gelatinase,








92 kD type IV collagenase) (CLG4B))






346




AX011001






Homo sapiens


MMP-9




2375




100






347




X89416






Homo sapiens


protein phosphatase 5




451




87






348




Y64786




Human 5′ EST related polypeptide SEQ ID NO: 947.




200




100






349




AC005167






Arabidopsis thaliana


putative ubiquitin-conjugating enzyme




509




44






350




AF074901






Caenorhabditis elegans


hemicentin precursor




1205




29






351




U12623






Rattus norvegicus


cyclic nucleotide gated cation channel




2811




93






352




AF245516






Drosophila melanogaster


Ran binding protein 9




140




40






353




AF300649






Homo sapiens


regulator of G-protein signaling




926




100






354




X16396






Homo sapiens


precursor polypeptide (AA −29 to 315)




855




80






355




S70290






Homo sapiens


glutamine synthetase, GS {EC 6.3.1.2}




1787




100






356




X59834






Homo sapiens


glutamate-ammonia ligase




435




89






357




X84801






Homo sapiens


ZNF165




467




55






358




AL365234






Arabidopsis thaliana


putative protein




224




34






359




AJ272034






Homo sapiens


putative capacitative calcium channel




4470




100






360




AJ007798






Homo sapiens


stromal antigen 3, (STAG3)




278




74






361




AJ007798






Homo sapiens


stromal antigen 3, (STAG3)




292




82






362




AF193807






Homo sapiens


Rh type B glycoprotein




2176




99






363




AC006963






Homo sapiens


similar to Kelch proteins; similar to BAA77027




895




37








(PID:g4650844)






364




Y86297




Human secreted protein HLDCE79, SEQ ID NO: 212.




530




100






365




AB039903






Homo sapiens


interferon-responsive finger protein 1 long form




4302




99






366




AF143003






Perca flavescens


lysyl oxidase related protein homolog




410




55






367




AF151840






Homo sapiens


CGI-82 protein




1166




71






368




AB036834






Drosophila melanogaster


MAP kinase phosphate




188




41






369




M30262






Homo sapiens


preprocardiodilatin-atrial natriuretic factor




771




100






370




AF135253






Mus musculus


fibulin-2




386




41






371




AF112361






Schmidtea mediterranea


opsin




109




32






372




X89416






Homo sapiens


protein phosphatase 5




387




100






373




M96256






Homo sapiens


rapamycin binding protein




1169




100






374




AF121859






Homo sapiens


sorting nexin 9




660




43






375




Y07566






Homo sapiens


RIT (Ric-related gene expressed in many tissues)




266




28






376




AF208291






Homo sapiens


protein kinase HIPK2




6242




99






377




AF170301






Mus musculus


nuclear body associated kinase 1a




5972




97






378




AL133230






Homo sapiens


dJ530I15.2




1170




99








(novel protein similar to placental protein DIFF40)






379




AB001928






Homo sapiens


cathepsin V




656




98






380




AB001928






Homo sapiens


cathepsin V




725




90






381




Y87329




Human signal peptide containing protein HSPP-106 SEQ ID NO: 106.




692




94






382




AL356276






Homo sapiens


bA367J7.2.1




479




51








(novel Immunoglobulin domains containing protein (isoform 1))






383




AK024551






Homo sapiens


unnamed protein product




824




100






384




U70851






Caenorhabditis elegans


similar to


S. cerevisiae


protein transport protein




612




39








SEC7 (SP:P11075)






385




D83348






Rattus norvegicus


long type PB-cadherin




3904




92






386




AF162149






Mycoplasma bovis


variable surface lipoprotein




216




46






387




U96149






Mus musculus


perforatorial protein PERF 15




481




68






388




U12535






Homo sapiens


epidermal growth factor receptor kinase substrate




1051




46






389




U00059






Saccharomyces cerevisiae


Yhr116wp




114




41






390




U70369






Mus musculus


hematopoietic-specific IL-2 deubiquitinating enzyme




873




47






391




AF247679






Xenopus laevis


putative N-terminal acetyltransferase




978




57






392




AB007646






Arabidopsis thaliana


UVB-resistance protein UVR8




315




31






393




AF026246






Homo sapiens


HERV-E envelope glycoprotein




361




91






394




AF198489






Homo sapiens


LBP-32




1747




57






395




Y91356




Human secreted protein sequence encoded by gene 11 SEQ ID NO: 77.




801




100






396




Y94978




Human secreted protein clone pw337_6 2nd protein sequence




444




100








SEQ ID NO: 238.






397




AC005996






Homo sapiens


similar to


Xenopus laevis


gamma-crystallin 6;




818




100








similar to AF071563 (PID:g3930581)






398




Y76216




Human secreted protein encoded by gene 93.




225




97






399




Y11447




Human 5′ EST secreted protein SEQ ID No 269.




210




97






400




U12392






Haematobia irritans


putative ATPase




917




49






401




U12392






Haematobia irritans


putative ATPase




609




49






402




AF002109






Arabidopsis thaliana


putative ABC transporter




487




40






403




AF110645






Homo sapiens


candidate tumor suppressor p33 ING1 homolog




860




69






404




J03941






Mus musculus


ferritin heavy chain




947




100






405




AL023513






Homo sapiens


dJ268D13.1.1




808




46








(seizure related gene 6 (mouse)-like (KIAA0927)








(isoform 1))






407




D87458






Homo sapiens


Similar to Human estrogen-responsive finger protein,




2575




99








efp (A49656)






408




AJ278475






Homo sapiens


transport-secretion protein 2.1 (TTS-2.1)




346




98






409




AF282886






Homo sapiens


heparanase-like protein HPA2b




2785




100






410




AF015454






Xenopus laevis


ER1




777




42






411




AF163762






Homo sapiens


zinc metalloendopeptidase




5940




99






412




Z34277






Homo sapiens


mucin




978




95






413




Y76179




Human secreted protein encoded by gene 56.




634




100






414




AF168362






Rattus norvegicus


protein associating with small stress protein PASS1




232




68






415




AF156777






Homo sapiens


ASB-1 protein




486




39






416




AK024169






Homo sapiens


unnamed protein product




752




100






417




Y08625




Human secreted protein BL341 4.




367




100






418




AB026803






Mus musculus


synaptotagmin VI




2153




96






419




AB026803






Mus musculus


synaptotagmin VI




2098




94






420




AJ278474






Sus scrofa


cytochrome P450




1316




50






421




W78135




Human secreted protein encoded by gene 10 clone HPNGQ80.




385




100






422




U70476






Rattus norvegicus


cationic amino acid transporter-1




646




62






423




AF188634






Drosophila melanogaster


F protein




546




55






424




AJ132889






Mus musculus


kinesin like protein 9




3618




88






425




D86983






Homo sapiens


similar to


D. melanogaster


peroxidasin (U11052)




328




32






426




AL035464






Homo sapiens


dJ1043E3.1 (novel protein)




945




99






427




ALC34380






Homo sapiens


dJ50O24.4




1131




67








(novel protein with DHHC zinc finger domain)






428




AJ400877






Homo sapiens


CEGP1 protein




5605




100






429




AF060570






Mus musculus


rig-1 protein




1545




73






430




Y02697




Human secreted protein encoded by gene 48 clone HTNBR95.




165




100






431




Y73386




HTRM clone 3279329 protein sequence.




529




100






432




AF279890






Homo sapiens


2P domain potassium channel TREK2




2760




100






433




AB023658






Rattus norvegicus


Ca/calmodulin-dependent protein




2468




93








kinase kinase alpha, CaM-kiriase kinase alpha






434




J05056






Oryctolagus cuniculus


oxysterol-binding protein




181




42






435




Y99437




Human PRO1508 (UNQ761) amino acid sequence SEQ ID NO: 336.




672




83






436




AF043179






Homo sapiens


T cell receptor beta chain




681




73






437




AF151042






Homo sapiens


HSPC208




585




79






438




Y87328




Human signal peptide containing protein HSPP-105 SEQ ID NO: 105.




681




100






439




Y66734




Membrane-bound protein PRO1097.




297




70






440




D78572






Mus musculus


membrane glycoprotein




262




28






441




L20315






Mus musculus


MPS1 protein




2716




77






442




Y19588




Amino acid sequence of a human secreted protein.




329




100






443




AF306550






Sinorhizobium meliloti


(p)ppGpp synthetase




213




36






444




M38379






Arabidopsis thaliana


calmodulin-1




108




37






445




Y12512




Human 5′ EST secreted protein SEQ ID NO: 543.




320




98






446




AC013482






Arabidopsis thaliana


T26F17.15




309




37






447




AC013482






Arabidopsis thaliana


T26F17.15




289




35






448




AL035703






Homo sapiens


dJ61A9.1 (tyrosine kinase)




5326




99






449




X59720






Saccharomyces cerevisiae


YCR017C, len: 953




731




30






450




AL035526






Arabidopsis thaliana


extensin-like protein




154




33






451




U16282






Homo sapiens


ELL




147




29






452




M80537






Drosophila melanogaster


fat protein




207




28






453




AB029334






Halocynthia roretzi


HrPET-1




687




38






454




AL031349






Schizosaccharomyces pombe


putative vesicular transport protein




643




28






455




AB041533






Homo sapiens


sperm antigen




3737




98






456




U27109






Homo sapiens


prepromultimerin




208




24






457




AJ010949






Mus musculus


calcium channel alpha-2-delta-C subunit




225




22






458




AJ271643






Homo sapiens


putative acid-sensing ion channel




2893




100






459




G02479




Human secreted protein, SEQ ID NO: 6560.




442




100






460




D16111






Homo sapiens


human homologue of rat phosphatidylethanolamine




846




98








binding protein






461




AF180470






Mus musculus


Kiaa0575




1139




57






462




Z54270






Caenorhabditis elegans


F11C1.4




183




33






463




AB019527






Homo sapiens


LDOC1 protein




129




31






464




U58105






Mus musculus


Murine homolog of human ftp-3




303




36






465




AF172449






Homo sapiens


opioid growth factor receptor




325




47






466




AL035106






Homo sapiens


dJ998C11.1 (continues in Em:AL445192 as bA269H4.1)




2438




58






467




AC004561






Arabidopsis thaliana


putative proline-rich protein




256




29






468




AF000198






Caenorhabditis elegans


weak similarity to HSP90




144




28






469




X59720






Saccharomyces cerevisiae


YCR016W, len: 290




128




26






470




AF027219






Homo sapiens


ZNF202 beta




620




32






471




AF122924






Xenopus laevis


Wnt inhibitory factor-1




484




44






472




X67704






Drosophila melanogaster


sperm protein




111




32






473




AF317889






Homo sapiens


NOX5




3015




99






474




L46815






Mus musculus


DNA binding protein Rc




6027




77






475




AF190665






Mus musculus


LMBR1 long form




2391




96






476




AF041382






Drosophila melanogaster


microtubule binding protein D-CLIP-190




244




33






477




AL033545






Arabidopsis thaliana


putative protein




138




49






478




Z98745






Homo sapiens


dJ29K1.2




1123




56






479




AL118502






Homo sapiens


bA371L19.1 (novel protein)




2471




100






480




Z94160






Homo sapiens


dJ63G5.3 (putative Leucine rich protein)




953




100






481




X98263






Homo sapiens


M-phase phosphoprotein 6




469




93






482




B08904




Human secreted protein sequence encoded by gene 14 SEQ ID NO: 61.




239




100






483




AY007233






Homo sapiens


phosphoinositol 3-phosphate binding protein-1




3830




99






484




AY007233






Homo sapiens


phosphoinositol 3-phosphate binding protein-1




4152




99






485




Y94947




Human secreted protein clone cw1292_8 protein sequence SEQ ID NO: 100.




408




100






486




J03614






Oryctolagus cuniculus


myosin heavy chain




225




26






487




AB026256






Homo sapiens


organic anion transporter OATP-B




188




92






488




AJ277748






Rattus norvegicus


NTPDase6




501




96






489




U02289






Caenorhabditis elegans


GTPase-activating protein




551




36






490




B08894




Human secreted protein sequence encoded by gene 4 SEQ ID NO: 51.




211




69






491




AF238862






Xenopus laevis


Churchill protein




495




71






492




D87076






Homo sapiens


similar to human bromodomain protein BR140 (JC2069)




107




30






493




A11959




synthetic construct PP4X




1122




99






494




Y87252




Human signal peptide containing protein HSPP-29 SEQ ID NO: 29.




391




100






495




AF015565






Dictyostelium discoideum


VacA




245




31






496




AF305427






Mus musculus


cAMP-dependent protein kinase regulatory subunit




940




78






497




K02581






Homo sapiens


thymidine kinase (EC 2.7.1.21)




1216




99






498




AB048364






Mus musculus


osteoblast differentiation promoting factor




1017




46






499




AJ006239






Homo sapiens


dihydropteridine reductase




933




100






500




Y59795




Human normal ovarian tissue derived protein 72.




554




100






501




AL109965






Homo sapiens


dJ1121G12.1.2




537




54








(A novel protein containing a putative PHD finger domain, isoform 2)






502




Y12021




Human 5′ EST secreted protein SEQ ID NO: 334.




265




97






503




G01878




Human secreted protein, SEQ ID NO: 5959.




348




92






504




AL023777






Schizosaccharomyces pombe


putative RNA-binding protein




318




39






505




M11902






Mus musculus


proline-rich salivary protein




109




45






506




J01998




AKV murine leukemia virus gag-pol polyprotein




621




42








(tag amber codon at 2250-2252 inserts Gln in Mo-MuLV)






507




AC012396






Arabidopsis thaliana


tumor-related protein, putative




154




45






508




X06160






Homo sapiens


precursor polypeptide (AA −24 to 140)




742




97






509




Y99351




Human PRO1481 (UNQ750) amino acid sequence SEQ ID NO: 41.




1725




100






510




M22760






Homo sapiens


cytochrome c oxidase subunit Va




461




97






511




AF199008






Mus musculus


PALS1




3396




96






512




AF145021






Mus musculus


exportin 4




3814




98






513




AF119837






Cyprinus carpio


hexokinase I




2020




75






514




Z69043






Homo sapiens


translocon-associated protein delta subunit precursor




459




98






515




AF276512






Homo sapiens


RNA polymerase II elongation factor ELL3




2071




99






516




M34379






Homo sapiens


elastase/medullasin precursor (EC 3.4.21.37)




1320




96






517




Y11385




Human 5′ EST secreted protein SEQ ID No 207.




220




100






518




AF045022






Bos taurus


phosphatidic acid-preferring phospholipase A1




4254




91






519




U00031






Caenorhabditis elegans


Contains similarity to Pfam domain: PF00957




980




47








(synaptobrevin), Score = 100.3, E-value = 1.2e-26, N = 1






520




X92098






Homo sapiens


transmembrane protein




597




89






521




X92098






Homo sapiens


transmembrane protein




1003




96






522




X57346






Homo sapiens


HS1




1218




95






523




AF207901






Xenopus laevis


cingulin




317




24






524




AL035678






Arabidopsis thaliana


putative protein




246




36






525




Y30721




Amino acid sequence of a human secreted protein.




231




100






526




AL035659






Homo sapiens


dJ979N1.1 (dJ979N1.1)




1822




47






527




AL035548






Schizosaccharomyces pombe


putative ribose methyltransferase




263




38






528




AB015617






Homo sapiens


ELKS




3464




72






529




L25665






Homo sapiens


GTP-binding protein




2227




99






530




Y11794




Human 5′ EST secreted protein SEQ ID No: 394.




375




100






531




Y14685






Arabidopsis thaliana


polynucleotide phosphorylase




1375




40






532




X52574






Mus musculus


GTP binding protein




4571




91






533




AB040119






Homo sapiens


mitochondrial import receptor Tom22




718




100






534




U23822






Danio rerio


collagen II A1




295




43






535




U39621






Gallus gallus


type V collagen




371




46






536




X79682






Felis catus


neuronal protein




617




96






537




AF099935






Homo sapiens


MDC-3.13 isoform 2




532




55






538




AF145672






Drosophila melanogaster


BcDNA.GH12174




230




36






539




AL035709






Arabidopsis thaliana


putative protein




572




37






540




AL022104






Schizosaccharcmyces pombe


putative pre-mrna splicing factor rna helicase




431




31






541




AF113615






Homo sapiens


FH1/FH2 domain-containing protein FHOS




1138




55






542




U85494






Zea mays


LON1 protease




1797




47






543




X54162






Homo sapiens


64 Kd autoantigen




587




38






544




AB037158






Homo sapiens


DSCR6a




165




40






545




Y07754




Human secreted protein fragment encoded from gene 11.




1723




100






546




AL009171






Drosophila melanogaster


62D9.o




184




33






547




M18963






Homo sapiens


islet regenerating protein




817




89






548




U29463






Homo sapiens


cytochrome b561




1125




91






549




Z25420






Gallus gallus


class II INCENP protein




237




26






550




AL031324






Schizosaccharomyces pombe


membrane atpase




1150




41






551




AF145609






Drosophila melanogaster


BcDNA.GH02833




666




41






552




AF302079






Homo sapiens


HSP22-like protein interacting protein 17




459




100






553




AJ242730






Homo sapiens


polyhomeotic 2




451




59






554




U88167






Caenorhabditis elegans


contains similarity to C2 domains




428




45






555




AF156779






Homo sapiens


ASB-4 protein




513




33






556




AF305081






Homo sapiens


tankyrase-related protein




5992




98






557




AB028968






Homo sapiens


KIAA1045 protein




2090




99






558




AB028968






Homo sapiens


KIAA1045 protein




1448




99






559




AJ250998






Mucor circinelloides


carotenoid regulatory protein




381




40






560




AL109804






Homo sapiens


dJ1009E24.1.1




6334




99








(A novel protein similar to the mouse sialoadhesin, a macrophage sialic acid








binding receptor, isoform 1)






561




Y76539




Human ovarian tumor EST fragment encoded protein 35.




261




98






562




L07765






Homo sapiens


carboxylesterase




209




51






563




Z38061






Saccharomyces cerevisiae


mal5, stal, len: 1367, CAI: 0.3,




206




21








AMYH_YEAST P08640 GLUCOAMYLASE S1 (EC 3.2.1.3)






564




AY009133






Homo sapiens


FYVE-finger-containing Rab5 effector protein Rabenosyn-5




4027




99






565




AF287478






Lytechinus variegatus


embryonic blastocoelar extracellular matrix




1116




35








protein precursor






566




AF223427






Xenopus laevis


RRM-containing protein SEB-4




793




88






567




AP000693






Homo sapiens


partial CDS




470




56






568




AJ131244






Homo sapiens


Sec24A protein




5357




98






569




Y12049




Human 5′ EST secreted protein SEQ ID NO: 362.




253




92






570




AF255342






Homo sapiens


putative pheromone receptor V1RL1 long form




1859




99






571




Y99355




Human PRO1295 (UNQ664) amino acid sequence SEQ ID NO: 54.




1265




100






572




ACC02397






Mus musculus


C9




678




51






573




M28821






Mus musculus


Tcte-1 peptide




1884




74






574




AF000196






Caenorhabditis elegans


strong similarity to the SNF2/RAD54 family of




185




36








helicases; partial CDS






575




L34587






Homo sapiens


RNA polymerase II elongation factor SIII, p15 subunit




426




100






576




AL035419






Homo sapiens


dJ1100H13.4 (putative RhoGAP domain containing protein)




212




35






577




D83004






Homo sapiens


ubiquitin-conjugating enzyme E2 UbcH-ben




385




100






578




Y87271




Human signal peptide containing protein HSPP-48 SEQ ID NO: 48.




474




86






579




U96626






Mus musculus


chondroadherin




341




29






580




Z85986






Homo sapiens


dJ108K11.3




1468




99








(similar to yeast suppressor protein SRP40)






581




AF061529






Mus musculus


rjs




230




32






582




AF228738






Homo sapiens


profilin IIa




750




100






583




AF228738






Homo sapiens


profilin IIa




624




87






584




AF181640






Drosophila melanogaster


BcDNA.GH09817




190




47






585




AB037901






Homo sapiens


gene amplified in squamous cell carcinoma-1




1340




54






586




AF149285






Caenorhabditis elegans


Osm-3




806




62






587




AF056021






Xenopus laevis


p80 katanin




146




31






588




U80931






Caenorhabditis elegans


strong similarity to class-III of pyridoxal-




866




49








phoshate-dependent aminotransferases






589




AK026962






Homo sapiens


unnamed protein product




1725




99






590




AB011483






Arabidopsis thaliana






288




28






591




AF152243






Mus musculus


putative E1-E2 ATPase




3942




98






592




AJ243460




Leishmania major proteophosphoglycan




204




25






593




AX021519






Homo sapiens


unnamed protein product




1317




99






594




AL121581






Homo sapiens


dJ1022E24.4




1162




69








(A novel protein weakly similar to protein-L-isoaspartate








o-methyltransferase (EC 2.1.1.77))






595




AL031177






Homo sapiens


dJ889M15.3 (novel protein)




177




44






596




AL035601






Arabidopsis thaliana


putative protein




301




28






597




AF202118






Homo sapiens


HOX D1 protein




1726




100






598




AC007228






Homo sapiens


BC37295_1




955




42






599




Z69727






Schizosaccharomyces pombe


probable ribosomal protein




249




34






600




L08134






Rattus norvegicus


glycoprotein




278




24






601




AK025598






Homo sapiens


unnamed protein product




3738




99






602




X89453






Rattus norvegicus


DRPLA




173




27






603




AF137030






Homo sapiens


transmembrane protein 2




1285




57






604




AL356014






Arabidopsis thaliana


putative protein




436




44






605




G03490




Human secreted protein, SEQ ID NO: 7571.




450




96






606




AY007380






Homo sapiens


F-box protein FBX30




1211




100






607




D38076






Homo sapiens


Ran-BP1 (Ran-binding protein 1)




916




100






608




U27837






Diphyllobothrium dendriticum


actin




1033




50






609




M25750






Oryctolagus cuniculus


sarcolumenin precursor




2307




97






610




AF180920






Homo sapiens


cyclin L ania-6a




630




51






611




AF168990






Homo sapiens


putative GTP-binding protein




2742




99






612




U77942






Homo sapiens


syntaxin 7




421




98






613




U00046






Caenorhabditis elegans


similar to yeast heat shock protein STI1




362




27






614




AB001928






Homo sapiens


cathepsin V




226




100






615




AB016687






Arabidopsis thaliana


SMC-like protein




326




22






616




AL163279






Homo sapiens


homolog to cAMP response element binding and




181




53








beta transducin family proteins






617




AF176524






Mus musculus


F-box protein FBL10




525




50






618




AF070656






Homo sapiens


HSPC002




731




86






619




Y00339






Homo sapiens


carbonic anhydrase II (AA 1-260)




910




88






620




M29913






Homo sapiens


eosinophil peroxidase




3757




100






621




AC006768






Caenorhabditis elegans


contains similarity to


Mycoplasma genitalium






1216




54








glycerol-3-phospate dehydrogenase (SW: P47285)






622




AB033595






Mus musculus


gasdermin




369




31






623




AF132449






Mus musculus


smoothelin small isoform S1




646




54






624




AF247039




porcine adenovirus 3 163R*




141




31






625




L43065






Saccharomyces cerevisiae


suppresses the respiratory deficiency of a yeast




403




36








pet mutant






626




AF071172






Homo sapiens


HERC2




483




35






627




Y40090




Peptide sequence derived from a human secreted protein.




921




98






628




AP001306






Arabidopsis thaliana


contains similarity to cell wall-plasma membrane




717




36








linker protein-gene_id: MKA23.5






629




AE004543






Pseudomonas aeruginosa


probable MFS transporter




207




32






630




AK025204






Homo sapiens


unnamed protein product




1925




99






631




AL050321






Homo sapiens


dJ717M23.1 (novel gene)




2443




98






632




AL050321






Homo sapiens


dJ717M23.1 (novel gene)




3988




99






634




U87804






Caulobacter crescentus


GidA




396




65






635




AF004161






Oryctolagus cuniculus


peroxisomal Ca-dependent solute carrier




154




52






636




U78090






Rattus norvegicus


potassium channel regulator 1




682




91






637




AC002394






Homo sapiens


Gene product with similarity to dynein beta subunit




536




41






638




AL109640






Homo sapiens


dJ543J19.5 (CGI-107 Protein)




478




98






639




AB017065






Arabidopsis thaliana


contains similarity to small nuclear




125




34








ribonucleoprotein-gene id: MFC16.18






640




AK024498






Homo sapiens


FLJ00106 protein




927




99






641




AF187064






Homo sapiens


p75NTR-associated cell death executor; NADE




303




57






642




AB017507






Homo sapiens


Apg12




724




100






643




AF035632






Rattus norvegicus


syntaxin 12




1231




90






644




U48852






Cricetulus griseus


HT protein




974




57






645




AB015798






Homo sapiens


DnaJ homolog




1572




93






646




AB031046






Homo sapiens


HMG-box transcription factor TCF-3




3096




100






647




U78547






Chlamydomonas reinhardtii


PF20




209




38






648




AK024452






Homo sapiens


FLJ00044 protein




2019




63






649




Y13117




Human secreted protein encoded by 5′ EST SEQ ID NO: 131.




268




100






650




AB023419






Mus musculus


mSox7




1803




87






651




AF176688






Rattus norvegicus


sodium/calcium/potassium exchanger NCKX1




184




34






652




AK026622






Homo sapiens


unnamed protein product




4385




99






653




AFC16712






Mus musculus


testicular condensing enzyme




191




36






654




AF135440






Mus musculus


huntington yeast partner C




4306




94






655




AF135440






Mus musculus


huntington yeast partner C




3709




86






656




AK022184






Homo sapiens


unnamed protein product




680




100






657




U02467






Lilium longiflorum


meiotin-1




182




33






658




M17099






Oryctolagus cuniculus


progesterone-induced protein




1832




93






659




AF113132






Homo sapiens


phosphoserine aminotransferase




1673




100






660




AF294790






Mus musculus


RING-finger protein MURF




137




27






661




X56203






Plasmodium falciparum


liver stage antigen




157




22






662




Y64994




Human 5′ EST related polypeptide SEQ ID NO: 1155.




372




100






663




U82626






Rattus norvegicus


basement membrane-associated chondroitin proteoglycan




146




22








Bamacan






664




AF155739






Mus musculus


axotrophin




3007




85






665




AB037596






Mus musculus


beta-1,6-N-acetylglucosaminyltransferase B




1257




73






666




AF168132






Homo sapiens


RU1




2485




55






667




AF132961






Homo sapiens


CGI-27 protein




1197




100






668




AB025258






Mus musculus


granuphilin-a




729




42






669




AL391688






Homo sapiens


bA524D16A.2.1




762




43








(novel protein similar to mouse granuphilin-a)






670




AP001745






Homo sapiens


human cDNA DKFZp586F0422, Accession No. AL050173




3747




100






671




X58454






Homo sapiens


DS dopamine receptor




1024




85






672




AB017059






Arabidopsis thaliana


FH protein interacting protein FIP2




212




35






673




AF255908






Streptococcus pneumoniae


PspA




177




29






674




Y02669




Human secreted protein encoded by gene 20 clone HMKAH10.




288




100






675




AB024986






Oryza sativa


cyclin




142




48






676




X70681






Xenopus laevis


zinc finger protein




144




33






677




AL390026






Homo sapiens


dJ336K20B.1 (novel protein based on FGENESH)




964




100






678




AC026815






Oryza sativa


putative ATP-dependent RNA helicase (5′-partial)




437




43






679




AF196779






Homo sapiens


JM10 protein




1182




98






680




U40802






Caenorhabditis elegans


similar to other protein phosphatases 1, 2A and 2B




325




36






681




AJ238854






Rattus norvegicus


type A/B hnRNP p40




1659




91






682




Y36083




Extended human secreted protein sequence, SEQ. ID NO. 468.




430




98






683




U37283






Homo sapiens


microfibril-associated glycoprotein-2 MAGP-2




846




94






684




AJ235270






Rickettsia prowazekii


50S RIBOSOMAL PROTEIN L10 (rplJ)




122




23






685




W67828




Human secreted protein encoded by gene 22 clone HFEAF41.




510




100






686




W67828




Human secreted protein encoded by gene 22 clone HFEAF41.




403




85






687




AK026226






Homo sapiens


unnamed protein product




1937




100






688




AK026226






Homo sapiens


unnamed protein product




578




100






689




D26549






Bos taurus


bovine adseverin




3527




92






690




AK024644






Homo sapiens


unnamed protein product




245




67






691




X63546






Homo sapiens


oncogene




1083




78






692




X63546






Homo sapiens


oncogene




1613




83






693




AB029334






Halocynthia roretzi


HrPET-1




614




37






694




AF286473






Mus musculus


retinitis pigmentosa GTPase regulator




181




35






695




AB002405






Homo sapiens


LAK-4p




346




34






696




Y14448




Human secreted protein encoded by gene 38 clone HFGAH44.




316




100






697




AL133216






Homo sapiens


bA291L22.2




256




100








(similar to CDC10 (cell division cycle 10,


S. cerevisiae


, homolog))






698




AF065389






Homo sapiens


tetraspan NET-4




1111




74






699




AF161390






Homo sapiens


HSPC272




281




81






700




Z83114






Caenorhabditis elegans


K09B11.2




233




37






701




Y41360




Human secreted protein encoded by gene 53 clone H3PAD75.




490




100






702




Y41360




Human secreted protein encoded by gene 53 clone HJPAD75.




233




92






703




AL049481






Arabidopsis thaliana


putative protein




829




53






704




U67949






Caenorhabditis elegans


contains similarity t to sugar and other




486




36








transporters (Pfam: sugar_tr.hmm.score: 13.46)






705




J02883






Homo sapiens


colipase precursor




243




100






706




X66286






Gallus gallus


tensin




1009




73






707




G01118




Human secreted protein, SEQ ID NO: 5199.




360




100






708




D50857






Homo sapiens


DOCK180 protein




1060




51






709




AE003511






Drosophila melanogaster


CG14194 gene product




542




54






710




L26247






Homo sapiens


isolog of yeast sui1 and rice gos2; putative




470




86






711




Y12952




Amino acid sequence of a human secreted peptide.




362




98






712




U41849






Saccharomyces cerevisiae


Yta6p




156




23






713




AF168362






Rattus norvegicus


protein associating with small stress protein PASS1




220




35






714




Y48256




Human prostate cancer-associated protein 42.




242




75






715




AF053130






Mus musculus


unconventional myosin MYO15




579




36






716




AF083385






Homo sapiens


30 kDa splicing factor; SPF 30




133




40






717




AF182417






Homo sapiens


MDS016




406




100






718




AF065389






Homo sapiens


tetraspan NET-4




584




43






719




W78132




Human secreted protein encoded by gene 7 clone HPEBD85.




246




100






720




AK023971






Homo sapiens


unnamed protein product




1358




100






721




AF288813






Mus musculus


synembryn




2384




86






722




X56203






Plasmodium falciparum


liver stage antigen




188




19






723




AF190501






Homo sapiens


leucine-rich repeat-containing G protein-coupled receptor 6




4286




99






724




AF022789






Homo sapiens


ubiquitin hydrolyzing enzyme I




1689




88






725




AF176531






Mus musculus


F-box protein FBX16




1412




80






726




U10991






Homo sapiens


G2




756




30






727




AE001795






Thermotoga maritima


glycerol kinase




845




38






728




AJ006692






Homo sapiens


ultra high sulfer keratin




868




71






729




Y76177




Human secreted protein encoded by gene 54.




866




99






730




AL162295






Arabidopsis thaliana


guanine nucleotide exchange factor-like protein




261




24






731




AF016036






Drosophila tsacasi


repressor-like protein




220




27






732




Z54327






Caenorhabditis elegans


contains similarity to Pfam domain: PF00070




339




32








(pyridine nucleotide-disulphide oxidoreductase),








Score = 82.7, E-value = 4e-24, N = 2






733




AC011713






Arabidopsis thaliana


Contains similarity to gb|AF092102 G-protein




303




26








beta subunit git5p from


Schizosaccharomyces pombe


and contains








2 PF|00400WD doinain, G-beta repeat domains.






734




AL109965






Homo sapiens


dJ1121G12.1.2




323




60








(A novel protein containing a putative PPD finger domain, isoform 2)






735




AB000910






Sus scrofa


ribosomal protein




483




87






736




Y07931




Human secreted protein fragment encoded from gene 80.




213




100






737




M95718






Oryctolagus cuniculus


keratin




233




65






738




AL034382






Schizosaccharomyces pombe


putative Trp-Asp repeat protein




453




35






739




Y38401




Human secreted protein encoded by gene No. 16.




233




88






740




U18917






Saccharomyces cerevisiae


Yer157wp




338




26






741




W64471




Human secreted protein from clone DF989_3.




545




97






742




AJ000474






Homo sapiens


cytidine deaminase




389




100






743




AL132954






Arabidopsis thaliana


putative protein




361




34






744




AF026292






Homo sapiens


chaperonin containing t-complex polypeptide 1,




890




100








eta subunit; CCT-eta






745




Y60251




Human endometrium tumour EST encoded protein 311.




392




98






746




U07817






Dictyostelium discoideum


glutamine-asparagine rich protein




148




30






747




M58529






Homo sapiens


pro-alpha-2 type V collagen




158




100






748




AL118506






Homo sapiens


dJ591C20.5 (KIAA1196)




952




45






749




AF181645






Drosophila melanogaster


BcDNA.GH12144




105




54






750




AF067136






Homo sapiens


protein phosphatase-1 regulatory subunit 7 alpha2




147




38






751




AJ006973






Homo sapiens


TOM1




638




82






752




AF145664






Drosophila melanogaster


BcDNA.GH11110




766




37






753




Z70310






Caenorhabditis elegans


contains similarity to Pfam domain: PF00013




510




34








(KH domain), Score = 42.8, E-value = 3.7e-12, N = 1;








PF00023 (Ank repeat), Score = 428.2, E-value = 2.4e-125, N = 19






754




AF276514






Mus musculus


105-kDa kinase-like protein




3769




88






755




Y30734




Amino acid sequence of a human secreted protein.




258




96






756




AF102129






Rattus norvegicus


KPL2




1436




79






757




L15313






Caenorhabditis elegans


putative




829




58






758




AE003584






Drosophila melanogaster


CG7289 gene product




529




34






759




Y66693




Membrane-bound protein PRO1004.




569




92






760




AC007766






Homo sapiens


R26610_1




1574




80






761




AF073958






Homo sapiens


cytokine-inducible SH2 protein 6




1142




54






762




Z71181






Caenorhabditis elegans


contains similarity to Pfam domain: PF00561




498




34








(alpha/beta hydrolase fold)








Score = 83.7, E-value = 1.2e-21, N = 1






763




AC005724






Arabidopsis thaliana


putative C3HC4-type RING zinc finger protein




166




45






764




AF052433






Strongylocentrotus purpuratus


katanin p80 subunit




170




30






765




X56044






Mus musculus


protein Htf9C




759




44






766




U56418






Homo sapiens


lysophosphatidic acid acyltransferase-beta




834




100






767




AK024388






Homo sapiens


unnamed protein product




675




100






768




W67828




Human secreted protein encoded by gene 22 clone HFEAF41.




278




100






769




AF055993






Homo sapiens


mSin3A associated polypeptide p30




637




71






770




AK024512






Homo sapiens


unnamed protein product




454




93






771




AF095844






Homo sapiens


melanoma differentiation associated protein-5




687




40






772




AF169257






Homo sapiens


sodium/calcium exchanger NCKX3




3084




99






773




D00824






Gallus gallus


alpha 1 chain of type XII collagen




302




30






774




X82557






Rattus norvegicus


myelin and lymphocyte protein




257




39






775




G03200




Human secreted protein, SEQ ID NO: 7281.




548




98






776




AK024793






Homo sapiens


unnamed protein product




1730




100






777




AB019397






Homo sapiens


DNA topoisomerase II binding protein




108




23






778




AK025033






Homo sapiens


unnamed protein product




817




99






779




AB028069






Homo sapiens


activator of S phase Kinase




474




36






780




AF273052






Homo sapiens


CTCL tumor antigen se70-2




1294




100






781




AF217512






Homo sapiens


uncharacterized bone marrow protein BM036




130




100






782




AF121857






Homo sapiens


sorting nexin 7




2019




100






783




AF305941






Homo sapiens


LIM2




933




100






784




AF153085






Homo sapiens


phosphoprotein pp75




178




31






785




AF106584






Caenorhabditis elegans


contains similarity to homeobox domains




255




32








(Pfam: PF00046, score = 16.2, E = 0.015, N = 1)






786




AF106584






Caenorhabditis elegans


contains similarity to homeobox domains




244




31








(Pfam: PF00046, score = 16.2, E = 0.015, N = 1)






787




AJ278508






Mus musculus


MDM2 binding protein




567




76






788




Z21507






Homo sapiens


human elongation factor-1-delta




269




96






789




AF096286






Mus musculus


pecanex 1




1498




52






790




AC004882






Homo sapiens


similar to calmodulin; similar to P24044 (PID: g115520)




525




60






791




AJ133500






Xenopus laevis


p33 ringo




547




49






792




U41264






Caenorhabditis elegans


coded for by


C. elegans


CDNA cm13gl;




212




50








Similar to bumetanide-sensitive Na—K—Cl cotransporter






793




X16078






Torpedo californica


4-acetamido-4′-isothiocyanostilbene-2,2′-disulphonic




423




38








acid-binding protein






794




AK026447






Homo sapiens


unnamed protein product




833




100






795




AL035417






Homo sapiens


dJ891H21.1 (HYPOTHETICAL 43.1 KD PROTEIN)




465




100






796




M83751






Homo sapiens


arginine-rich protein




525




62






797




AB037834






Homo sapiens


KIAA1413 protein




7273




99






798




AB037834






Homo sapiens


KIAA1413 protein




6449




98






799




AC011717






Arabidopsis thaliana


putative carnitine/acylcarnitine translocase;




450




37








50581-51656






800




AC069143






Arabidopsis thaliana


Contains similarity to a transposable element Tip100




305




25








protein for transposase from


Ipomoea purpurea


gb|4063769 and is a








member of the transmembrane 4 family PF|00335.






801




AY008372






Homo sapiens


oxysterol binding protein-related protein 3




4671




100






802




AC004557






Arabidopsis thaliana


F17L21.20




404




39






803




AF182218






Homo sapiens


epidermal lipoxygenase




3830




99






804




U90353






Strongyloides stercoralis


IgG and IgE immuncreactive antigen recognized




108




35








by sera from patients with strongyloidiasis






805




D13989






Homo sapiens


human rho GDI




818




99






806




U00482






Homo sapiens


gamma-subunit of rod cGMP-phosphodiesterase




472




100






807




Y19599




SEQ ID NO 317 from WO9922243.




590




98






808




D50617






Saccharomyces cerevisiae


YFL042C




227




31






809




X58430






Homo sapiens


homeobox protein




478




100






810




M98539






Homo sapiens


prostaglandin D2 synthase




464




97






811




AJ004872






Homo sapiens


TCR beta chain




1296




92






812




G00329




Human secreted protein, SEQ ID NO: 4410.




524




100






813




AL031431






Homo sapiens


dJ462O23.2 (novel protein)




824




45






814




M37760






Mus musculus


serine 2 ultra high sulfur protein




633




55






815




AL121673






Homo sapiens


bA305P22.2 (novel protein)




2396




100






816




AB027568






Mus musculus


thiamin pyrophosphokinase




664




91






817




M37190






Homo sapiens


ras inhibitor




792




40






818




AL109659






Homo sapiens


dJ1024N4.1




2858




100








(novel Sodium:solute symporter family member similar to SLC5A1








(SGLT1))






819




AB000170






Sus scrofa


endopeptidase 24.16 type M1




3472




94






820




AF058789






Rattus norvegicus


SynGAP-a




6651




99






821




AK024408






Homo sapiens


unnamed protein product




1040




100






822




Y08260






Mus musculus


cytoplasmic polyadenylation element-binding protein (CPEB)




2453




95






823




Y36090




Extended human secreted protein sequence, SEQ ID NO. 475.




420




95






824




AC003682






Homo sapiens


R28530_2




1398




48






825




D89052






Homo sapiens


proton-ATPase-like protein




356




100






826




AB039903






Homo sapiens


interferon-responsive finger protein 1 long form




978




61






827




U96963






Mus musculus


p140mDia




471




26






828




AJ251641






Mus musculus


syncoilin




656




89






829




Z98595






Schizosaccharomyces pombe


coronin-like protein




196




24






830




AF116660






Homo sapiens


PRO1430




299




100






831




AJ277442






Homo sapiens


xylosyltransferase II




4625




100






832




D86081






Mus musculus


S-II-T1




290




36






833




L21671






Mus musculus


Eps8




547




31






834




Z34801






Caenorhabditis elegans


Similarity with drosphila MSP-300 protein




283




25








(PIR acc. no. S30431) , contains similarity to Pfam domain: PF01465








(GRIP domain), Score = 90.2, E-value = 1.4e-23, N = 1






835




AF264750






Homo sapiens


ALR-like protein




21361




100






836




AF176814






Mus musculus


Ab1-philin 2




247




41






837




AL121771






Homo sapiens


dJ548G19.1.2




166




24








(novel protein (ortholog of mouse zinc finger protein ZFP64)








(translation of cDNA NT2RP4001938 (Em: AK001744)) (isoform 2))






838




G00405




Human secreted protein, SEQ ID NO: 4486.




397




98






839




AF130357






Mus musculus domesticus


similar to RNA binding protein




222




53






840




AJ272034






Homo sapiens


putative capacitative calcium channel




4470




100






841




G02337




Human secreted protein, SEQ ID NO: 6418.




377




98






842




AF286475






Takifugu rubripes


retinitis pigmentosa GTPase regulator-like protein




226




22






843




AB030183






Mus musculus


contains transmembrane (TM) region




1159




89






844




Y18890




Human endogenous retrovirus K gag protein




273




47






845




AC004890






Homo sapiens


similar to HUB1; similar to BAA24380 (PID: g2789430)




435




79






846




Y66666




Membrane-bound protein PRO1013.




1858




99






847




Y66666




Membrane-bound protein PRO1013.




1658




93






848




AF119913






Homo sapiens


PRO3077




620




100






849




Y36233




Human secreted protein encoded by gene 10.




302




96






850




D14530






Homo sapiens


ribosomal protein




341




100






851




AC011713






Arabidopsis thaliana


Is a member of the PP|01553 Acyltransferase family.




162




39






852




Y36154




Human secreted protein #26.




296




98






853




AC004877






Homo sapiens


zinc finger-like; similar to P52742 (PID: g1731411)




2830




100






854




M34163






Mus musculus


low affinity IgE receptor (FC-epsilon-RII)




293




29






855




X73579






Rattus norvegicus


CD23




333




31






856




M34427






Homo sapiens


T-plastin




414




98






857




Y19456




Amino acid sequence of a human secreted protein.




307




90






858




AF067136






Homo sapiens


protein phosphatase-1 regulatory subunit 7 betal




199




47






859




AC023279






Arabidopsis thaliana


F12K21.21




441




38






860




G04069




Human secreted protein, SEQ ID NO: 8150.




491




100






861




AP001297






Arabidopsis thaliana gene


_id:F14O13.28˜




488




28






862




Y19587




Amino acid sequence of a human secreted protein.




160




96






863




AB039861






Schizosaccharomyces pombe


Esol




201




31






864




U34932






Rattus norvegicus


Fos-related antigen




2062




84






865




Y60152




Human endometrium tumour EST encoded protein 212.




379




100






866




AK027028






Homo sapiens


unnamed protein product




2294




99






867




AF039718






Caenorhabditis elegans


contains similarity to lupus LA protein homologs




329




44






868




AK024480






Homo sapiens


FLJ00074 protein




1001




100






869




AF320909






Homo sapiens


MAGE-E1




634




100






870




AF126484






Homo sapiens


CARD4




181




30






871




Z83844






Homo sapiens


dJ37E16.5




651




46








(novel protein similar to nitrophenylphosphatases from various organisms)






872




AF128406






Homo sapiens


nuclear prelamin A recognition factor




1121




47






873




Y16790






Homo sapiens


keratin type I




2029




99






874




AJ006692






Homo sapiens


ultra high sulfer keratin




1073




88






875




U43281






Saccharomyces cerevisiae


Lpg21p




323




40






876




AF078844






Homo sapiens


hqp0376 protein




362




81






877




AB025258






Mus musculus


granuphilin-a




3193




90






878




D37991






Homo sapiens


beta-signal sequence receptor




847




99






879




AL035427






Homo sapiens


dJ769N13.1 (KIAA0443 protein.)




510




28






880




AL109925






Homo sapiens


dJ534K7.2 (novel protein)




3323




100






881




AF254411






Homo sapiens


ser/arg-rich pre-mRNA splicing factor SR-A1




6833




100






882




AL137082






Arabidopsis thaliana


putative protein




310




42






883




M97188






Strongylocentrotus purpuratus


tektin A1




292




40






884




Y10837




Amino acid sequence of a human secreted protein.




434




100






885




Z12172






Homo sapiens


putative homeotic protein




1388




79






886




AJ000479






Homo sapiens


putative G-Protein coupled receptor, EDG6




177




30






887




X56044






Mus musculus


protein Htf9C




2360




75






888




AC006963






Homo sapiens


similar to Kelch proteins; similar to BAA77027 (PID: g4650844)




268




27






889




AF205357






Drosophila melanogaster


extracellular matrix protein papilin




503




33






890




AB027757






Cicer arietinum


NADPH oxidoreductase homolog




719




45






891




AL022140






Arabidopsis thaliana


serine/threonine protein kinase like protein




153




30






892




AF286473






Mus musculus


retinitis pigmentosa GTPase regulator




208




28






893




AL050328






Homo sapiens


bA145L22.2




2072




99








(novel KRAB box containing C2H2 type zinc finger protein)






894




AL096856






Arabidopsis thaliana


putative protein




223




44






895




AF245517






Homo sapiens


vacuolar proton pump 116 kDa accessory subunit




4378




100






896




AB000113






Rattus norvegicus


cationic amino acid transporter 3




2644




83






897




AF142406






Babesia bigemina


200 kDa antigen p200




588




24






898




AF062476






Mus musculus


retinoic acid-responsive protein; STRA6




2184




75






899




Y48507




Human breast tumour-associated protein 52.




308




98






900




U79745






Homo sapiens


monocarboxylate transporter homologue MCT6




465




46






901




AF145661






Drosophila melanogaster


BcDNA.GH10646




376




24






902




AC009519






Arabidopsis thaliana


F1N19.17




241




29






903




Y27576




Human secreted protein encoded by gene No. 10.




394




96






904




AF192968






Homo sapiens


high-glucose-regulated protein 8




2041




67






905




AP001743






Homo sapiens


putative gene, ankirin like, possible dual specifity




4306




99








Ser/Thr/Tyr kinase domain






906




U71205






Mus musculus


rit




252




35






907




M77003






Mus musculus


glycerol-3-phosphate acyltransferase




3366




93






908




AF286368






Homo sapiens


eppin-1




222




54






909




AC006439






Arabidopsis thaliana


putative ADP-ribosylation factor




261




34






910




AF230808






Homo sapiens


zinc finger transcription factor Pegasus




2279




99






911




AF134804






Mus musculus


putative zinc finger transcription factor OVO1




698




52






912




AF049907






Homo sapiens


zinc finger transcription factor




530




31






913




AF217319






Mus musculus


putative repair and recominbination helicase RAD26L




3218




89






914




X80473






Mus musculus


rab19




1008




88






915




U88315






Caenorhabditis elegans


weak similarity to Plasmodium vivax




609




36








reticulocyte-binding protein 2 (GI: 160628)






916




AF273052






Homo sapiens


CTCL tumor antigen se70-2




1237




99






917




U34932






Rattus norvegicus


Fos-related antigen




2258




84






918




AF116911






Mus musculus


thymic dendritic cell-derived factor 1




305




92






919




AF205935






Mus musculus


MGA protein




4822




84






920




AL157413






Homo sapiens


bA526K17.1 (novel protein).




1076




100






921




Y10823




Amino acid sequence of a human secreted protein.




183




100






922




Z97653






Homo sapiens


c380A1.1b (novel protein)




1106




100






923




Z24725






Homo sapiens


mitogen inducible gene mig-2




1461




56






924




AF113917






Homo sapiens


NADP+-dependent isocitrate dehydrogenase




1428




100






925




AF064604






Homo sapiens


KE03 protein




1025




57






926




AF064604






Homo sapiens


KE03 protein




972




58






927




AL033545






Arabidopsis thaliana


putative protein




84




35






928




AJ007014






Homo sapiens


AMMECR1 protein




733




64






929




AJ276316






Homo sapiens


zinc finger protein 304




1007




51






930




AC004077






Arabidopsis thaliana


putative katanin




877




57






931




D16226






Oryctolagus cuniculus


one of the members of sodium-glucose




2008




71








cotransporter family






932




D16226






Oryctolagus cuniculus


one of the members of sodium-glucose




3026




83








cotransporter family






933




AF234676






Sus scrofa


adipose differentiation-related protein




336




29






934




U70732






Homo sapiens


glutamate pyruvate transaminase




1821




68






935




AB027004






Homo sapiens


protein phosphatase




462




48






936




AF043180






Homo sapiens


T cell receptor beta chain




1097




69






937




AC006528






Arabidopsis thaliana


putative DNA replication licensing factor




742




41






938




L08483






Drosophila melanogaster


ring canal protein




660




31






939




AL022238






Homo sapiens


dJ1042K10.4 (novel protein)




758




46






940




AF275151






Rattus norvegicus


androgen receptor-related apoptosis-associated protein




619




55








CBL27






941




AF275151






Rattus norvegicus


androgen receptor-related apoptosis-associated protein




619




55








CBL27






942




AE000854






Methanothermobacter thermoautotrophicus


Na+/H+-exchanging protein:




498




35








Na+/H+ antiporter






943




D88894






Homo sapiens


brain acyl-CoA hydrolase




1742




100






944




AP001072






Oryza sativa


Similar to


Arabidopsis thaliana


DNA chromosome 4,




248




33








BAC clone F22K18, putative protein. (AL035356)






945




AB033744






Mus musculus


type II cytokeratin




835




74






946




AP186461






Rattus norvegicus


ring finger protein Fxy




351




30






947




AK025539






Homo sapiens


unnamed protein product




2555




99






948




J03407






Homo sapiens


rfp transforming protein




507




40

























TABLE 3









SEQ ID NO:




ACCESSION NO.




DESCRIPTION




RESULTS*


























2




BL00972




Ubiquitin carboxyl-




BL00972A 11.93 2.500e-20 267-285








terminal hydrolases




BL00972D 22.55 5.179e-17 828-853








family 2 proteins.




BL00972E 20.72 8.650e-13 855-877









BL00972C 16.48 7.120e-11 411-426









BL00972B 9.45 7.923e-10 353-363






3




BL00018




EF-hand calcium-




BL00018 7.41 5.696e-09 331-344








binding domain proteins.






4




BL00018




EF-hand calcium-




BL00018 7.41 5.696e-09 331-344








binding domain proteins.






5




PR00401




SH2 DOMAIN SIGNATURE




PR00401B 12.94 1.000e-08 340-351






6




PR00401




SH2 DOMAIN SIGNATURE




PR00401B 12.94 1.000e-08 367-378






7




BL00625




Regulator of chromosome




BL00625A 16.21 7.787e-16 308-337








condensation (RCC1)




BL00625A 16.21 7.369e-15 190-219








proteins.




BL00625B 17.69 1.514e-13 302-336









BL00625B 17.69 2.286e-13 184-218









BL00625B 17.69 3.957e-13 132-166









BL00625A 16.21 5.690e-13 138-167









BL00625A 16.21 5.731e-11 360-389









BL00625B 17.69 3.333e-10 354-388






9




BL00120




Lipases, serine proteins.




BL00120B 11.37 9.486e-12 166-181






15




BL00183




Ubiquitin-conjugating




BL00183 28.97 1.329e-10 45-93








enzymes proteins.






18




PR00049




WILM'S TUMOUR




PR00049D 0.00 6.034e-09 262-277








PROTEIN SIGNATURE






19




PR00049




WILM'S TUMOUR




PR00049D 0.00 6.034e-09 74-89








PROTEIN SIGNATURE






20




BL00790




Receptor tyrosine




BL00790D 12.41 8.297e-09 804-829








kinase class V proteins.






21




BL00790




Receptor tyrosine




BL00790D 12.41 8.297e-09 878-903








kinase class V proteins.






23




PR00380




KINESIN HEAVY CHAIN




PR00380D 9.93 2.080e-22 321-343








SIGNATURE




PR00380A 14.18 1.486e-21 79-101









PR00380B 12.64 6.571e-18 217-235









PR00380C 13.18 6.927e-13 269-288






25




BL01242




Formamidopyrimidine-




BL01242F 17.92 5.300e-11 32-66








DNA glycosylase proteins.






27




PF00651




BTB (also known as BR-C/Ttk)




PF00651 15.00 2.500e-14 46-59








domain proteins.






28




DM00215




PROLINE-RICH PROTEIN 3.




DM00215 19.43 3.898e-09 99-132






29




PR00380




KINESIN HEAVY CHAIN




PR00380A 14.18 9.250e-25 93-115








SIGNATURE




PR00380D 9.93 4.857e-19 302-324









PR00380B 12.64 4.429e-18 212-230









PR00380C 13.18 1.692e-16 247-266






30




BL00107




Protein kinases ATP-binding




BL00107A 18.39 3.368e-18 36-67








region proteins.






32




BL00594




Aromatic amino acids




BL00594A 16.75 9.376e-09 76-120








permeases proteins.






34




BL00790




Receptor tyrosine




BL00790E 29.58 1.111e-12 614-662








kinase class V proteins.




BL00790E 29.58 3.111e-12 668-716









BL00790E 29.58 7.000e-10 560-608






38




BL00290




Immunoglobulins and




BL00290A 20.89 4.150e-12 126-149








major histocompatibility








complex proteins.






39




PR00019




LEUCINE-RICH REPEAT




PR00019A 11.19 6.087e-10 93-107








SIGNATURE




PR00019B 11.36 7.840e-09 90-104






42




PD01443




INHIBITOR CALPAIN




PD01443D 8.36 4.670e-09 815-837








CALPASTATIN REPEAT








THIOL PROT.






43




BL01101




Casein kinase II




BL01101A 16.07 1.000e-40 9-54








regulatory subunit proteins.




BL01101B 10.94 9.000e-31 72-97






46




BL01166




RNA polymerases beta




BL01166G 18.10 2.500e-34 824-866








chain proteins.




BL01166H 19.05 9.410e-30 936-986









BL01166D 17.37 4.396e-19 612-642









BL01166E 13.47 8.244e-17 682-706









BL01166C 12.21 9.357e-12 431-456






47




BL00518




Zinc finger, C3HC4 type




BL00518 12.23 7.000e-09 25-34








(RING finger), proteins.






48




BL00038




Myc-type, ′helix-




BL00038A 13.61 6.625e-11 284-300








loop-helix′








dimerization domain proteins.






49




BL00905




GTP1/OBG family proteins.




BL00905D 15.00 4.214e-10 125-140






50




BL00107




Protein kinases ATP-




BL00107B 13.31 7.300e-15 64-80








binding region proteins.






53




BL00383




Tyrosine specific




BL00383E 10.35 5.263e-09 328-339








protein phosphatases proteins.






55




BL00383




Tyrosine specific




BL00383E 10.35 5.263e-09 246-257








protein phosphatases proteins.






56




BL00383




Tyrosine specific




BL00383E 10.35 5.263e-09 328-339








protein phosphatases proteins.






62




BL00226




Intermediate filaments proteins.




BL00226B 23.86 5.919e-09 560-608






64




PR00322




G10 PROTETN




PR00322E 6.62 1.720e-10 30-40






65




BL00269




Mammalian defensins proteins.




BL00269C 16.52 6.786e-26 110-139









BL00269A 8.53 2.607e-20 45-65









BL00269B 19.17 5.500e-17 72-101






66




BL01160




Kinesin light chain




BL01160B 19.54 8.297e-10 6-60








repeat proteins.






67




BL00028




Zinc finger, C2H2




BL00028 16.07 5.846e-11 476-493








type, domain proteins.




BL00028 16.07 6.192e-11 989-1006






71




BL00289




Pentaxin family proteins.




BL00289E 18.00 4.375e-13 22-37






74




BL00348




p53 tumor antigen proteins.




BL00348F 23.19 4.571e-09 140-183






75




BL00455




Putative AMP-binding




BL00455 13.31 6.684e-13 248-264








domain proteins.






78




BL00421




Transmembrane 4




BL00421E 20.97 1.851e-09 17-47








family proteins.






80




PD01066




PROTEIN ZINC FINGER




PD01066 19.43 2.149e-29 6-45








ZINC-FINGER METAL-








BINDINC NU.






81




PR00014




FIBRONECTIN TYPE III




PR00014D 12.04 2.059e-10 215-230








REPEAT SIGNATURE






84




BL00269




Mammalian defensins proteins.




BL00269C 16.52 6.786e-26 133-162









BL00269A 8.53 2.607e-20 68-88









BL00269B 19.17 5.500e-17 95-124






85




PD01876




ANTIGEN MELANOMA-




PD01876C 21.73 1.231e-20 75-128








ASSOCIATED MULTIGENE








FAMILY TUM.






86




PD02870




RECEPTOR




PD02870B 18.83 8.835e-11 326-359








INTERLEUKIN-1








PRECURSOR.






87




PR00988




URIDINE KINASE




PR00988A 6.39 6.276e-12 386-376








SIGNATURE






90




BL00120




Lipases, serine proteins.




BL00120C 12.62 9.053e-12 95-106






96




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 5.500e-30 169-212






100




BL01128




Shikimate kinase proteins.




BL01128A 18.84 8.200e-14 7-41






101




PR00014




FIBRONECTIN TYPE III




PR00014C 15.44 1.783e-09 211-230








REPEAT SIGNATURE




PR00014A 8.22 3.045e-09 373-383









PR00014C 15.44 6.087e-09 309-328






102




DM00372




CARCINOEMBRYONIC




DM00372C 23.69 4.919e-12 67-103








ANTIGEN PRECURSOR








AMINO-TERMINAL








DOMAIN.






105




BL01282




BIR repeat proteins.




BL01282B 30.49 1.000e-11 194-233






109




PR00464




E-CLASS P450 GROUP




PR00464A 20.47 9.591e-16 149-170








II SIGNATURE




PR00464C 18.84 1.000e-15 324-353









PR00464D 17.40 6.250e-15 353-371









PR00464B 20.41 1.844e-12 205-224






110




PD02382




RECEPTOR CHAIN




PD02382A 17.43 9.321e-09 99-115








PRECURSOR TRANSME.






112




BL00795




Involucrin proteins.




BL00795C 17.06 6.442e-10 905-950






115




DM00215




PROLINE-RICH PROTEIN 3.




DM00215 19.43 6.644e-09 603-636






116




BL01282




BIR repeat proteins.




BL01282B 30.49 1.000e-11 137-176






117




BL01282




BIR repeat proteins.




BL01282B 30.49 1.000e-11 187-226






118




BL00218




Amino acid permeases proteins.




BL00218D 21.49 7.324e-11 226-271









BL00218E 23.30 3.475e-09 307-347






119




BL00994




Bacterial export




BL00994A 15.15 1.086e-09 71-118








FHIPEP family proteins.






120




PD01066




PROTEIN ZINC FINGER




PD01066 19.43 8.385e-33 6-45








ZINC-FINGER METAL-








BINDING NU.






121




PD01427




TRANSFERASE




PD01427B 22.45 1.545e-11 117-158








METHYLTRANSFERASE BI.






122




PF00168




C2 domain proteins.




PR00168C 27.49 1.750e-09 202-228






127




PR00962




LETHAL(2) GIANT




PR00962D 10.40 3.054e-10 178-202








LARVAE PROTEIN








SIGNATURE






130




DM01970




0 kw ZK632.12




DM01970B 8.60 2.478e-13 310-323








YDR313C ENDOSOMAL III.






131




PF00774




Dihydropyridine sensitive




PF00774D 10.59 8.396e-09 339-365








L-type calcium channel








(Beta subuni.






132




BL00615




C-type lectin domain proteins.




BL00615A 16.68 3.160e-11 129-147






133




PD01066




PROTEIN ZINC FINGER




PD01066 19.43 2.705e-11 47-86








ZINC-FINGER METAL-








BINDING NU.






134




BL00359




Ribosomal protein




BL00359B 23.07 7.462e-24 160-201








L11 proteins.




BL00359C 22.18 6.586e-22 215-249









BL00359A 20.66 4.000e-21 124-160






136




PD02462




PROTEIN BOLA




PD02462A 22.48 1.220e-09 104-139








TRANSCRIPTION








REGULATION AC.






137




PR00679




PROHIBITIN SIGNATURE




PR00679F 8.03 6.478e-28 178-202









PR00679C 14.44 7.677e-22 107-126









PR00679E 12.82 5.171e-19 153-173









PR00679D 11.91 9.053e-18 130-147









PR00679G 6.13 7.882e-17 201-218









PR00679B 13.63 2.444e-10 84-104






138




PR00245




OLFACTORY RECEPTOR




PR00245E 12.40 8.286e-12 45-60








SIGNATURE






139




PD00126




PROTEIN REPEAT




PD00126A 22.53 6.885e-10 99-120








DOMAIN TPR NUCLEA.






140




BL01145




Ribosomal protein




BL01145A 13.73 1.000e-12 3-45








L34e proteins.






145




BL00154




E1-E2 ATPases




BL00154D 12.57 7.387e-09 95-106








phosphorylation site proteins.






147




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 7.923e-15 439-452








METAL-BINDI.




PD00066 13.92 2.800e-14 411-424









PD00066 13.92 2.800e-14 467-480









PD00066 13.92 5.800e-14 495-508









PD00066 13.92 5.800e-14 523-536









PD00066 13.92 8.200e-14 355-368









PD00066 13.92 5.500e-13 579-592









PD00066 13.92 3.143e-12 551-564









PD00066 13.92 4.857e-12 383-396






149




BL00649




G-protein coupled




BL00649C 17.82 9.542e-12 400-426








receptors family 2 proteins.






152




BL00479




Phorbol esters/diacylglycerol




BL00479B 12.57 8.875e-09 886-902








binding domain proteins.






153




PR00205




CADHERIN SIGNATURE




PR00205B 11.39 5.655e-16 255-273









PR00205A 14.73 1.000e-12 180-196









PR00205B 11.39 4.927e-10 475-493









PR00205C 13.65 9.438e-10 515-530






155




BL00122




Carboxylesterases




BL00122A 12.04 3.152e-15 86-107








type-B serine proteins.




BL00122D 12.53 7.097e-14 197-213









BL00122B 16.84 1.346e-13 148-159









BL00122C 7.91 9.550e-10 168-179






157




BL00018




EF-hand calcium-




BL00018 7.41 2.800e-10 217-230








binding domain proteins.




BL00018 7.41 8.650e-10 133-146






160




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 6.143e-12 927-940








METAL-BINDI.




PD00066 13.92 7.000e-09 343-356






161




DM01857




5 kw NUCLEOSIDE




DM01857B 14.94 6.471e-19 284-312








TRANSPORT DEPENDENT




DM01B57E 18.90 7.313e-18 488-527








NA.




DM01857F 12.86 7.045e-15 548-575









DM01857C 15.62 4.500e-14 312-344









DM01857A 20.25 1.667e-13 207-250









DM01857D 16.80 3.165e-12 372-410






164




DM01840




kw SPAC24B11.09




DM01840B 22.04 1.844e-40 59-103








R07E5.13.




DM01840A 10.95 9.571e-13 31-43






166




BL01115




GTP-binding nuclear




BL01115A 10.22 3.438e-14 53-97








protein ran proteins.






167




PF00622




Domain in SPla and




PF00622B 21.00 2.500e-13 265-287








the Ryanodine Receptor.






168




PR00019




LEUCINE-RICH REPEAT




PR00019A 11.19 5.050e-11 66-80








SIGNATURE




PR00019B 11.36 6.850e-10 63-77






169




BL00509




Ras GTPase-activating




BL00509B 10.28 5.263e-10 429-440








proteins.






172




PR00720




MAMMALIAN LMW




PR00720C 12.41 1.099e-27 88-109








PHOSPHOTYROSINE




PR00720B 10.61 4.789e-20 71-87








PROTEIN PHOSPHATASE




PR00720A 16.54 2.000e-17 28-41








SIGNATURE




PR00720E 10.01 1.342e-16 117-139









PR00720D 17.32 1.778e-15 110-127






173




PD00131




ATP-BINDING




PD00131B 34.97 7.987e-09 108-162








TRANSPORT








TRANSMEMBR.






175




BL00615




C-type lectin domain




BL00615A 16.68 9.526e-13 573-591








proteins.






179




BL00134




Serine proteases,




BL00134A 11.96 5.781e-15 493-510








trypsin family,




BL00134B 15.99 4.194e-14 675-699








histidine proteins.






180




BL00236




Neurotransmitter-gated




BL00236D 25.66 4.000e-30 64-106








ion-channels proteins.






181




BL00604




Synaptophysin/




BL00604F 5.96 7.718e-10 367-412








synaptoporin proteins.






184




PR00042




FOS TRANSFORMING




PR00042E 9.69 7.652e-09 234-258








PROTEIN SIGNATURE






187




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 9.400e-14 365-378








METAL-BINDI.




PD00066 13.92 6.143e-12 335-348









PD00066 13.92 2.174e-11 395-408






188




BL00962




Ribosomal protein S2




BL00962D 22.51 5.500e-35 131-175








proteins.




BL00962C 15.90 9.591e-17 106-124









BL00962B 36.15 9.060e-15 40-94






189




BL00152




ATP synthase alpha




BL00152A 15.38 5.109e-14 128-154








and beta subunits proteins.






191




BL00152




ATP synthase alpha




BL00152B 21.40 4.273e-37 124-162








and beta subunits proteins.




BL00152A 15.38 8.364e-23 67-93






192




BL00152




ATP synthase alpha




BL00152B 21.40 2.000e-32 185-223








and beta subunits proteins.




BL00152A 15.38 8.364e-23 128-154






193




PR00493




BREAST CANCER TYPE I




PR00493G 7.57 1.184e-10 652-673








SUSCEPTIBILITY








PROTEIN SIGNATURE






195




BL00301




GTP-binding




BL00301A 12.41 1.750e-12 72-84








elongation factors proteins.






197




BL00745




Prokaryotic-type




BL00745C 13.66 7.398e-18 59-106








class I peptide








chain release








factors signat.






198




BL00745




Prokaryotic-type class I peptide




BL00745C 13.66 4.706e-12 59-106








chain release factors signat.






201




BL00660




Band 4.1 family




BL00660B 17.33 4.800e-27 136-176








domain proteins.




BL00660A 31.50 7.911e-20 52-105









BL00660C 23.36 2.241e-19 215-259









BL00660E 23.41 9.647e-13 301-324






205




PR00109




TYROSINE KINASE




PR00109B 12.27 1.882e-12 155-174








CATALYTIC DOMAIN








SIGNATURE






207




PR00837




ALLERGEN V5/TPX-1




PR00837C 17.21 4.064e-11 155-172








FAMILY SIGNATURE




PR00837A 14.77 4.960e-10 78-97









PR00837B 11.64 1.310e-09 133-147






208




BL01115




GTP-binding nuclear




BL01115A 10.22 8.909e-13 4-48








protein ran proteins.






211




BL00175




Phosphoglycerate




BL00175D 27.67 4.000e-40 367-419








mutase famiiy




BL00175C 23.75 6.870e-28 316-348








phosphohistidine




BL00175A 15.42 8.200e-19 252-272








proteins.




BL00175B 12.60 8.714e-17 299-312






212




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 1.000e-14 547-560








METAL-BINDI.




PD00066 13.92 2.200e-14 353-366









PD00066 13.92 3.400e-14 241-254









PD00066 13.92 6.400e-14 325-338









PD00066 13.92 1.500e-13 297-310









PD00066 13.92 6.500e-13 465-478









PD00066 13.92 7.500e-13 437-450









PD00066 13.92 8.500e-13 409-422









PD00066 13.92 2.714e-12 269-282









PD00066 13.92 3.571e-12 381-394









PD00066 13.92 7.577e-10 519-532






213




PD02331




CYCLIN CELL CYCLE




PD02331C 13.84 1.913e-11 9-36








DIVISION PROTE.






215




BL00239




Receptor tyrosine




BL00239B 25.15 3.915e-15 100-148








kinase class II proteins.






216




BL01013




Oxysterol-binding




BL01013D 26.81 9.135e-22 501-545








protein famiiy




BL01013A 25.14 4.600e-14 220-256








proteins.




BL01013C 9.97 4.906e-12 330-340









BL01013B 11.33 3.017e-11 287-298






219




BL00289




Pentaxin family protiens.




BL00289A 30.36 6.850e-26 25-56









BL00289E 18.00 6.684e-14 78-93






220




PR00217




43 KD POSTSYNAPTIC




PR00217C 10.91 7.527e-09 547-563








PROTEIN SIGNATRRE






221




PR00756




MEMBRANE ALANYL




PR00756D 10.58 1.529e-21 367-383








DIPEPTIDASE (M1)




PR00756B 14.06 5.737e-16 253-269








FAMILY SIGNATURE




PR00756A 12.90 1.237e-13 205-221









PR00756E 11.91 4.094e-13 386-399









PR00756C 11.60 6.108e-11 331-342






222




DM01688




2 POLY-IG RECEPTOR.




DM01688I 14.97 6.279e-09 75-123






224




PR00308




TYPE I ANTIFREEZE




PR00308C 3.83 2.523e-10 40-50








PROTEIN SIGNATURE




PR00308C 3.83 8.892e-10 41-51









PR00308C 3.83 8.892e-10 42-52









PR00308B 4.28 6.671e-09 40-52






231




BL00300




SRP54-type proteins




BL00300C 25.57 6.000e-09 215-269








GTP-binding domain proteins.






232




BL00514




Fibrinogen beta and gamma




BL00514C 17.41 9.463e-19 233-270








chains C-terminal domain




BL00514E 14.28 7.750e-12 293-310








proteins.




BL00514D 15.35 9.824e-11 274-287









BL00514G 15.98 4.273e-10 356-386









BL00514H 14.95 6.217e-09 391-416






233




BL01158




Macrophage migration




BL01158A 21.81 4.130e-30 2-47








inhibitory factor




BL01158B 17.07 4.316e-29 47-74








family proteins.






234




BL00456




Sodium: solute




BL00456A 22.59 6.250e-40 46-101








symporter family




BL00456C 24.55 6.586e-40 184-239








proteins.




BL00456B 18.94 8.125e-25 122-152









BL00456D 6.92 5.500e-10 476-486






237




PR00830




ENDOPEPTIDASE LA (LON)




PR00830A 8.41 4.780e-14 241-261








SERINE PROTEASE (S16)








SIGNATURE






238




PR00165




ANION EXCHANGER




PR00165I 10.02 8.412e-14 829-849








SIGNATURE




PR00165A 9.84 6.423e-13 495-518









PR00165B 15.26 9.090e-11 520-541









PR00165F 10.39 6.663e-10 639-658






239




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 7.943e-14 65-108






241




BL00115




Eukaryotic RNA polymerase II




BL00115Z 3.12 2.047e-10 469-518








heptapeptide repeat proteins.






244




BL01215




Mrp family proteins.




BL01215A 9.75 2.436e-09 466-493






245




PR00303




PREPROTEIN




PR00303G 10.45 8.759e-09 88-111








TRANSLOCASE SECY








SUBUNIT SIGNATURE






249




PF00429




ENV polyprotein




PF00429 31.08 8.015e-16 415-465








(coat polyprotein).






250




BL00415




Synapsins proteins.




BL00415N 4.29 7.115e-10 224-268






252




BL00183




Ubiquitin-conjugating




BL00183 28.97 4.326e-22 81-129








enzymes proteins.






254




BL00237




G-protein coupled




BL00237A 27.68 4.214e-16 108-148








receptors proteins.




BL00237C 13.19 3.323e-11 245-272









BL00237B 5.28 2.227e-09 182-194






255




BL00237




G-protein coupled




BL00237A 27.68 4.214e-16 108-148








receptors proteins.




BL00237C 13.19 3.323e-11 280-307









BL00237B 5.28 2.227e-09 217-229






259




PR00259




TRANSMEMBRANE FOUR




PR00259B 14.81 3.769e-21 50-77








FAMILY SIGNATURE




PR00259C 16.40 4.000e-20 77-106









PR00259A 9.27 3.600e-16 12-36






260




BL00282




Kazal serine protease inhibitors




BL00282 16.88 7.207e-14 562-585








family proteins.






261




PF00922




Vesiculovirus phosphoprotein.




PF00922A 19.17 7.724e-09 88-122






266




PR00049




WILM'S TUMOUR




PR00049D 0.00 7.143e-10 17-32








PROTEIN SIGNATURE






267




BL00612




Osteonectin domain proteins.




BL00612E 13.12 3.947e-11 379-424






268




BL00223




Annexins repeat proteins




BL00223A 15.59 1.000e-33 147-181








domain proteins.




BL00223A 15.59 1.435e-16 75-109









BL00223C 24.79 3.928e-15 134-189






269




BL00223




Annexins repeat proteins




BL00223B 28.47 1.000e-40 188-238








domain proteins.




BL00223A 15.59 1.000e-33 119-153









BL00223A 15.59 1.435e-16 47-81









BL00223C 24.79 3.928e-15 106-161






270




BL00223




Annexins repeat proteins




BL00223A 15.59 1.000e-33 119-153








domain proteins.




BL00223A 15.59 1.435e-16 47-81









BL00223C 24.79 3.928e-15 106-161






273




BL00086




Cytochrome P450 cysteine




BL00086 20.87 8.615e-27 423-455








heme-iron ligand proteins.






274




PR00385




P450 SUPERFAMILY




PR00385A 14.97 5.696e-13 295-313








SIGNATURE






275




BL00477




Alpha-2-macroglobulin




BL00477A 13.50 9.182e-19 70-99








family thiolester region








proteins.






277




BL00500




Thymosin beta-4 family




BL00500 9.77 2.565e-28 2-42








proteins.






279




PR00320




G-PROTEIN BETA WD-40




PR00320A 16.74 4.971e-10 231-246








REPEAT SIGNATURE




PR00320C 13.01 8.200e-10 231-246









PR00320B 12.19 9.486e-10 231-246









PR00320B 12.19 3.475e-09 188-203









PR00320B 12.19 4.600e-09 315-330









PR00320C 13.01 4.900e-09 315-330






288




PF00580




UvrD/REP helicase.




PR00580D 13.15 8.920e-13 670-684









PF00580E 13.89 2.800e-11 867-886









PF00580F 8.62 9.438e-10 913-926






289




PR00019




LEUCINE-RICH REPEAT




PR00019B 11.36 1.000e-09 64-78








SIGNATURE




PR00019A 11.19 8.000e-09 90-104






290




PD00126




PROTEIN REPEAT




PD00126A 22.53 5.500e-10 229-250








DOMAIN TPR NUCLEA.






295




DM01206




CORONAVIRUS




DM01206B 10.69 4.759e-09 464-484








NUCLEOCAPSID PROTEIN.






297




BL00972




Ubiquitin carboxyl-terminal




BL00972A 11.93 8.054e-15 191-209








hydrolases family 2 proteins.






298




PF00023




Ank repeat proteins.




PF00023A 16.03 9.500e-12 347-363









PF00023A 16.03 8.500e-10 283-299









PF00023A 16.03 8.875e-10 184-200






300




BL00415




Synapsins proteins.




BL00415Q 2.23 8.297e-09 13-49






302




BL01113




C1q domain proteins.




BL01113B 18.26 2.500e-13 841-877






304




BL01248




Laminin-type EGF-like




BL01248 11.02 7.171e-12 258-271








(LE) domain proteins.




BL01248 11.02 7.943e-12 325-338






305




BL00164




Enolase proteins.




BL00164A 11.58 2.800e-28 41-64






307




BL01153




NOL1/NOP2/sun family




BL01153D 19.69 8.322e-14 102-128








proteins.




BL01153C 13.67 6.507e-10 51-65






308




BL00892




HIT family proteins.




BL00892B 16.86 1.000e-20 130-154









BL00892A 18.17 6.657e-20 64-95






309




PF00791




Domain present in ZO-1 and




PF00791B 28.49 4.146e-10 73-128








Unc5-like netrin receptors.






310




BL00615




C-type lectin domain proteins.




BL00615B 12.25 5.200e-12 166-180






311




BL00071




Glyceraldehyde 3-phosphate




BL00071B 21.70 1.000e-40 80-126








dehydrogenase proteins.




BL00071C 11.81 1.000e-40 146-181









BL00071D 19.39 3.118e-25 184-239









BL00071E 11.48 4.600e-24 308-329









BL00071A 5.81 2.607e-14 5-17






314




PR00926




MITOCHONDRIAL




PR00926F 17.75 2.688e-10 15-38








CARRIER PROTEIN




PR00926D 10.53 6.625e-10 21-40








SIGNATURE






315




PR00654




ANGIOTENSINOGEN




PR00654A 15.64 1.540e-26 23-44








SIGNATURE




PR00654D 10.48 3.538e-26 153-175









PR00654F 15.16 8.071e-26 255-275









PR00654E 9.81 2.241e-25 194-215









PR006540 9.50 5.500e-21 115-135






316




BL00290




Immunoglobulins and major




BL00290A 20.89 8.071e-17 34-57








histocompatibility








complex proteins.






317




BL00290




Immunoglobulins and major




BL00290A 20.89 7.600e-16 34-57








histocompatibility








complex proteins.






318




BL00290




Immunoglobulins and major




BL00290A 20.89 2.800e-16 31-54








histocompatibility








complex proteins.






319




BL00290




Immunoglobulins and major




BL00290A 20.89 9.400e-16 34-57








histocompatibility








complex proteins.






320




BL00290




Immunoglobulins and major




BL00290B 13.17 4.000e-21 282-300








histocompatibility




BL00290A 20.89 4.600e-16 34-57








complex proteins.




BL00290A 20.89 2.421e-13 225-248






321




BL00290




Immunoglobulins and major




BL00290A 20.89 4.600e-16 34-57








histocompatibility








complex proteins.






323




BL00290




Immunoglobulins and major




BL00290A 20.89 8.071e-17 34-57








histocompatibility








complex proteins.






324




BL00290




Immunoglobulins and major




BL00290A 20.89 4.600e-16 34-57








histocompatibility








complex proteins.






325




BL00290




Immunoglobulins and major




BL00290A 20.89 4.600e-16 34-57








histocompatibility








complex proteins.






326




PF00651




BTB (also known as BR-C/Ttk)




PF00651 15.00 2.500e-14 46-59








domain proteins.






333




BL00972




Ubiquitin carboxyl-




BL00972A 11.93 3.919e-15 101-119








terminal hydrolases




BL00972B 9.45 7.577e-10 180-190








family 2 proteins.






337




PF00789




Domain present in




PF00789B 19.70 5.941e-09 213-234








ubiquitin-regulatory proteins.






338




PF00789




Domain present in




PF00789B 19.70 5.941e-09 259-280








ubiquitin-regulatory proteins.






339




PR00625




DNAJ PROTEIN FAMILY




PR00625A 12.84 3.000e-19 19-39








SIGNATURE




PR00625B 13.48 2.756e-17 47-68






340




BL00518




Zinc finger, C3HC4 type




BL00518 12.23 5.714e-10 24-33








(RING finger) proteins.






341




PR00500




POLYCYSTIC KIDNEY




PR00500I 9.22 1.107e-31 2810-2833








DISEASE PROTEIN




PR00500G 3.68 1.087e-30 2525-2548








SIGNATURE




PR00500H 17.80 1.107e-29 2662-2684









PR00500E 6.99 1.106e-27 2350-2370









PR00500F 9.44 1.108e-26 2483-2503






343




BL00415




Synapsins proteins.




BL00415N 4.29 5.401e-09 136-180






344




PD00930




PROTEIN GTPASE




PD00930B 33.72 2.800e-23 229-270








DOMAIN ACTIVATION.




PD00930A 25.62 5.021e-12 125-151






345




BL00023




Type II fibronectin




BL00023 24.31 8.043e-34 281-318








collagen-binding




BL00023 24.31 5.320e-32 223-260








domain proteins.




BL00023 24.31 5.800e-29 340-377






346




BL00023




Type II fibronectin




BL00023 24.31 8.043e-34 281-318








collagen-binding




BL00023 24.31 5.320e-32 223-260








domain proteins.




BL00023 24.31 5.800e-29 340-377






350




DM01354




kw TRANSCRIPTASE




DM01354R 8.50 2.969e-22 2115-2145








REVERSE II ORF2.




DM01354S 11.61 1.692e-14 2145-2166






351




BL00888




Cyclic nucleotide-




BL00888B 14.79 4.706e-18 372-396








binding domain proteins.




BL00868A 18.03 1.000e-08 354-371






353




PF00615




Regulator of G protein




PR00615B 16.25 9.625e-16 73-90








signalling domain proteins.




PR00615C 10.06 9.206e-12 150-164






354




BL00766




Tetrahydrofolate




BL00766E 13.78 9.625e-39 191-228








dehydrogenase/cyclohydrolase




BL00766C 25.86 4.375e-31 77-125








proteins.




BL00766D 17.05 5.966e-25 152-182






355




BL00180




Glutamine synthetase proteins.




BL00180E 17.60 1.000e-40 154-206









BL00180D 13.26 2.174e-24 119-141









BL00180F 10.05 6.211e-17 218-231









BL00180G 10.20 8.435e-17 307-322









BL00180C 12.14 4.600e-14 102-112









BL00180B 18.03 4.971e-14 68-87









BL00180A 13.20 5.065e-14 32-45






356




BL00180




Glutamine synthetase proteins.




BL00180F 10.05 6.750e-15 49-62






358




BL01131




Ribosomal RNA adenine




BL01131A 26.62 1.000e-08 77-123








dimethylases proteins.






360




DM00191




w SPAC8A4.04C




DM00191A 8.16 5.440e-09 36-49








RESISTANCE SPAC8A4.05C








DAUNORUBICIN.






361




DM00191




w SPAC8A4.04C




DM00191A 8.16 5.440e-09 61-74








RESISTANCE SPAC8A4.05C








DAUNORUBICIN.






362




PF00606




Herpesviral Glycoprotein B.




PF00606I 20.74 7.894e-09 264-316






363




PR00209




ALPHA/BETA GLIADIN




PR00209B 4.88 9.080e-11 80-99








FAMILY SIGNATURE




PR00209B 4.88 6.967e-10 86-105






364




PR00528




GLUCOCORTICOID




PR00528F 9.13 9.063e-09 31-51








RECEPTOR SIGNATURE






365




PF00622




Domain in SPla and the




PF00622C 12.62 6.625e-13 759-773








RYanodine Receptor.






366




BL00420




Speract receptor repeat




BL00420B 22.67 2.824e-25 37-92








proteins domain proteins.




BL00420C 11.90 9.250e-12 122-133






367




PR00080




ALCOHOL




PR00080A 9.32 8.548e-10 122-134








DEHYDROGENASE








SUPERFAMILY








SIGNATURE






369




BL00263




Natriuretic peptides proteins.




BL00263 11.87 5.909e-22 129-147






370




BL00609




Glycosyl hydrolases family




BL00609C 13.27 9.270e-11 249-261








32 proteins.






371




PR00237




RHODOPSIN-LIKE GPCR




PR00237E 13.03 4.000e-10 26-50








SUPERFAMILY








SIGNATURE






372




BL00125




Serine/threonine specific




BL00125D 33.11 9.719e-35 23-78








protein phosphatases proteins.






373




BL00453




FKBP-type peptidyl-prolyl




BL00453B 23.86 6.538e-26 281-315








cis-trans isomerase proteins.




BL00453A 15.57 8.364e-12 249-264









BL00453C 9.72 3.250e-11 323-336






374




PR00497




NEUTROPHIL CYTOSOL




PR00497A 6.92 8.261e-09 310-328








FACTOR P40 SIGNATURE






375




PR00449




TRANSFORMING PROTEIN




PR00449A 13.20 8.269e-16 34-56








P21 RAS SIGNATURE






376




PR00109




TYROSINE KINASE




PR00109B 12.27 9.847e-10 314-333








CATALYTIC DOMAIN








SIGNATURE






377




PR00109




TYROSINE KINASE




PR00109B 12.27 9.847e-10 314-333








CATALYTIC DOMAIN








SIGNATURE






378




BL00472




Small cytokines




BL00472C 20.76 8.225e-09 50-87








(intercrine/chemokine)








C—C subfamily signatur.






379




PR00704




CALPAIN CYSTEINE




PR007040 11.88 6.162e-09 132-149








PROTEASE (C2) FAMILY








SIGNATURE






380




PR00705




PAPAIN CYSTEINE




PR00705A 10.55 8.667e-13 155-171








PROTEASE (C1) FAMILY




PR00705B 10.22 2.385e-10 334-345








SIGNATURE






382




PD01270




RECEPTOR FC




PD01270A 17.22 7.443e-10 129-169








IMMUNOGLOBULIN




PD01270A 17.22 7.387e-09 36-76








AFFIN.






384




BL00412




Neuromodulin (GAP-43)




BL00412D 16.54 6.772e-10 250-301








proteins.






385




BL00232




Cadherins extracellular




BL00232B 32.79 8.594e-35 151-199








repeat proteins domain




BL00232B 32.79 5.579e-22 260-308








proteins.




BL00232A 27.72 1.000e-20 57-90









BL00232C 10.65 3.613e-14 258-276









BL00232B 32.79 4.872e-11 377-425









BL00232C 10.65 3.211e-09 480-498






387




BL00214




Cytosolic fatty-acid




BL00214B 26.51 9.000e-29 47-92








binding proteins.




BL00214A 21.17 1.000e-24 6-32






388




PR00452




SH3 DOMAIN SIGNATURE




PR00452B 11.65 8.250e-09 509-525






390




BL00972




Ubiquitin carboxyl-terminal




BL00972A 11.93 8.759e-17 112-130








hydrolases family 2 proteins.




BL00972D 22.55 8.116e-12 354-379









BL00972B 9.45 7.088e-09 193-203






392




BL00243




Integrins beta chain




BL00243I 31.77 3.155e-09 1-44








cysteine-rich domain proteins.






394




BL00476




Fatty acid desaturases family 1




BL00476F 12.75 6.551e-09 45-90








proteins.






397




PR00320




G-PROTEIN BETA WD-40




PR00320A 16.74 9.690e-11 292-307








REPEAT SIGNATURE




PR00320B 12.19 4.343e-10 292-307









PR00320C 13.01 7.840e-10 292-307






402




PD01823




PROTEIN INTERGENIC




PD01823D 16.66 3.093e-15 21-42








REGION ABC1 PRECURSOR




PD01823E 9.30 5.909e-15 75-88








MITOCHONDRION T.






404




BL00540




Ferritin iron-binding regions




BL00540A 15.06 1.000e-40 9-50








proteins.




BL00540B 18.82 1.000e-40 100-155









BL00540C 13.00 7.500e-15 165-177






405




PR00294




STREPTOMYCES




PR00294A 10.44 6.444e-10 159-186








SUBTILISIN INHIBITOR








SIGNATURE






408




BL00428




Cell cycle proteins




BL00428A 14.30 3.613e-09 91-110








ftsW/rodA/spoVE proteins.






411




BL00142




Neutral zinc metallopeptidases,




BL00142 8.38 7.188e-10 389-400








zinc-binding region proteins.






412




DM00191




w SPAC8A4.04C




DM00191D 13.94 6.330e-11 232-271








RESISTANCE




DM00191D 13.94 7.728e-11 48-87








SPAC8A4.05C




DM00191D 13.94 5.000e-10 112-151








DAUNORUBICIN.




DM00191D 13.94 5.667e-10 59-98









DM00191D 13.94 5.667e-10 123-162









DM00191D 13.94 6.583e-10 56-95









DM00191D 13.94 8.417e-10 280-319









DM00191D 13.94 8.917e-10 192-231









DM00191D 13.94 1.391e-09 224-263









DM00191D 13.94 2.409e-09 208-247









DM00191D 13.94 4.835e-09 120-159









DM00191D 13.94 5.304e-09 149-188









DM00191D 13.94 5.461e-09 211-250









DM00191D 13.94 6.322e-09 80-119









DM00191D 13.94 7.652e-09 243-282









DM00191D 13.94 8.513e-09 216-255









DM00191D 13.94 9.452e-09 177-216






415




PR00023




Ank repeat proteins.




PR00023A 16.03 1.321e-09 110-126






418




PF00168




C2 domain proteins.




PR00168C 27.49 9.250e-17 320-346






419




PF00168




C2 domain proteins.




PR00168C 27.49 9.250e-17 320-346






420




BL00086




Cytochrome P450 cysteine




BL00086 20.87 1.857e-20 444-476








heme-iron ligand proteins.






422




BL00218




Amino acid permeases




BL00218D 21.49 9.757e-11 263-308








proteins.






423




PR00049




WILM'S TUMOUR




PR00049D 0.00 3.288e-09 35-50








PROTEIN SIGNATURE






424




PR00380




KINESIN HEAVY CHAIN




PR00380A 14.18 4.086e-22 84-106








SIGNATURE




PR00380C 13.18 5.286e-17 240-259









PR00380D 9.93 7.698e-17 290-312









PR00380B 12.64 7.805e-14 207-225






425




PR00049




WILM'S TUMOUR




PR00049D 0.00 1.915e-09 590-605








PROTEIN SIGNATURE






426




BL00411




Kinesin motor domain




BL00411H 15.66 7.811e-22 79-110








proteins.




BL00411G 21.39 8.683e-22 31-73






428




BL00790




Receptor tyrosine kinase




BL00790E 29.58 6.667e-12 767-815








class V proteins.






429




BL00048




Protamine P1 proteins.




BL00048 6.39 4.038e-09 396-423






433




BL00107




Protein kinases ATP-binding




BL00107A 18.39 8.500e-27 342-373








region proteins.






434




PR00910




LUTEOVIRUS ORF6




PR00910A 2.51 6.036e-09 48-61








PROTEIN SIGNATURE






440




PR00019




LEUCINE-RICH REPEAT




PR00019A 11.19 7.261e-10 69-83








SIGNATURE




PR00019B 11.36 4.600e-09 66-80






444




BL00018




EF-hand calcium-binding




BL00018 7.41 6.870e-09 42-55








domain proteins.






448




BL00790




Receptor tyrosine




BL00790B 21.59 1.000e-40 61-113








kinase class V proteins.




BL00790C 16.65 1.000e-40 165-219









BL00790K 9.30 1.000e-40 657-711









BL00790Q 15.61 1.000e-40 855-904









BL00790O 7.68 5.929e-39 797-830









BL00790G 22.06 5.114e-36 376-420









BL00790R 16.20 7.469e-36 951-995









BL00790E 29.58 7.250e-35 273-321









BL00790J 14.21 8.200e-33 605-645









BL00790N 13.25 1.214e-31 763-790









BL00790I 20.01 1.931e-29 501-532









BL00790D 12.41 2.500e-27 243-268









BL00790H 13.42 6.478e-27 455-481









BL00790M 8.74 8.683e-25 741-763









BL00790P 12.33 3.755e-24 830-855









BL00790F 15.90 5.200e-24 339-366









BL00790L 11.16 5.909e-21 721-741









BL00790A 19.74 1.964e-19 31-53






453




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 2.000e-11 84-127






455




BL01160




Kinesin light chain




BL01160B 19.54 5.958e-09 387-441








repeat proteins.






456




BL01113




Clq domain proteins.




BL01113B 18.26 2.500e-13 841-877






458




BL01206




Amiloride-sensitive




BL01206D 30.58 3.025e-28 363-412








sodium channels proteins.




BL01206G 21.72 6.063e-27 530-576









BL01206F 16.40 7.643e-15 485-506









BL01206E 20.72 5.650e-14 427-454









BL01206C 12.30 3.455e-12 333-352









BL01206B 13.56 1.205e-10 313-327






460




BL01220




Phosphatidylethanolamine-




BL01220B 16.65 1.000e-40 59-100








binding protein




BL01220C 14.75 5.846e-34 100-128








family proteins.




BL01220A 22.62 3.400e-31 21-52






461




BL00815




Alpha-isopropylmalate and




BL00815C 21.36 3.118e-09 786-815








homocitrate synthases proteins.






464




PR00049




WILM'S TUMOUR




PR00049D 0.00 4.051e-09 1-16








PROTEIN SIGNATURE






470




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 5.200e-09 453-466








METAL-BINDI.






471




BL00022




EGF-like domain proteins.




BL00022A 7.48 5.000e-09 177-184









BL00022A 7.48 5.000e-09 241-248









BL00022A 7.48 8.000e-09 49-56






473




PR00371




FLAVOPROTEIN




PR00371D 14.55 4.536e-11 385-405








PYRIDINE NUCLEOTIDE








CYTOCHROME REDUCTASE








SIGNATURE






474




BL00028




Zinc finger, C2H2 type,




BL00028 16.07 4.462e-11 1087-1104








domain proteins.






475




PR00260




BACTERIAL CHEMOTAXIS




PR00260C 10.26 9.294e-09 146-167








SENSORY TRANSDUCER








SIGNATURE






476




BL00845




CAP-Gly domain proteins.




BL00845 16.43 6.442e-21 405-430









BL00845 16.43 9.820e-19 203-228






478




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 3.769e-15 369-382








METAL-BINDI.




PD00066 13.92 4.462e-15 285-298









PD00066 13.92 2.800e-14 257-270









PD00066 13.92 5.200e-14 313-326









PD00066 13.92 8.962e-10 341-354






481




PR00671




INHIBIN BETA B CHAIN




PR00671C 4.18 5.345e-09 9-29








SIGNATURE






483




DM01803




1 HERPESVIRUS




DM01803A 10.51 6.855e-09 215-236








GLYCOPROTEIN H.






484




DM01803




1 HERPESVIRUS




DM01803A 10.51 6.855e-09 251-272








GLYCOPROTEIN H.






486




PR00545




RETINOIC ACID




PR00545A 5.35 9.430e-09 383-398








RECEPTOR SIGNATURE






488




BL01238




GDA1/CD39 family of




BL01238A 11.72 7.840e-16 76-91








nucleoside phosphatases








proteins.






489




PD00930




PROTEIN GTPASE




PD00930B 33.72 2.800e-26 1256-1297








DOMAIN ACTIVATION.




PD00930A 25.62 3.864e-13 1152-1178






493




BL00223




Annexins repeat proteins




BL00223B 28.47 1.000e-40 140-190








domain proteins.




BL00223C 24.79 1.000e-40 217-272









BL00223A 15.59 5.500e-32 21-55









BL00223A 15.59 4.783e-14 230-264









BL00223C 24.79 2.515e-10 8-63









BL00223A 15.59 6.250e-10 71-105






496




DM01513




CAMP-DEPENDENT




DM01513A 13.61 8.568e-14 15-56








PROTEIN KINASE








REGULATORY CHAIN.






497




BL00603




Thymidine kinase




BL00603C 30.02 1.000e-40 152-207








cellular-type proteins.




BL00603A 20.71 4.500e-33 63-96









BL00603D 10.53 5.091e-18 217-232









BL00603B 11.39 3.455e-15 132-147






498




PD01922




PROTEIN




PD01922B 21.83 7.328e-14 162-198








PHOSPHODIESTERASE








HYDROL.






499




BL00061




Short-chain




BL00061B 25.79 1.931e-13 99-137








dehydrogenases/reductases








family proteins.






500




BL01160




Kinesin light chain




BL01160B 19.54 5.958e-09 64-118








repeat proteins.






501




PF00856




SET domain proteins.




PF00856A 26.14 8.579e-11 5-42






504




BL00030




Eukaryotic RNA-binding region




BL00030B 7.03 3.400e-10 116-126








RNP-1 proteins.






506




PF00075




RNase H.




PF00075D 10.71 7.000e-11 517-528









PF00075C 11.58 9.786e-11 484-496









PF00075B 12.56 4.073e-10 449-460









PF00075A 14.44 2.143e-09 402-419






508




BL00262




Insulin family proteins.




BL00262B 16.89 8.286e-17 68-88









BL00262A 12.48 4.600e-15 32-50






509




PR00213




MYELIN P0 PROTEIN




PR00213E 5.51 9.775e-12 264-289








SIGNATURE






511




BL00856




Guanylate kinase proteins.




BL00856C 29.21 2.658e-26 539-587









BL00856B 9.61 2.946e-18 511-532






513




PR00475




HEXOKINASE FAMILY




PR00475B 14.92 6.143e-26 186-212








SIGNATURE




PR00475E 16.08 2.742e-22 327-350









PR00475F 9.68 4.000e-20 407-430









PR00475A 14.06 3.118e-19 118-135









PR00475C 11.92 6.684e-19 239-256









PR00475G 9.08 1.692e-16 479-496









PR00475D 13.30 2.653e-13 262-277









PR00475G 9.08 2.650e-10 32-49






516




PR00722




CHYMOTRYPSIN SERINE




PR00722A 12.27 8.448e-14 56-72








PROTEASE FAMILY (S1)








SIGNATURE






518




BL00291




Prion protein.




BL00291A 4.49 9.379e-09 105-140






519




PF00534




Glycosyl transferases




PF00534B 14.47 9.581e-12 398-422








group 1.






520




PF01105




emp24/gp25L/p24 family.




PF01105B 25.12 2.868e-25 126-178






521




PF01105




emp24/gp25L/p24 family.




PF01105B 25.12 2.868e-25 151-203






522




PR00305




14-3-3 PROTEIN ZETA




PR00305A 9.33 9.500e-36 37-67








SIGNATURE




PR00305E 13.01 4.316e-32 177-204









PR00305D 16.34 3.647e-30 150-177









PR00305F 15.95 1.964e-26 204-234









PR00305C 8.68 3.182e-26 115-138









PR00305B 9.99 4.857e-24 84-109









PR00305F 15.95 8.975e-15 215-245






526




PF00642




Zinc finger C-x8-C-x5-C-x3-H




PF00642 11.59 7.796e-10 676-687








type (and similar).




PF00642 11.59 7.055e-09 276-287






527




PF00588




SpoU rRNA Methylase family.




PF00588B 17.18 8.200e-10 281-303






528




BL01160




Kinesin light chain




BL01160B 19.54 5.653e-09 791-845








repeat proteins.






529




PR00326




GTP1/OBG GTP-BINDING




PR00326A 8.75 4.255e-14 364-385








PROTEIN FAMILY








SIGNATURE






531




BL01305




moaA/nifB/pqqE family




BL01305D 14.97 7.279e-09 7-22








proteins.






532




PR00918




CALICIVIRUS NON-




PR00915A 13.76 5.807e-09 458-479








STRUCTURAL








POLYPROTEIN FAMILY








SIGNATURE






533




PR00171




SUGAR TRANSPORTER




PR00171E 14.87 1.000e-08 73-86








SIGNATURE






534




DM01418




352 FIBRILLAR




DM01418A 20.83 5.650e-23 101-149








COLLAGEN CARBOXYL-




DM01418B 22.51 8.500e-11 166-208








TERMINAL.




DM01418C 20.48 8.655e-10 236-278






535




DM01418




352 FIBRILLAR




DM01418A 20.83 5.650e-23 117-165








COLLAGEN CARBOXYL-




DM01418B 22.51 8.500e-11 182-224








TERMINAL.




DM01418C 20.48 8.655e-10 252-294






536




BL01052




Calponin family repeat




BL01052B 15.31 3.308e-11 30-56








proteins.






538




BL00795




Involucrin proteins.




BL00795C 17.06 7.600e-09 239-284






542




BL01046




ATP-dependent serine




BL01046D 19.61 4.938e-35 452-493








proteases, lon family,




BL01046C 17.03 9.581e-31 377-421








serine active sit.




BL01046B 19.24 4.977e-29 331-377






543




BL00824




Elongation factor 1




BL00824B 9.21 2.338e-09 150-170








beta/beta′/delta








chain proteins.






547




BL00615




C-type lectin domain proteins.




BL00615A 16.68 3.880e-11 47-65









BL00615B 12.25 2.286e-10 149-163






550




PR00119




P-TYPE CATION-




PR00119B 13.94 8.714e-12 35-50








TRANSPORTING ATPASE




PR00119E 8.48 7.716e-11 420-440








SUPERFAMILY








SIGNATURE






551




BL00039




DEAD-box subfamily




BL00039D 21.67 3.455e-33 476-522








ATP-dependent




BL00039A 18.44 8.548e-23 145-184








helicases proteins.




BL00039C 15.63 8.500e-16 277-301









BL00039B 19.19 1.837e-12 191-217






553




BL00115




Eukaryotic RNA polymerase II




BL00115Z 3.12 9.669e-09 116-165








heptapeptide repeat proteins.






554




PF00168




C2 domain proteins.




PF00168B 11.83 8.000e-10 38-49






555




PP00791




Domain present in




PF00791B 28.49 4.165e-13 780-835








ZO-1 and Unc5-like




PF00791B 28.49 6.767e-10 888-943








netrin receptors.




PF00791C 20.98 8.059e-09 794-833






556




PF00023




Ank repeat proteins.




PF00023A 16.03 5.875e-10 437-453









PF00023A 16.03 7.000e-10 563-579









PF00023A 16.03 8.500e-10 248-264









PF00023A 16.03 9.250e-10 95-111









PF00023A 16.03 3.250e-09 596-612









PF00023A 16.03 3.893e-09 716-732









PF00023A 16.03 6.786e-09 62-78









PF00023A 16.03 9.036e-09 496-512






557




BL00479




Phorbol esters/diacylglycerol




BL00479B 12.57 8.714e-09 143-159








binding domain proteins.






558




BL00479




Phorbol esters/diacylglycerol




BL00479B 12.57 8.714e-09 167-183








binding domain proteins.






559




BL00518




Zinc finger, C3HC4 type




BL00518 12.23 5.286e-10 239-248








(RING finger) proteins.






562




BL00122




Carboxylesterases type-B




BL00122G 11.67 2.500e-15 15-26








serine proteins.






563




PR00910




LUTEOVIRUS ORF6




PR00910A 2.51 1.986e-11 340-353








PROTEIN SIGNATURE




PR00910A 2.51 1.986e-11 342-355









PR00910A 2.51 1.986e-11 344-357









PR00910A 2.51 9.778e-10 346-359









PR00910A 2.51 1.107e-09 338-351









PR00910A 2.51 3.464e-09 336-349






564




DM01970




0 kw ZK632.12




DM01970B 8.60 8.475e-15 175-188








YDR313C ENDOSOMAL III.






566




PR00833




POLLEN ALLERGEN POA




PR00833H 2.30 8.375e-10 149-164








PI SIGNATURE




PR00833H 2.30 2.846e-09 147-162






567




PF00992




Troponin.




PF00992A 16.67 3.368e-09 448-483






573




PR00019




LEUCINE-RICH REPEAT




PR00019A 11.19 7.333e-09 322-336








SIGNATURE




PR00019B 11.36 9.280e-09 319-333






574




BL00315




Dehydrins proteins.




BL00315A 9.35 7.197e-10 93-121






576




PD00930




PROTEIN GTPASE




PD00930B 33.72 4.240e-16 235-276








DOMAIN ACTIVATION.






577




BL00183




Ubiquitin-conjugating




BL00183 28.97 8.338e-14 44-92








enzymes proteins.






579




PR00019




LEUCINE-RICH REPEAT




PR00019B 11.36 6.850e-10 132-146








SIGNATURE




PR00019A 11.19 2.667e-09 135-149









PR00019B 11.36 9.640e-09 180-194









PR00019B 11.36 1.000e-08 277-291






581




BL00625




Regulator of chromosome




BL00625A 16.21 2.033e-16 567-596








condensation (RCC1)




BL00625B 17.69 4.205e-12 561-595








proteins.




BL00625R 17.69 9.423e-11 93-127









BL00625B 17.69 1.444e-10 152-186









BL00625A 16.21 1.759e-10 99-128









BL00625A 16.21 2.739e-09 515-544









BL00625B 17.69 3.172e-09 43-77









BL00625A 16.21 4.170e-09 158-187






582




BL00414




Profilin proteins.




BL00414A 13.85 6.344e-13 2-16









BL00414E 15.46 6.283e-09 121-136






583




BL00414




Profilin proteins.




BL00414A 13.85 6.344e-13 2-16









BL00414E 15.46 6.283e-09 105-120






584




PD00301




PROTEIN REPEAT




PD00301A 10.24 8.200e-09 131-142








MUSCLE CALCIUM-BI.






585




DM01930




2 kw FINGER SMCX




DM01930F 14.16 1.310e-27 24-60








SMCY YDR096W.






586




BL00411




Kinesin motor domain proteins.




BL00411G 21.39 2.200e-39 77-119









BL00411H 15.66 8.800e-33 125-156









BL00411F 14.77 6.250e-18 33-58






588




BL00600




Aminotransferases class-III




BL00600E 16.43 5.725e-15 164-193








pyridoxal-phosphate




BL00600G 12.43 7.000e-14 242-261








attachment si.




BL00600F 8.77 7.480e-11 207-220









BL00600D 8.71 1.750e-10 143-157






589




BL00838




Interleukins −4 and −13




BL00838A 12.35 8.696e-09 136-155








proteins.






591




PR00121




SODIUM/POTASSIUM-




PR00121D 16.72 3.012e-12 261-283








TRANSPORTING ATPASE








SIGNATURE






592




BL00289




Pentaxin family proteins.




BL00289A 30.36 9.03le-09 331-362






594




BL01279




Protein-L-isoaspartate




BL01279A 24.27 1.000e-11 67-115








(D-aspartate)








O-methyltransferase signa.






597




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 4.462e-32 244-287






598




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 6.400e-16 245-258








METAL-BINDI.




PD00066 13.92 8.615e-15 329-342









PD00066 13.92 6.000e-13 301-314









PD00066 13.92 4.857e-12 217-230









PD00066 13.92 1.346e-10 273-286









PD00066 13.92 8.200e-09 357-370






599




BL00585




Ribosomal protein S5 proteins.




BL00585B 18.78 6.143e-18 303-340









BL00585A 28.43 4.286e-16 220-272






600




PR00482




OMPTIN SERINE




PR00482C 11.02 7.968e-09 816-842








PROTEASE SIGNATURE






601




PR00500




POLYCYSTIC KIDNEY




PR00500B 7.74 7.359e-10 56-77








DISEASE PROTEIN








SIGNATURE






603




PR00917




SMALL ROUND




PR00917G 10.59 8.990e-09 812-830








STRUCTURED VIRUS








(C37) CYSTEINE








PROTEASE FAMILY








SIGNATURE






605




BL00028




Zinc finger, C2H2 type,




BL00028 16.07 9.486e-09 109-126








domain proteins.






607




PF00638




RanBP1 domain proteins.




PR00638 11.91 4.600e-18 67-82






608




BL00406




Actins proteins.




BL00406E 8.44 8.541e-28 323-373









BL00406B 5.47 1.375e-27 82-137









BL00406D 12.58 3.160e-26 266-321









BL00406C 6.75 6.943e-25 141-196









BL00406A 9.95 2.575e-20 7-42






610




BL00048




Protamine P1 proteins.




BL0004B 6.39 3.700e-09 153-180






611




PR00315




GTP-BINDING




PR00315A 11.81 5.688e-10 126-140








ELONGATION FACTOR








SIGNATURE






615




PF00780




Domain found in NIK1-like




PR00780B 23.03 9.908e-09 14-57








kinases, mouse citron and








yeast ROM.






619




BL00162




Eukaryotic-type




BL00162C 17.78 1.000e-40 88-125








carbonic anhydrases




BL00162E 14.93 7.231e-39 171-204








proteins.




BL00162F 22.68 5.050e-31 208-242









BL00162A 22.92 8.714e-30 16-47









BL00162D 15.06 7.158e-24 126-151









BL00162B 21.43 1.375e-19 51-74






620




PR00457




ANIMAL HAEM




PR00457E 20.67 1.621e-24 414-441








PEROXIDASE SIGNATURE




PR00457D 16.81 8.258e-21 389-410









PR00457B 13.29 3.455e-18 223-239









PR00457G 17.45 7.000e-18 595-616









PR00457C 19.25 4.414e-16 371-390









PR00457H 15.90 8.650e-14 666-681









PR00457A 15.80 5.645e-12 169-181









PR00457F 13.69 8.875e-11 467-478






621




BL01304




ubiH/COQ6 monooxygenase




BL01304A 8.05 3.571e-11 50-64








family proteins.






623




BL00019




Actinin-type actin-binding




BL00019D 15.33 3.880e-17 145-175








domain proteins.






625




BL00893




mutT domain proteins.




BL00893 18.99 5.500e-16 127-152






626




PF00632




HECT-domain




PF00632B 18.45 7.000e-16 488-516








(ubiquitin-transferase).




PF00632C 20.66 7.851e-14 533-565






628




PR00239




MOLLUSCAN RHODOPSIN




PR00239E 1.58 9.566e-10 292-304








C-TERMINAL TAIL








SIGNATURE






634




BL01280




Glucose inhibited division




BL01280A 15.97 6.727e-36 69-110








protein A family proteins.




BL01280B 23.56 8.105e-27 128-180






638




PR00413




HALOACID




PR00413E 15.78 6.714e-09 70-87








DEHALOGENASE/EPOXIDE








HYDROLASE FAMILY








SIGNATURE






639




PD01861




PROTEIN NUCLEAR




PD01861A 14.06 6.318e-10 60-84








RIBONUCLEOPROTEIN








SMALL MRNA RNA.






640




PD00289




PROTEIN SH3 DOMAIN




PD00289 9.97 6.586e-09 46-60








REPEAT PRESYNA.






643




BL00914




Syntaxin/




BL00914 24.91 1.250e-29 184-234








epimorphin family proteins.






644




PR00010




TYPE II EGF-LIKE




PR00010C 11.16 6.667e-11 363-374








SIGNATURE






645




PR00625




DNAJ PROTEIN FAMILY




PR00625A 12.84 4.600e-20 14-34








SIGNATURE




PR00625B 13.48 8.759e-20 46-67






646




PD02448




TRANSCRIPTION




PD02448A 9.37 3.854e-09 351-390








PROTEIN DNA-BINDIN.






650




PD02446




TRANSCRIPTION




PD02448A 9.37 1.511e-20 50-89








PROTEIN DNA-BINDIN.




PD02448B 10.17 8.071e-19 89-137






654




PR00403




WW DOMAIN SIGNATURE




PR00403B 12.19 9.816e-11 144-159









PR00403B 12.19 8.167e-10 103-118






655




PR00403




WW DOMAIN SIGNATURE




PR00403B 12.19 9.816e-11 107-122






657




PR00929




AT-HOOK-LIKE DOMAIN




PR00929B 4.38 4.600e-10 358-370








SIGNATURE






658




PD02379




AMINOTRANSFERASE




PD02379E 11.43 1.000e-40 194-236








BIOSYNTHESIS




PD02379F 18.62 6.029e-35 245-284








PHOSPHOSERINE SER.




PD02379H 16.03 5.235e-33 352-385









PD02379B 12.05 3.613e-31 80-113









PD02379A 15.57 2.800e-25 29-60









PD023790 13.34 3.700e-21 119-139









PD02379D 11.83 9.419e-16 168-181









PD02379G 10.62 2.537e-14 313-328






659




PD02379




AMINOTRANSFERASE




PD02379E 11.43 1.000e-40 194-236








BIOSYNTHESIS




PD02379F 18.62 6.029e-35 245-284








PHOSPHOSERINE SER.




PD02379B 12.05 3.613e-31 80-113









PD02379A 15.57 2.800e-25 29-60









PD02379H 16.03 7.864e-23 306-339









PD023790 13.34 3.700e-21 119-139









PD02379D 11.83 9.419e-16 168-181






662




PR00874




FUNGI-IV




PR00874C 4.37 6.625e-09 33-48








METALLOTHIONEIN








SIGNATURE






666




BL00035




′POU′ domain proteins.




BL00035B 14.46 6.236e-09 683-704






668




PF00168




C2 domain proteins.




PF00168C 27.49 8.412e-13 634-660






669




PR00168




C2 domain proteins.




PR00168C 27.49 8.412e-13 115-141






671




PR00566




DOPAMINE 1B RECEPTOR




PR00566E 13.44 5.255e-18 466-483








SIGNATURE




PR00566A 9.32 3.000e-17 200-214









PR00566D 9.35 1.600e-12 446-455









PR00566C 11.44 2.184e-12 401-412









PR00566B 8.20 3.053e-11 341-351






672




PR00169




POTASSIUN CHANNEL




PR00169A 16.77 7.851e-11 46-66








SIGNATURE






673




DM00215




PROLINE-RICH PROTEIN 3.




DM00215 19.43 7.529e-11 183-216






674




BL00951




ER lumen protein retaining




BL00951B 14.23 1.670e-09 43-74








receptor proteins.






675




BL00292




Cyclins proteins.




BL00292B 20.31 3.925e-11 120-151






676




PR00048




C2H2-TYPE ZINC




PR00048A 10.52 3.160e-09 111-125








FINGER SIGNATURE






677




BL01226




Hydroxymethylglutary




BL01226I 25.06 8.560e-09 256-304








1-coenzyme A








synthase proteins.






681




BL00030




Eukaryotic RNA-binding region




BL00030A 14.39 1.563e-12 72-91








RNP-1 proteins.




BL00030A 14.39 2.125e-12 156-175






689




BL00740




MAM domain proteins.




BL00740B 19.76 3.813e-09 637-658






693




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 7.000e-11 93-136






698




PR00259




TRANSMEMBRANE FOUR




PR00259A 9.27 3.308e-18 19-43








FAMILY SIGNATURE




PR00259C 16.40 9.800e-18 88-117









PR00259D 13.50 2.756e-15 238-265






703




PR00669




INHIBIN ALPHA CHAIN




PR00669F 5.57 9.899e-09 223-241








SIGNATURE






704




DM01292




ESICULAR LUMEN




DM01292L 12.54 9.505e-09 240-265








DOMAIN.






705




PR00128




COLIPASE SIGNATURE




PR00128D 9.77 6.250e-25 47-66









PR00128C 9.28 5.299e-20 24-47






708




PR00049




WILM'S TUMOUR




PR00049D 0.00 9.929e-10 384-399








PROTEIN SIGNATURE






710




BL01118




Translation initiation




BL01118B 26.75 8.579e-26 94-132








factor SUI1 proteins.




BL01118A 12.46 4.000e-13 77-92






711




BL00811




Oleosins proteins.




BL00811A 8.26 3.310e-09 120-158






712




BL00674




AAA-protein family proteins.




BL00674B 4.46 9.182e-11 184-206






713




DM01871




kw SSR LIGASE




DM018710 20.79 9.836e-10 270-296








CYCLOFORMYL-








TETRAHYDROFOLATE.






715




PR00049




WILM'S TUMOUR




PR00049D 0.00 7.712e-09 95-110








PROTEIN SIGNATURE






717




BL01181




Ribosomal protein




BL01181 15.43 2.500e-10 13-49








S21 proteins.






718




PR00259




TRANSMEMBRANE FOUR




PR002590 16.40 6.824e-16 88-117








FAMILY SIGNATURE




PR00259A 9.27 3.423e-14 24-48









PR00259D 13.50 1.574e-13 238-265









PR00259B 14.81 8.714e-13 61-88






720




PR00496




NAPIN SIGNATURE




PR00496A 6.68 6.276e-09 21-43






723




PR00237




RHODOPSIN-LIKE GPCR




PR00237G 19.63 2.543e-11 670-697








SUPERFAMILY




PR00237A 11.48 3.000e-10 424-449








SIGNATURE






724




BL00972




Ubiquitin carboxyl-terminal




BL00972A 11.93 7.500e-20 36-54








terminal hydrolases




BL00972D 22.55 6.806e-16 296-321








family 2 proteins.




BL00972B 9.45 1.000e-13 116-126









BL00972E 20.72 8.773e-12 321-343






725




PF00646




F-box domain proteins.




PF00646A 14.37 6.906e-09 92-106






727




BL00933




FGGY family of




BL00933D 24.01 7.545e-15 212-249








carbohydrate kinases




BL00933B 15.94 2.200e-09 54-65








proteins.




BL00933E 13.80 3.543e-09 439-455









BL00933A 17.50 4.857e-09 20-44






728




PR00876




NEMATODE




PR00876B 7.66 1.887e-10 137-151








METALLOTHIONEIN








SIGNATURE






731




PD01976




KINASE DEHYDROGENASE




PD01976A 8.95 1.493e-09 83-96








TRANSFERASE.






732




BL00623




GMC oxidoreductases proteins.




BL00623A 12.60 9.859e-10 12-31






735




BL01172




Ribosomal protein




BL01172B 14.10 8.909e-38 15-57








L44e proteins.




BL01172C 16.78 7.188e-31 63-102






737




DM01724




kw ALLERGEN POLLEN




DM01724 8.14 5.909e-11 11-31








CIM1 HOL-LI.




DM01724 8.14 6.591e-11 41-61









DM01724 8.14 6.831e-10 39-59









DM01724 8.14 8.697e-09 55-75






738




PR00320




G-PROTEIN BETA WD-40




PR00320A 16.74 8.463e-09 73-88








REPEAT SIGNATURE






742




PD02269




CYTIDINE DEAMINASE




PD022690 16.36 7.882e-17 79-92








HYDROLASE ZINC




PD02269A 10.06 1.000e-15 29-41








AMINOHY.




PD02269D 11.98 5.000e-14 110-125






743




BL00790




Receptor tyrosine




BL00790D 12.41 8.297e-09 429-454








kinase class V proteins.






744




BL00750




Chaperonins TCP-1 proteins.




BL00750B 16.17 2.000e-39 69-119









BL00750A 20.07 8.286e-36 25-68









BL00750C 25.65 8.579e-23 152-184






746




BL00415




Synapsins proteins.




BL00415N 4.29 4.710e-10 225-269






748




BL00028




Zinc finger, C2H2 type




BL00028 16.07 1.000e-09 212-229








domain proteins.




BL00028 16.07 6.143e-09 365-382






752




BL00030




Eukaryotic RNA-binding region




BL00030A 14.39 6.143e-13 332-351








RNP-1 proteins.






753




PF00023




Ank repeat proteins.




PR00023A 16.03 8.500e-10 283-299









PF00023A 16.03 9.625e-10 347-363









PF00023A 16.03 1.321e-09 184-200









PF00023A 16.03 1.643e-09 150-166






754




BL00107




Protein kinases ATP-binding




BL00107B 13.31 1.643e-10 202-218








region proteins.






760




PR00671




INHIBIN BETA B CHAIN




PR006710 4.18 8.966e-09 212-232








SIGNATURE






761




PR00678




P13 KINASE P85




PR00678H 9.13 7.805e-12 292-315








REGULATORY SUBUNIT








SIGNATURE






762




PR00412




EPOXIDE HYDROLASE




PR00412C 11.30 2.421e-12 169-183








SIGNATURE




PR00412A 13.23 7.947e-12 104-123









PR00412B 12.59 7.429e-10 123-139






763




PR00217




43 KD POSTSYNAPTIC




PR00217C 10.91 7.247e-10 293-309








PROTEIN SIGNATURE






764




PR00320




G-PROTEIN BETA WD-40




PR00320A 16.74 9.122e-09 277-292








REPEAT SIGNATURE




PR00320A 16.74 9.780e-09 233-248









PR00320C 13.01 1.000e-08 233-248






765




BL01230




RNA methyltransferase




BL01230E 15.79 2.918e-11 487-503








trmA family proteins.






771




BL00039




DEAD-box subfamily




BL00039D 21.67 2.957e-09 434-480








ATP-dependent








helicases proteins.






773




PR00453




VON WILLEBRAND




PR00453A 12.79 2.957e-10 33-51








FACTOR TYPE A DOMAIN








SIGNATURE






777




PR00493




BREAST CANCER TYPE I




PR00493G 7.57 3.711e-14 693-714








SUSCEPTIBILITY








PROTEIN SIGNATURE






778




BL00443




Glutamine amidotransferases




BL00443F 16.68 8.714e-09 85-101








class-II proteins.






780




DM01206




CORONAVIRUS




DM01206B 10.69 7.288e-10 167-187








NUCLEOCAPSID








PROTEIN.






783




BL01221




PMP-22/EMP/MP20




BL012210 26.20 1.281e-34 59-104








family proteins.




BL01221D 13.99 5.966e-27 136-163









BL01221A 17.26 2.385e-26 1-29









BL01221B 13.29 1.000e-14 38-52






785




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 4.000e-10 297-340






786




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 4.000e-10 297-340






790




BL00303




S-100/ICaBP type calcium




BL00303B 26.15 5.075e-13 73-110








binding protein.






792




PD01941




TRANSMEMBRANE




PD01941C 19.96 4.960e-16 84-139








COTRANSPORTER SYMP.




PD01941B 15.02 2.093e-11 4-51






794




BL00672




Serine proteases, V8 family




BL00672B 9.84 3.554e-09 214-231








histidine proteins.






797




BL00674




AAA-protein family proteins.




BL00674B 4.46 7.814e-10 360-382






798




BL00674




AAA-protein family proteins.




BL00674B 4.46 7.814e-10 360-382






799




BL00215




Mitochondrial energy




BL00215A 15.82 9.591e-16 206-231








transfer proteins.




BL00215A 15.82 4.000e-15 104-129









BL00215A 15.82 9.400e-15 7-32









BL00215B 10.44 1.000e-10 154-167






801




BL01013




Oxysterol-binding protein




BL01013A 25.14 5.500e-21 537-573








family proteins.




BL01013D 26.81 2.16le-18 807-851









BL01013C 9.97 4.231e-13 625-635









BL01013B 11.33 3.017e-11 603-614






803




BL00711




Lipoxygenases iron-binding




BL00711I 18.56 8.630e-28 577-615








binding region proteins.




BL00711E 19.66 3.550e-22 414-451









BL00711G 21.83 9.100e-22 503-535









BL00711C 20.75 5.959e-19 268-297









BL00711D 17.56 1.923e-16 347-373









BL00711H 23.34 1.771e-12 535-574









BL00711F 19.79 2.086e-10 484-501






805




PR00492




RHO PROTEIN GDP




PR00492C 9.68 1.900e-23 122-139








DISSOCIATION




PR00492B 9.77 8.579e-23 76-95








INHIBITOR SIGNATURE




PR004920 14.82 8.200e-21 139-155









PR00492A 11.92 1.643e-18 60-76






808




BL00378




Hexokinases proteins.




BL00378A 19.01 8.500e-09 403-431






809




BL00027




′Homeobox′ domain proteins.




BL00027 26.43 8.615e-33 35-78






810




PR00179




LIPOCALIN SIGNATURE




PR00179B 9.56 1.000e-12 102-115









PR001790 19.02 1.000e-10 130-146









PR00179A 13.78 5.680e-10 37-50






811




BL00290




Immunoglobulins and major




BL00290A 20.89 1.818e-11 164-187








histocompatibility








complex proteins.






814




BL00269




Mammalian defensins proteins.




BL002690 16.52 7.158e-09 171-200






815




BL00216




Sugar transport proteins.




BL00216B 27.64 5.846e-09 141-191






818




BL00456




Sodium:solute symporter




BL00456A 22.59 2.080e-30 83-138








family proteins.




BL00456C 24.55 3.721e-29 221-276









BL00456B 18.94 1.000e-22 159-189






819




BL00142




Neutral zinc metallopeptidases,




BL00142 8.38 1.857e-09 494-505








zinc-binding region proteins.






820




BL00509




Ras GTPase-activating




BL00509B 10.28 1.643e-12 610-621








proteins.






824




PR00048




C2H2-TYPE ZINC




PR00048A 10.52 6.143e-13 252-266








FINGER SIGNATURE




PR00048A 10.52 7.429e-13 476-490









PR00048A 10.52 3.118e-12 336-350









PR00048A 10.52 3.118e-12 364-378









PR00048A 10.52 4.706e-12 504-518









PR00048A 10.52 8.412e-12 224-238









PR00048A 10.52 3.842e-11 392-406









PR00046A 10.52 6.211e-11 308-322









PR00048A 10.52 6.211e-11 448-462









PR00048B 6.02 7.231e-11 492-502









PR00048B 6.02 3.250e-10 240-250









PR00048A 10.52 6.870e-10 420-434









PR00048B 6.02 2.421e-09 380-390






825




PR00122




VACUOLAR ATP




PR00122D 9.97 7.214e-11 103-127








SYNTRASE 16 KD




PR00122C 8.20 9.526e-10 76-103








SUBUNIT SIGNATURE






826




BL00518




Zinc finger, C3HC4 type




BL00518 12.23 6.571e-10 30-39








(RING finger), proteins.






828




BL01160




Kinesin light chain




BL01160B 19.54 1.610e-09 33-87








repeat proteins.




BL01160B 19.54 9.619e-09 65-119






835




PD02411




PROTEIN




PD02411 21.89 6.786e-15 3967-4001








TRANSCRIPTION








REGULATION NUCLEAR.






836




DM01970




0 kw ZK632.12




DM01970B 8.60 9.423e-10 111-124








YDR313C ENDOSOMAL III.






837




PR00048




C2H2-TYPE ZINC




PR00048A 10.52 2.174e-10 177-191








FINGER SIGNATURE






842




PF00922




Vesiculovirus phosphoprotein.




PF00922A 19.17 7.724e-09 276-310






844




PD02059




CORE POLYPROTEIN




PD02059A 28.10 5.950e-10 34-75








PROTEIN GAG








CONTAINS: P.






846




BL00326




Tropomyosins proteins.




BL00326D 8.76 8.065e-09 165-206






847




BL00326




Tropomyosins proteins.




BL00326D 8.76 8.065e-09 173-214






849




PR00563




BETA-3 ADRENERGIC




PR00563B 3.98 8.141e-09 8-28








RECEPTOR SIGNATURE






851




PR00450




RECOVERIN FAMILY




PR00450C 12.22 1.570e-09 285-307








SIGNATURE






853




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 8.800e-14 290-303








METAL-BINDI.




PD00066 13.92 4.000e-13 234-247









PD00066 13.92 4.429e-12 262-275









PD00066 13.92 9.217e-11 206-219









PD00066 13.92 3.769e-10 505-518









PD00066 13.92 4.115e-10 449-462









PD00066 13.92 4.462e-10 533-546









PD00066 13.92 6.538e-10 477-490






854




BL00615




C-type lectin domain proteins.




BL00615A 16.68 8.920e-11 137-155






855




BL00615




C-type lectin domain proteins.




BL00615A 16.68 8.920e-11 176-194






856




BL00018




EF-hand calcium-binding




BL00018 7.41 9.000e-14 65-78








domain proteins.






858




PR00019




LEUCINE-RICH REPEAT




PR00019B 11.36 1.000e-09 219-233








SIGNATURE






860




PD02474




SYNTHASE SMALL




PD02474B 21.08 8.568e-09 199-238








SUBUNIT ACETOLACT.






861




PF00922




Vesiculovirus phosphoprotein.




PF00922A 19.17 1.000e-08 249-283






864




PR00289




DISINTEGRIN




PR00289B 11.79 1.947e-09 522-535








SIGNATURE






866




PF00242




DNA polymerase (viral)




PF00242F 12.18 8.522e-09 197-219








N-terminal domain proteins.






867




PR00780




LEUSERPIN 2




PR00780B 4.89 4.491e-09 262-285








SIGNATURE






868




BL00226




Intermediate filaments proteins.




BL00226D 19.10 8.027e-13 208-255






869




PD01876




ANTIGEN MELANOMA-




PD01876C 21.73 3.326e-15 461-534








ASSOCIATED MULTIGENE




PD01876C 21.73 3.045e-10 735-788








FAMILY TUM.






870




PR00747




GLYCOSYL HYDROLASE




PR00747C 12.06 8.767e-09 337-356








FAMILY 47 SIGNATURE






872




DM01782




HYDROGENASE (FE)




DM01782C 13.88 4.400e-19 349-368








LARGE CHAIN.




DM01782F 9.01 4.375e-18 499-515









DM01782B 17.29 3.412e-10 294-327






873




BL00226




Intermediate filaments proteins.




BL00226D 19.10 7.375e-38 321-368









BL00226B 23.86 7.107e-32 155-203









BL00226C 13.23 3.100e-19 220-251









BL00226A 12.77 7.000e-15 55-70









BL00226D 19.10 7.800e-09 254-301






874




DM01415




6 SALIVARY GLUE




DM01415B 13.78 9.518e-10 4-52








PROTEIN.






876




PR00860




VERTEBRATE




PR00860B 7.04 2.929e-20 74-88








METALLOTHIONEIN




PR00860A 5.46 5.655e-13 52-65








SIGNATURE




PR00860C 9.61 2.400e-12 88-98






877




PR00360




C2 DOMAIN SIGNATURE




PR00360B 13.61 7.136e-09 572-586






881




DM01206




CORONAVIRUS




DM01206B 10.69 8.767e-10 567-587








NUCLEOCAPSID




DM01206B 10.69 1.000e-09 563-583








PROTEIN.






882




BL00092




N-6 Adenine-specific DNA




BL00092 5.35 2.000e-09 136-145








methylases proteins.






883




PR00511




TEKTIN SIGNATURE




PR00511A 13.59 3.700e-14 113-130






885




PR00764




COMPLEMENT C9




PR00764F 16.69 2.286e-09 158-179








SIGNATURE






887




BL01279




Protein-L-isoaspartate




BL01279A 24.27 3.691e-09 419-467








(D-aspartate)








O-methyltransferase signa.






889




PD01719




PRECURSOR




PD01719A 12.89 2.603e-11 259-287








GLYCOPROTEIN SIGNAL




PD01719A 12.89 8.105e-10 199-227








RE.






890




BL01162




Quinone oxidoreductase/




BL01162C 22.80 1.269e-18 151-195








zeta-crystallin proteins.




BL01162A 15.38 1.265e-11 64-87






893




PD01066




PROTEIN ZINC FINGER




PD01066 19.43 5.415e-26 46-85








ZINC-FINGER METAL-








BINDING NU.






894




PD02910




TRANSCRIPTION




PD02910A 15.43 9.839e-09 62-97








PROTEIN FACTOR








REGULATION A.






895




PD02199




SUBUNIT HYDROGEN ION




PD02199A 20.58 1.000e-40 10-61








TRANSPORT T.




PD02199D 13.18 1.000e-40 364-405









PD02199F 15.02 1.000e-40 440-482









PD02199J 11.42 1.000e-40 723-762









PD02199K 15.22 1.000e-40 792-831









PD02199G 9.43 4.447e-24 531-555









PD02199B 27.90 1.474e-22 263-306









PD02199H 13.62 2.636e-21 576-599









PD02199E 7.56 8.642e-19 405-424









PD02199C 17.60 8.085e-14 313-329









PD02199I 8.90 4.780e-09 616-624






896




BL00218




Amino acid permeases proteins.




BL00218E 23.30 5.920e-10 343-383






897




BL00048




Protamine P1 proteins.




BL00048 6.39 9.526e-10 160-187






900




BL00811




Oleosins proteins.




BL00811B 10.57 9.791e-09 307-336






904




BL00415




Synapsins proteins.




BL00415N 4.29 4.153e-09 301-345






905




BL00107




Protein kinases ATP-binding




BL00107A 18.39 3.250e-17 133-164








region proteins.






906




PR00449




TRANSFORMING PROTEIN




PR00449A 13.20 4.971e-14 4-26








P21 RAS SIGNATURE






908




BL00317




WAP-type ‘four-disulfide core’




BL00317B 14.58 3.550e-13 48-70








domain proteins.






909




BL01019




ADP-ribosylation factors




BL01019B 19.49 7.517e-21 95-150








family proteins.






910




PD00066




PROTEIN ZINC-FINGER




PD00066 13.92 2.385e-15 128-141








METAL-BINDI.




PD00066 13.92 5.714e-12 100-113






911




PR00048




C2H2-TYPE ZINC




PR00045A 10.52 1.000e-11 174-188








FINGER SIGNATURE




PR00048B 6.02 1.692e-11 162-172






912




PF00651




BTB (also known as BR-C/Ttk)




PR00651 15.00 2.895e-11 45-58








domain proteins.






913




DM00547




1 kw CHROMO




DM00547F 23.43 7.643e-34 606-653








BROMODOMAIN SHADOW




DM00547B 11.28 7.907e-16 155-169








GLOBAL.




DM00547C 17.30 8.650e-14 209-231









DM00547D 11.60 6.500e-13 277-291









DM00547E 13.94 1.000e-11 307-330






914




BL01115




GTP-binding nuclear protein




BL01115A 10.22 5.330e-11 18-62








ran proteins.






919




BL01283




T-box domain proteins.




BL01283D 11.70 7.868e-31 59-92









BL01283C 13.05 2.537e-14 25-39






922




DM01503




1 HERPESVIRUS




DM01803A 10.51 8.699e-09 100-121








GLYCOPROTEIN H.






924




BL00470




Isocitrate and isopropylmalate




BL00470A 16.25 5.179e-14 10-31








dehydrogenases proteins.




BL00470C 15.43 4.103e-10 223-238









BL00470E 16.52 1.900e-09 287-297






925




PF00023




Ank repeat proteins.




PF00023A 16.03 3.893e-09 44-60









PF00023B 14.20 9.182e-09 40-50






926




PF00023




Ank repeat proteins.




PF00023A 16.03 3.893e-09 72-88









PF00023B 14.20 9.182e-09 68-78






929




PD01066




PROTEIN ZINC FINGER




PD01066 19.43 2.019e-26 51-90








ZINC-FINGER METAL-








BINDING NU.






930




PR00830




ENDOPEPTIDASE LA




PR00830A 8.41 4.927e-13 222-242








(LON) SERINE








PROTEASE (S16)








SIGNATURE






931




BL00456




Sodium:solute symporter




BL00456A 22.59 1.957e-32 35-90








family proteins.




BL00456B 18.94 9.780e-17 111-141






932




BL00456




Sodium:solute symporter




BL00456A 22.59 1.957e-32 35-90








family proteins.




BL00456C 24.55 1.225e-31 173-228









BL00456B 18.94 9.780e-17 111-141






936




BL00290




Immunoglobulins and major




BL00290A 20.89 1.818e-11 159-182








histocompatibility








complex proteins.






937




PR00830




ENDOPEPTIDASE LA




PR0083CA 8.41 5.897e-10 352-372








(LON) SERINE








PROTEASE (S16)








SIGNATURE






938




PF00651




BTB (also known as BR-C/TtK)




PR00651 15.00 7.000e-10 50-63








domain proteins.






939




PD00306




PROTEIN GLYCOPROTEIN




PD00306A 10.26 6.625e-13 544-558








PRECURSOR RE.






940




PD00306




PROTEIN GLYCOPROTEIN




PD00306A 10.26 6.625e-13 544-558








PRECURSOR RE.






941




PD00306




PROTEIN GLYCOPROTEIN




PD00306A 10.26 6.625e-13 544-558








PRECURSOR RE.






942




PF00938




Lipoprotein.




PF00938E 19.50 6.096e-09 272-307






943




PF00925




GTP cyclohydrolase II.




PF00925F 13.23 9.850e-09 356-367






945




BL00226




Intermediate filaments proteins.




BL00226A 12.77 5.355e-13 139-154






947




PF00035




Double-stranded RNA




PF00035B 12.06 7.750e-09 273-287








binding motif.






948




PF00622




Domain in SPla and the




PF00622B 21.00 9.250e-11 170-192








RYanodine Receptor.











*Results include in order: accession number subtype; raw score; p-value; position of signature in amino acid sequence.

























TABLE 4









SEQ










ID NO:




pFAM NAME




DESCRIPTION




p-value




pFAM SCORE



























2




UCH-2




Ubiquitin carboxyl-terminal hydrolase family 2




5.9e-26




99.7






5




SH2




Src homology domain 2




8.5e-22




66.5






6




SH2




Src homology domain 2




8.5e-22




66.5






7




RCC1




Regulator of chromosome condensation (RCC1)




4.4e-18




68.4






14




UQ_con




Ubiquitin-conjugating enzyme




3.2e-49




176.9






15




UQ_con




Ubiquitin-conjugating enzyme




1.2e-68




241.5






20




gntR




Bacterial regulatory proteins, gntR family




0.062




11.4






21




gntR




Bacterial regulatory proteins, gntR family




0.062




11.4






23




kinesin




Kinesin motor domain




2.9e-128




439.5






27




Kelch




Kelch motif




7.9e-71




248.7






29




kinesin




Kinesin motor domain




1.9e-171




583.0






30




pkinase




Eukaryotic protein kinase domain




8.5e-09




35.7






32




Aa_trans




Transmembrane amino acid transporter protein




3.1e-53




190.3






34




EGF




EGF-like domain




3.9e-24




93.6






38




ig




Immunoglobulin domain




6.2e-06




24.2






39




LRR




Leucine Rich Repeat




1.1e-10




48.9






43




CK_II_beta




Casein kinase II regulatory subunit




4.5e-69




242.9






45




COX6B




Cytochrome oxidase c subunit VIb




0.064




−6.7






46




RNA_pol_B




RNA polymerase beta subunit




0




1206.8






47




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




  2e-07




28.3






48




HLH




Helix-loop-helix DNA-binding domain




1.5e-07




38.5






49




ras




Ras family




3.3e-12




2.0






50




pkinase




Eukaryotic protein kinase domain




1.9e-43




157.8






52




ig




Immunoglobulin domain




7.2e-08




30.4






54




PX




PX domain




3.7e-06




33.9






65




Defensin_propep




Defensin propeptide




  3e-25




97.3






67




zf-C2H2




Zinc finger, C2H2 type




  2e-55




197.5






71




pentaxin




Pentaxin family




3.4e-18




66.3






73




SSF




Sodium:solute symporter family




1.7e-05




−65.8






75




AMP-binding




AMP-binding enzyme




1.1e-12




−49.0






78




transmembrane4




Transmembrane 4 family




6.4e-05




18.7






80




zf-C2H2




Zinc finger, C2H2 type




4.9e-30




113.2






81




fn3




Fibronectin type III domain




1.8e-13




58.2






83




polyprenyl_synt




Polyprenyl synthetases




0.015




−83.6






84




Defensin_propep




Defensin propeptide




  3e-25




97.3






85




MAGE




MAGE family




3.5e-34




127.0






86




LRR




Leucine Rich Repeat




7.7e-15




62.7






87




Cytidylyltransf




Cytidylyltransferase




1.4e-05




29.3






90




lipase




Lipase




2.5e-15




55.2






96




homeobox




Homeobox domain




1.1e-30




115.3






101




fn3




Fibronectin type III domain




4.7e-78




272.7






102




ig




Immunoglobulin domain




0.00045




18.2






103




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




6.7e-08




32.8






104




Trans_recep




Transient receptor




1.9e-34




115.7






105




Tropomyosin




Tropomyosins




0.0086




11.3






109




p450




Cytochrome P450




1.8e-61




217.6






116




Tropomyosin




Tropomyosins




0.0086




11.3






117




Tropomyosin




Tropomyosins




0.0086




11.3






118




aa_permeases




Amino acid permease




3.2e-06




−173.3






120




zf-C2H2




Zinc finger, C2H2 type




1.3e-124




427.4






122




C2




C2 domain




1.1e-38




142.0






123




ig




Immunoglobulin domain




0.00079




17.4






127




WD40




WD domain, G-beta repeat




9.6e-15




62.4






130




FYVE




FYVE zinc finger




7.4e-23




86.0






131




PH




PH domain




3.9e-25




94.8






133




KRAB




KRAB box




1.6e-24




94.9






134




Ribosomal_L11




Ribosomal protein L11




3.9e-64




226.5






136




zf-C2H2




Zinc finger, C2H2 type




2.1e-185




629.4






137




Band_7




SPFH domain/Band 7 family




6.5e-35




129.4






139




TPR




TPR Domain




5.3e-16




66.6






142




WH1




WH1 domain




6.4e-05




29.8






143




zf-DHHC




DHHC zinc finger domain




0.033




−11.5






147




zf-C2H2




Zinc finger, C2H2 type




9.2e-82




285.0






149




7tm_2




7 transmembrane receptor (Secretin family)




1.2e-22




88.7






151




lectin_c




Lectin C-type domain




0.0097




3.8






152




PDZ




PDZ domain (Also known as DHR or GLGF).




0.0031




24.2






153




cadherin




Cadherin domain




3.7e-95




329.6






155




COesterase




Carboxylesterases




  7e-48




166.8






156




DSPc




Dual specificity phosphatase, catalytic domain




2.8e-29




110.7






157




efhand




EF hand




2.2e-14




61.2






159




A2M




Alpha-2-macroglobulin family




2.2e-07




25.5






160




zf-C2H2




Zinc finger, C2H2 type




8.8e-68




238.6






161




Nucleoside_tra2




Na+ dependent nucleoside transporter




8e-188




637.4






166




ras




Ras family




5.5e-31




116.4






167




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




3.2e-06




24.4






168




LRR




Leucine Rich Repeat




1.3e-06




35.4






169




RasGAP




GTPase-activator protein for Ras-like GTPase




6.4e-28




106.2






172




LMWPc




Low molecular weight phosphotyrosine protein phosphatase




9.7e-56




198.6






175




lectin_c




Lectin C-type domain




5.4e-06




33.3






178




PK




Pyruvate kinase




5.4e-12




46.3






179




vwa




von Willebrand factor type A domain




4.1e-63




223.1






180




neur_chan




Neurotransmitter-gated ion-channel




1.5e-120




413.9






181




C4




C-terminal tandem repeated domain in type 4 procollagen




1.4e-148




507.0






187




zf-C2H2




Zinc finger, C2H2 type




1.2e-25




98.6






188




Ribosomal_S2




Ribosomal protein S2




3.3e-79




276.5






191




ATP-synt_ab




ATP synthase alpha/beta family




5.7e-09




−1.3






192




ATP-synt_ab




ATP synthase alpha/beta family




3.1e-11




49.7






193




ank




Ank repeat




3.5e-33




123.7






195




GTP_EFTU




Elongation factor Tu family




8.1e-33




113.3






197




RF-1




Peptidyl-tRNA hydrolase domain




0.00034




10.7






200




WD40




WD domain, G-beta repeat




  1e-05




32.4






201




Band_41




FERM domain (Band 4.1 family)




5.3e-86




269.7






205




pkinase




Eukaryotic protein kinase domain




1.6e-90




314.1






207




SCP




SCP-like extracellular protein




  1e-15




60.4






208




ras




Ras family




1.5e-15




40.0






211




6PF2K




6-phosphofructo-2-kinase




5.2e-152




518.4






212




zf-C2H2




Zinc finger, C2H2 type




  3e-104




359.8






215




pkinase




Eukaryotic protein kinase domain




3.2e-92




319.8






216




Oxysterol_BP




Oxysterol-binding protein




4.3e-48




173.2






219




pentaxin




Pentaxin family




2.6e-40




142.7






220




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




6.2e-08




30.0






221




Peptidase_M1




Peptidase family M1




1.5e-182




529.4






222




ig




Immunoglobulin domain




3.1e-07




28.4






224




BTB




BTB/POZ domain




1.8e-27




104.7






225




F-box




F-box domain.




3.5e-05




30.6






229




Na_Ca_Ex




Sodium/calcium exchanger protein




1.1e-23




92.1






231




lactamase_B




Metallo-beta-lactamase superfamily




0.01




−5.3






232




fibrinogen_C




Fibrinogen beta and gamma chains, C-terminal globular domain




7.5e-40




140.2






233




MIF




Macrophage migration inhibitory factor (MIF)




5.4e-66




232.7






234




SSF




Sodium:solute symporter family




1.7e-234




792.4






235




Cation_efflux




Cation efflux family




2.2e-63




224.0






237




AAA




ATPases associated with various cellular activities (AAA)




2.6e-85




296.8






238




HCO3_cotransp




HCO3- transporter family




0




1395.3






239




homeobox




Homeobox domain




1.1e-14




62.2






243




GST




Glutathione S-transferases.




0.0024




14.4






244




Viral_helicasel




Viral (Superfamily 1) RNA helicase




0.0019




14.7






245




Na_Galacto_symp




Sodium:galactoside symporter family




0.0068




−94.4






246




PPR




PPR repeat




0.0024




24.5






249




ENV_polyprotein




ENV polyprotein (coat polyprotein)




3.9e-45




155.2






252




UQ_con




Ubiquitin-conjugating enzyme




7.6e-15




62.8






254




7tm_1




7 transmembrane receptor (rhodopsin family)




  2e-32




105.2






255




7tm_1




7 transmembrane receptor (rhodopsin family)




9.6e-43




137.9






256




UBA




UBA domain




1.2e-08




42.1






259




transmembrane4




Transmembrane 4 family




4.2e-44




144.0






267




thyroglobulin_1




Thyroglobulin type-1 repeat




3.2e-35




130.4






268




annexin




Annexin




6.7e-80




278.9






269




annexin




Annexin




8.5e-122




418.0






270




annexin




Annexin




6.7e-80




278.9






271




Armadillo_seg




Armadillo/beta-catenin-like repeats




6.4e-06




33.1






273




p450




Cytochrome P450




2.3e-132




453.1






274




p450




Cytochrome P450




5.9e-52




186.0






275




A2M_N




Alpha-2-macroglobulin family N-terminal region




2.1e-72




247.4






277




Thymosin




Thymosin beta-4 family




2.3e-16




67.8






279




WD40




WD domain, G-beta repeat




5.8e-19




76.4






288




F-box




F-box domain.




0.012




22.3






289




LRR




Leucine Rich Repeat




1.3e-17




72.0






290




TPR




TPR Domain




7.5e-54




192.3






293




cadherin




Cadherin domain




8.2e-07




36.1






297




UCH-1




Ubiquitin carboxyl-terminal hydrolases family 2




4.9e-08




40.1






298




ank




Ank repeat




9.1e-74




258.5






302




Clq




Clq domain




0.023




14.1






304




laminin_Nterm




Laminin N-terminal (Domain VI)




3.2e-52




186.9






305




enolase




Enol-ase




2.4e-69




243.8






307




Nol1_Nop2_Sun




NOL1/NOP2/sun family




0.0015




15.2






308




HIT




HIT family




3.2e-50




180.3






309




ank




Ank repeat




1e-20




82.3






310




lectin_c




Lectin C-type domain




8.8e-19




75.8






311




gpdh




glyceraldehyde 3-phosphate dehydrogenases




8.4e-237




793.9






314




mito_carr




Mitochondrial carrier proteins




  2e-59




206.6






315




serpin




Serpins (serine protease inhibitors)




1.5e-109




372.9






316




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




5.1e-141




481.9






317




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




1.2e-132




454.1






318




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




3.2e-122




419.4






319




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




1.3e-119




410.8






320




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




1.8e-122




420.3






321




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




1.7e-131




450.2






323




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




  4e-144




492.2






324




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




3.8e-105




362.7






325




MHC_I




Class I Histocompatibility antigen, domains alpha 1 and 2




1.3e-139




477.2






326




Kelch




Kelch motif




1.4e-101




350.9






328




Glycos_transf_2




Glycosyl transferases




1.9e-09




44.8






332




ig




Immunoglobulin domain




3.3e-07




28.3






333




UCH-1




Ubiquitin carboxyl-terminal hydrolases family 2




1.1e-12




55.6






336




ATP-gua_Ptrans




ATP:guanido phosphotransferase




4.1e-15




56.6






337




UBX




UBX domain




6.5e-18




72.9






338




UBX




UBX domain




6.5e-18




72.9






339




DnaJ




DnaJ domain




  1e-36




135.4






340




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




5.3e-07




26.9






341




PKD




PKD domain




0




1485.5






342




CNH




CNH domain




2.7e-24




94.2






344




RhoGAP




RhOGAP domain




1.8e-59




211.0






345




Peptidase_M10




Matrixin




8.2e-110




378.2






346




Peptidase_M10




Matrixin




8.2e-110




378.2






349




UQ_con




Ubiquitin-conjugating enzyme




2.6e-06




0.9






350




ig




Immunoglobulin domain




1.6e-236




767.1






351




CNG_membrane




Transmembrane region cyclic Nucleotide Gated Channel




3.7e-108




372.7






353




RGS




Regulator of G protein signaling domain




4.6e-49




176.4






354




THF_DHG_CYH




Tetrahydrofolate dehydrogenase/cyclohydrolase




  5e-106




365.7






355




gln-synt




Glutamine synthetase




1.9e-194




612.3






356




gln-synt




Glutamine synthetase




  2e-39




125.5






357




SCAN




SCAN domain




9.7e-61




215.2






358




UPF0117




Domain of unknown function DUF36




1.3e-38




131.0






359




Trans_recep




Transient receptor




0




1115.3






362




Ammonium_transp




Ammonium Transporter Family




1.9e-56




200.9






363




Kelch




Kelch motif




3.2e-50




180.3






365




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




7.4e-26




88.2






366




SRCR




Scavenger receptor cysteine-rich domain




1.7e-25




98.1






367




adn_short




short chain dehydrogenase




1.1e-37




138.6






369




ANP




Atrial natriuretic peptide




1.5e-51




183.0






370




EGF




EGF-like domain




3.6e-26




100.4






372




STphosphatase




Ser/Thr protein phosphatase




  1e-31




112.7






373




FKBP




FKBP-type peptidyl-prolyl cis-trans isomerases




1.7e-57




185.6






374




PX




PX domain




7.6e-16




66.1






375




ras




Ras family




5.2e-16




45.4






376




pkinase




Eukaryotic protein kinase domain




1.9e-56




200.9






377




pkinase




Eukaryotic protein kinase domain




1.9e-56




200.9






379




Peptidase_C1




Papain family cysteine protease




4.6e-119




409.0






380




Peptidase_C1




Papain family cysteine protease




7.4e-109




375.1






382




ig




Immunoglobulin domain




8.7e-10




36.6






384




Sec7




Sec7 domain




4.5e-71




249.5






385




cadherin




Cadherin domain




2.7e-95




330.0






387




lipocalin




Lipocalin/cytosolic fatty-acid binding protein family




  1e-35




127.1






388




SH3




SH3 domain




1.5e-10




48.4






390




UCH-2




Ubiquitin carboxyl-terminal hydrolase family 2




1.8e-20




81.4






392




RCC1




Regulator of chromosome condensation (RCC1)




1.5e-14




56.1






397




crystall




Beta/Gamma crystallin




3.3e-38




140.4






403




PHD




PHD-finger




6.7e-15




62.9






404




ferritin




Ferritins




4.1e-114




386.1






405




CUB




CUB domain




2.4e-13




57.8






406




ATP-gua_Ptrans




ATP:guanido phosphotransferase




  5e-05




20.7






407




SPRY




SPRY domain




  2e-09




44.8






410




ELM2




ELM2 domain




9.4e-15




62.4






411




Reprolysin




Reprolysin (M12B) family zinc metalloprotease




7.2e-15




56.4






415




ank




Ank repeat




3.5e-18




73.8






418




C2




C2 domain




  1e-75




264.9






419




C2




C2 domain




  1e-75




264.9






420




p450




Cytochrome P450




9.8e-120




411.2






422




aa_permeases




Amino acid permease




1.3e-08




−108.1






424




kinesin




Kinesin motor domain




1.9e-115




397.0






425




LRR




Leucine Rich Repeat




8.3e-26




99.2






426




kinesin




Kinesin motor domain




  8e-63




222.1






427




zf-DHHC




DHHC zinc finger domain




9.1e-34




125.6






428




EGF




EGF-like domain




3.2e-45




163.6






432




TWIK_channel




TASK K+ channel




1.8e-09




7.9






433




pkinase




Eukaryotic protein kinase domain




2.3e-78




273.8






434




PH




PH domain




0.00018




21.0






436




ig




Immunoglobulin domain




9.2e-09




33.3






440




LRR




Leucine Rich Repeat




2.8e-16




67.5






441




MACPF




MAC/Perforin domain




0.016




−71.3






444




efhand




EF hand




0.00027




27.7






448




EPH_lbd




Ephrin receptor ligand binding domain




7.9e-135




461.3






456




C1q




C1q domain




0.023




14.1






458




ASC




Amiloride-sensitive sodium channel




9.2e-127




434.5






459




pkinase




Eukaryotic protein kinase domain




0.083




10.4






460




PBP




Phosphatidylethanolamine-binding protein




1.2e-71




251.4






464




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




2.8e-12




54.2






466




DEP




Domain found in Dishevelled, Eg1-10, and Pleckstrin




6.7e-18




72.9






470




SCAN




SCAN domain




4.3e-52




186.5






471




EGF




EGF-like domain




5.1e-28




106.5






473




Ferric_reduct




Ferric reductase like transmembrane component




6.8e-74




258.9






474




zf-C2H2




Zinc finger, C2H2 type




5.7e-12




53.2






478




CAP_GLY




CAP-Gly domain




3.4e-46




166.9






477




PAP2




PAP2 superfamily




4.9e-10




46.8






476




SCAN




SCAN domain




9.3e-70




245.2






480




LRR




Leucine Rich Repeat




  1e-13




59.0






483




PH




PH domain




  1e-15




61.2






484




PH




PH domain




2.3e-21




81.3






489




RhoGAP




RhoGAP domain




5.7e-57




202.7






493




annexin




Annexin




4.7e-70




246.2






495




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




4.2e-06




24.0






497




TK




Thymidine kinases




9.4e-118




338.0






501




PHD




PHD-finger




0.01




8.3






504




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




1.3e-19




78.6






506




rvt




Reverse transcriptase (RNA-dependent DNA polymerase)




3.2e-30




113.8






508




Insulin




Insulin/IGF/Relaxin family




6.6e-22




86.2






510




COX5A




Cytochrome c oxidase subunit Va




1.2e-55




198.3






511




Guanylate_kin




Guanylate kinase




6.2e-38




139.4






513




hexokinase




Hexokinase




0




1029.0






516




trypsin




Trypsin




1.4e-78




250.0






519




Glycos_transf_1




Glycosyl transferases group 1




2.2e-27




102.8






520




EMP24_GP25L




emp24/gp25L/p24 family




3.5e-70




246.6






521




EMP24_GP25L




emp24/gp25L/p24 family




3.5e-81




283.1






522




14-3-3




14-3-3 proteins




6.6e-150




511.4






526




zf-CCCH




Zinc finger C-x8-C-x5-C-x3-H type (and similar).




0.039




13.4






527




SpoU_methylase




SpoU rRNA Methylase family




2.1e-27




104.5






529




MMR_HSR1




GTPase of unknown function




1.5e-90




314.2






531




RNase PH




3′ exoribonuclease family




2.2e-96




333.6






534




COLFI




Fibrillar collagen C-terminal domain




5.6e-50




129.2






535




COLFI




Fibrillar collagen C-terminal domain




3.1e-58




150.3






536




CH




Calponin homology (CH) domain




5.6e-14




59.9






541




FH2




Formin Homology 2 Domain




4.5e-07




−23.3






542




AAA




ATPases associated with various cellular activities (AAA)




5.5e-33




123.0






547




lectin_c




Lectin C-type domain




2.3e-29




111.0






551




DEAD




DEAD/DEAH box helicase




9.2e-58




185.6






554




C2




C2 domain




1.3e-51




184.9






555




ank




Ank repeat




1.5e-26




101.6






556




ank




Ank repeat




  6e-137




468.4






559




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




1.1e-10




38.9






560




ig




Immunoglobulin domain




1.2e-81




268.2






562




COesterase




Carboxylesterases




1.4e-15




55.8






564




FYVE




FYVE zinc finger




3.8e-15




58.7






572




SPRY




SPRY domain




0.0059




2.8






573




LRR




Leucine Rich Repeat




0.0076




22.9






575




Skp1




Skp1 family




6.3e-10




46.4






576




RhoGAP




RhoGAP domain




4.2e-31




116.8






577




UQ_con




Ubiquitin-conjugating enzyme




6.3e-50




179.3






579




LRR




Leucine Rich Repeat




8.7e-34




125.7






580




K_tetra




K+ channel tetramerisation domain




0.0016




−5.0






581




RCC1




Regulator of chromosome condensation (RCC1)




8.1e-10




39.5






582




profilin




Profilins




5.4e-63




222.7






583




profilin




Profilins




  4e-48




173.3






585




PHD




PHD-finger




0.041




2.8






586




kinesin




Kinesin motor domain




5.2e-114




392.2






588




aminotran_3




Aminotransferases class-III pyridoxal-phosphate




  4e-75




217.6






593




TPR




TPR Domain




0.00036




27.3






594




PCMT




Protein-L-isoaspartate (D-aspartate) O-methyltransferase (PCMT)




1.6e-11




21.8






597




homeobox




Homeobox domain




6.1e-27




102.9






598




zf-C2H2




Zinc finger, C2H2 type




6.8e-85




295.5






599




Ribosomal_S5




Ribosomal protein S5




9.9e-12




45.7






600




IQ




IQ calmodulin-binding motif




1.8e-18




74.8






604




DUF6




Integral membrane protein DUF6




0.083




9.1






605




zf-C2H2




Zinc finger, C2H2 type




2.1e-05




31.4






606




F-box




F-box domain.




1.7e-05




31.7






607




Ran_BP1




RanBP1 domain.




1.1e-88




308.0






608




actin




Actin




4.4e-156




513.5






611




GTP_EFTU




Elongation factor Tu family




8.5e-22




76.1






619




carb_anhydrase




Eukaryotic-type carbonic anhydrase




2.5e-171




582.6






620




peroxidase




Peroxidase




5.8e-208




704.3






621




FAD_Gly3P_dh




FAD-dependent glycerol-3-phosphate dehydrogenase




0.029




−256.0






623




CH




Calponin homology (CH) domain




1.4e-25




98.4






625




mutT




Bacterial




4.3e-09




36.9






626




HECT




HECT-domain (ubiquitin-transferase).




4.3e-16




66.9






627




ig




Immunoglobulin domain




0.024




12.6






631




Acetyltransf




Acetyltransferase (GNAT) family




2.9e-12




54.2






632




Acetyltransf




Acetyltransferase (GNAT) family




2.9e-12




54.2






634




pyr_redox




Pyridine nucleotide-disulphide oxidoreductase




2.9e-05




20.5






639




Sm




Sm protein




1.3e-07




38.7






640




PDZ




PDZ domain (Also known as DHR or GLGF).




8.1e-15




62.7






643




Syntaxin




Syntaxin




2.3e-09




38.1






645




DnaJ




DnaJ domain




1.3e-39




145.1






646




HMG_box




HMG (high mobility group) box




2.7e-27




104.1






650




HMG_box




HMG (high mobiiity group) box




3.4e-30




113.7






653




DUF6




Integral membrane protein DUF6




  3e-12




54.1






654




FF




FF domain




1.2e-33




125.2






655




FF




FF domain




1.2e-33




125.2






658




aminotran_5




Aminotransferases class-V




1.3e-127




437.3






659




aminotran_5




Aminotransferases class-V




3.5e-94




326.3






666




SAM_PNT




Sterile alpha motif (SAM)/Pointed domain




0.0021




6.9






667




DUF52




Protein of unknown function DUF52




4.7e-64




226.2






668




C2




C2 domain




6.8e-36




132.7






669




C2




C2 domain




7.8e-35




129.2






671




7tm_1




7 transmembrane receptor (rhodopsin family)




2.4e-50




162.1






672




K_tetra




K+ channel tetramerisation domain




1.1e-25




98.7






675




cyclin




Cyclin




7.1e-14




52.2






676




zf-C2H2




Zinc finger, C2H2 type




1.4e-18




75.2






679




PDZ




PDZ domain (Also known as DHR or GLGF).




4.3e-16




66.9






681




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




1.7e-47




171.2






689




Gelsolin




Gelsolin repeat.




8.5e-89




308.4






691




TBC




TBC domain




  2e-08




15.3






692




TBC




TBC domain




  2e-07




1.3






697




GTP_CDC




Cell division protein




2.3e-14




54.4






698




transmembrane4




Transmembrane 4 family




  1e-38




126.8






705




Colipase




Colipase




4.9e-21




83.3






706




SH2




Src homology domain 2




1.9e-05




19.4






710




SUI1




Translation initiation factor SUI1




  5e-48




173.0






715




SH3




SH3 domain




0.013




8.0






716




UBA




UBA domain




1.3e-09




45.4






717




Ribosomal_S21




Ribosomal protein S21




0.0039




11.7






718




transmembrane4




Transmembrane 4 family




3.6e-52




169.8






723




LRR




Leucine Rich Repeat




1.1e-48




175.2






724




UCH-2




Ubiquitin carboxyl-terminal hydrolase family 2




5.6e-28




106.4






725




F-box




F-box domain.




0.0016




25.1






727




FGGY




FGGY family of carbohydrate kinases




5.8e-62




219.3






732




pyr_redox




Pyridine nucleotide-disulphide oxidoreductase




4.2e-23




80.9






733




WD40




WD domain, G-beta repeat




0.073




19.6






735




Ribosomal_L44




Ribosomal protein L44




  1e-38




142.1






738




WD40




WD domain, G-beta repeat




1.3e-08




42.1






742




dCMP_cyt_deam




Cytidine and deoxycytidylate deaminase zinc-binding region




1.7e-09




45.0






743




gntR




Bacterial regulatory proteins, gntR family




0.062




11.4






744




cpn60_TCP1




TCP-1/cpn60 chaperonin family




2.9e-74




260.1






748




zf-C2H2




Zinc finger, C2H2 type




6.5e-15




63.0






751




VHS




VHS domain




1.7e-61




217.7






752




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




4.2e-23




90.2






753




ank




Ank repeat




1.4e-80




281.1






757




cyclin




Cyclin




0.026




11.5






761




SH2




Src homology domain 2




5.6e-05




18.0






762




abhydrolase




alpha/beta hydrolase fold




1.2e-21




85.3






763




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




1.8e-05




22.0






764




WD40




WD domain, G-beta repeat




  1e-05




32.4






766




Acyltransferase




Acyltransferase




0.00021




15.9






771




helicase_C




Helicases conserved C-terminal domain




3e-15




64.1






772




Na_Ca_Ex




Sodium/calcium exchanger protein




  8e-76




265.3






773




vwa




von Willebrand factor type A domain




9.6e-29




108.9






775




DEAD




DEAD/DEAH box helicase




0.042




9.5






776




pkinase




Eukaryotic protein kinase domain




1.6e-07




31.0






780




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




1.1e-05




32.3






782




PX




PX domain




1.5e-23




91.7






783




PMP22_Claudin




PMP-22/EMP/MP20/Claudin family




1.5e-56




201.3






785




homeobox




Homeobox domain




0.00021




23.9






786




homeobox




Homeobox domain




0.00021




23.9






790




efhand




EF hand




3.6e-18




73.8






793




Glyco_hydro_31




Glycosyl hydrolases family 31




9.8e-18




62.0






797




Smr




Smr domain




0.0029




13.0






798




Smr




Smr domain




0.0029




13.0






799




mito_carr




Mitochondrial carrier proteins




3.1e-61




212.8






801




Oxysterol_BP




Oxysterol-binding protein




3.4e-78




273.2






803




lipoxygenase




Lipoxygenase




1.7e-140




480.2






805




Rho_GDI




RHO protein GDP dissociation inhibitor




1.1e-122




420.9






809




homeobox




Homeobox domain




3.9e-32




120.2






810




lipocalin




Lipocalin/cytosolic fatty-acid binding protein family




8.5e-31




110.2






811




ig




Immunoglobulin domain




8.8e-14




49.5






814




Keratin_B2




Keratin, high sulfur B2 protein




3.3e-07




32.9






815




sugar_tr




Sugar (and other) transporter




0.0057




−109.4






817




VPS9




Vacuolar sorting protein 9 (VPS9) domain




1.1e-37




138.7






818




SSF




Sodium:solute symporter family




5.9e-206




697.6






819




Peptidase_M3




Peptidase family M3




1.4e-280




945.5






820




RasGAP




GTPase-activator protein for Ras-like GRPase




1.3e-26




101.9






824




zf-C2H2




Zinc finger, C2H2 type




  2e-87




303.9






825




ATP-synt_C




ATP synthase subunit C




5.4e-08




40.0






826




zf-C3HC4




Zinc finger, C3HC4 type (RING finger)




4.7e-13




46.6






827




FH2




Formin Homology 2 Domain




  9e-55




195.4






833




SH3




SH3 domain




9.2e-14




59.2






835




SET




SET domain




9.9e-52




185.3






836




zf-DHHC




DHHC zinc finger domain




2.4e-25




97.7






837




zf-C2H2




ZinC finger, C2H2 type




8.2e-14




59.3






840




Trans_recep




Transient receptor




0




1115.3






850




Ribosomal_S12




Ribosomal protein S12




  1e-24




93.8






851




Acyltransferase




Acyltransferase




7.3e-06




32.2






853




zf-C2H2




Zinc finger, C2H2 type




5.1e-80




279.2






854




lectin_c




Lectin C-type domain




2.8e-24




94.1






855




lectin_c




Lectin C-type domain




2.8e-24




94.1






856




efhand




EF hand




9.7e-10




45.8






858




LRR




Leucine Rich Repeat




3.1e-09




44.1






868




filament




Intermediate filament proteins




0.00027




20.0






869




MAGE




MAGE family




2.3e-19




77.8






871




Hydrolase




haloacid dehalogenase-like hydrolase




0.00056




12.6






872




Fe_hyd_SSU




Iron hydrogenase small subunit




0.00025




21.9






873




filament




Intermediate filament proteins




3.7e-143




489.0






874




Keratin_B2




Keratin, high sulfur B2 protein




0.082




−51.3






876




metalthio




Metallothionein




4.7e-23




90.0






877




C2




C2 domain




4.7e-53




189.7






880




efhand




EF hand




0.041




20.5






885




EGF




EGF-like domain




1.1e-32




122.0






886




7tm_1




7 transmembrane receptor (rhodopsin family)




0.003




12.5






887




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




0.058




15.6






888




Kelch




Kelch motif




5.3e-22




86.5






889




tsp_1




Thrombospondin type 1 domain




0.0014




23.3






890




adh_zinc




Zinc-binding dehydrogenases




1.5e-55




197.9






891




pkinase




Eukaryotic protein kinase domain




  3e-05




22.8






893




zf-C2H2




Zinc finger, C2H2 type




2.1e-30




114.5






895




V_ATPase_sub_a




V-type ATPase 116kDa subunit family




0




1263.1






896




aa_permeases




Amino acid permease




5.6e-08




−125.3






900




MCT




Monocarboxylate transporter




  1e-42




155.3






901




Filamin




Filamin/ABP280 repeat.




2.3e-20




81.1






905




ank




Ank repeat




3.3e-88




306.5






906




ras




Ras family




1.4e-13




17.8






907




Acyltransferase




Acyltransferase




6.4e-34




126.1






908




wap




WAP-type (Whey Acidic Protein) ‘four-disulfide core’




0.039




7.8






909




arf




ADP-ribosylation factor family




3.7e-11




25.6






910




zf-C2H2




Zinc finger, C2H2 type




2.5e-09




44.4






911




zf-C2H2




Zinc finger, C2H2 type




5.6e-20




79.8






912




BTB




BTB/POZ domain




1.1e-27




105.4






913




SNF2_N




SNF2 and others N-terminal domain




1.7e-71




250.9






914




ras




Ras family




2.4e-69




243.8






916




rrm




RNA recognition motif. (a.k.a. RRM, RBD, or RNP domain)




0.061




15.4






919




T-box




T-box




2.7e-45




159.8






923




PH




PH domain




1.5e-14




57.0






924




isodh




Isocitrate and isopropylmalate dehydrogenases




3.7e-125




421.5






925




ank




Ank repeat




2.5e-08




41.1






926




ank




Ank repeat




2.5e-08




41.1






928




DUF51




Protein of unknown function DUF51




5.7e-13




52.5






929




zf-C2H2




Zinc finger, C2H2 type




2.1e-72




253.9






930




AAA




ATPases associated with various cellular activities (AAA)




1.1e-67




238.3






931




SSF




Sodium:solute symporter family




4.8e-170




578.3






932




SSF




Sodium:solute symporter family




1.3e-198




673.2






934




aminotran_1




Aminotransferases class-I




2e-10




10.6






935




DSPc




Dual specificity phosphatase, catalytic domain




1.1e-29




112.1






936




ig




Immunoglobulin domain




2.7e-15




54.3






937




MCM




MCM2/3/5 family




1.9e-11




−67.4






938




Kelch




Kelch motif




5.7e-56




199.4






939




UBA




UBA domain




2.7e-06




34.3






940




UBA




UBA domain




2.7e-06




34.3






941




UBA




UBA domain




2.7e-06




34.3






942




Na_H_Exchanger




Sodium/hydrogen exchanger family




0.00067




−108.1






943




Acyl-CoA_hydro




Cytosolic long-chain acyl-CoA thioester hydrolase




  9e-74




258.5






944




zf-DHHC




DHHC zinc finger domain




8.6e-18




72.5






945




filament




Intermediate filament proteins




5.6e-29




109.7






946




SPRY




SPRY domain




0.022




−3.0






947




dsrm




Double-stranded RNA binding motif




1.5e-12




55.1






948




SPRY




SPRY domain




2.8e-11




50.9

























TABLE 5










POSITION









OF SIGNAL




maxS








IN AMINO ACID




(MAXIMUM




meanS






SEQ ID NO:




SEQUENCE




SCORE)




(MEAN SCORE)


























1




1-24




0.926




0.738






8




1-48




0.994




0.655






9




1-31




0.984




0.921






11




1-36




0.994




0.757






12




1-20




0.977




0.902






16




1-25




0.921




0.787






17




1-33




0.967




0.803






22




1-26




0.949




0.664






24




1-28




0.929




0.700






26




1-17




0.919




0.828






31




1-19




0.890




0.552






33




1-19




0.981




0.916






35




1-21




0.980




0.904






36




1-21




0.980




0.904






38




1-26




0.951




0.801






41




1-43




0.994




0.659






44




1-19




0.942




0.693






52




1-21




0.989




0.925






57




1-18




0.964




0.812






58




1-19




0.972




0.915






59




1-24




0.997




0.929






60




1-16




0.945




0.737






61




1-31




0.957




0.775






68




1-28




0.988




0.938






69




1-23




0.976




0.897






72




1-33




0.948




0.776






75




1-31




0.991




0.925






76




1-41




0.942




0.703






77




1-36




0.910




0.749






79




1-27




0.962




0.696






82




1-24




0.943




0.832






86




1-27




0.962




0.856






87




1-19




0.967




0.909






88




1-39




0.986




0.922






89




1-28




0.982




0.924






91




1-29




0.984




0.763






92




1-22




0.974




0.796






93




1-29




0.928




0.725






94




1-44




0.995




0.811






95




1-36




0.901




0.766






97




1-25




0.921




0.787






103




1-23




0.966




0.812






106




1-19




0.951




0.895






107




1-16




0.927




0.827






108




1-25




0.949




0.823






110




1-28




0.980




0.848






113




1-24




0.965




0.891






114




1-25




0.946




0.860






119




1-36




0.964




0.648






126




1-32




0.941




0.669






128




1-17




0.995




0.974






135




1-18




0.968




0.799






141




1-24




0.882




0.599






143




1-38




0.991




0.904






146




1-29




0.963




0.888






148




1-19




0.892




0.715






153




1-34




0.921




0.652






154




1-20




0.951




0.839






158




1-31




0.921




0.659






162




1-36




0.992




0.917






176




1-30




0.989




0.910






177




1-28




0.974




0.851






179




1-25




0.937




0.812






182




1-30




0.978




0.786






183




1-27




0.987




0.879






185




1-23




0.923




0.655






196




1-28




0.980




0.893






199




1-27




0.963




0.833






202




1-24




0.976




0.913






203




1-24




0.988




0.967






204




1-22




0.968




0.831






206




1-21




0.952




0.822






207




1-42




0.939




0.682






209




1-22




0.984




0.928






210




1-22




0.984




0.928






217




1-21




0.942




0.713






219




1-18




0.922




0.838






222




1-18




0.988




0.944






226




1-18




0.975




0.958






227




1-18




0.975




0.958






228




1-18




0.975




0.958






229




1-48




0.989




0.889






230




1-23




0.996




0.936






232




1-16




0.967




0.933






245




1-15




0.948




0.907






247




1-27




0.936




0.689






248




1-42




0.978




0.750






249




1-15




0.977




0.966






251




1-26




0.976




0.875






252




1-28




0.973




0.822






253




1-28




0.990




0.925






257




1-22




0.982




0.933






258




1-15




0.986




0.919






259




1-27




0.994




0.900






267




1-21




0.989




0.871






272




1-28




0.976




0.653






278




1-20




0.987




0.916






281




1-30




0.996




0.894






282




1-41




0.983




0.791






286




1-20




0.978




0.893






291




1-17




0.953




0.784






292




1-25




0.950




0.897






293




1-20




0.974




0.912






294




1-15




0.974




0.817






299




1-35




0.973




0.795






302




1-22




0.982




0.872






303




1-18




0.983




0.927






306




1-20




0.934




0.828






307




1-16




0.952




0.807






308




1-19




0.904




0.656






312




1-35




0.957




0.640






313




1-35




0.957




0.640






315




1-33




0.953




0.707






316




1-24




0.981




0.884






317




1-24




0.987




0.914






318




1-21




0.977




0.905






319




1-24




0.978




0.911






320




1-18




0.984




0.958






321




1-18




0.984




0.958






322




1-24




0.989




0.922






323




1-18




0.984




0.956






324




1-18




0.986




0.965






325




1-18




0.986




0.965






326




1-32




0.956




0.706






329




1-48




0.983




0.616






330




1-20




0.965




0.878






334




1-16




0.921




0.828






335




1-20




0.937




0.700






345




1-19




0.995




0.971






346




1-19




0.995




0.971






348




1-20




0.926




0.751






354




1-29




0.981




0.937






362




1-27




0.977




0.849






366




1-24




0.977




0.845






367




1-23




0.990




0.833






369




1-25




0.971




0.894






370




1-16




0.961




0.916






371




1-41




0.980




0.681






379




1-17




0.977




0.921






380




1-17




0.977




0.921






381




1-26




0.993




0.894






383




1-25




0.986




0.939






385




1-33




0.977




0.811






392




1-43




0.992




0.943






393




1-20




0.943




0.882






395




1-20




0.995




0.933






396




1-26




0.938




0.663






398




1-21




0.955




0.767






399




1-19




0.920




0.692






400




1-41




0.937




0.604






401




1-41




0.937




0.604






405




1-19




0.986




0.961






409




1-41




0.923




0.559






411




1-25




0.973




0.853






413




1-20




0.935




0.817






416




1-29




0.958




0.637






417




1-23




0.991




0.740






420




1-29




0.986




0.848






421




1-18




0.997




0.979






425




1-16




0.979




0.964






427




1-25




0.980




0.952






428




1-37




0.989




0.822






430




1-37




0.984




0.878






431




1-42




0.978




0.698






432




1-18




0.969




0.913






435




1-31




0.981




0.856






438




1-36




0.987




0.595






439




1-27




0.955




0.786






441




1-17




0.915




0.825






442




1-34




0.954




0.783






445




1-19




0.941




0.839






448




1-27




0.944




0.778






449




1-20




0.941




0.734






456




1-22




0.982




0.872






479




1-19




0.993




0.931






480




1-22




0.992




0.807






482




1-22




0.918




0.716






485




1-38




0.994




0.887






488




1-24




0.914




0.588






490




1-28




0.990




0.919






494




1-26




0.990




0.969






498




1-36




0.954




0.817






502




1-23




0.896




0.747






503




1-23




0.969




0.855






508




1-24




0.985




0.932






514




1-23




0.985




0.951






516




1-27




0.985




0.927






517




1-21




0.960




0.649






519




1-41




0.990




0.922






520




1-20




0.991




0.954






521




1-20




0.991




0.954






525




1-21




0.975




0.909






530




1-11




0.900




0.758






545




1-18




0.933




0.634






547




1-22




0.899




0.639






548




1-40




0.953




0.668






561




1-16




0.881




0.607






568




1-36




0.924




0.590






569




1-25




0.919




0.718






570




1-28




0.928




0.590






571




1-27




0.967




0.872






578




1-25




0.998




0.934






579




1-30




0.973




0.829






599




1-20




0.937




0.728






603




1-30




0.962




0.801






604




1-17




0.925




0.779






609




1-20




0.981




0.910






614




1-17




0.977




0.921






617




1-25




0.938




0.677






620




1-22




0.975




0.822






629




1-34




0.934




0.552






630




1-28




0.946




0.799






635




1-15




0.954




0.725






636




1-30




0.938




0.808






644




1-24




0.973




0.910






649




1-24




0.920




0.596






652




1-29




0.968




0.769






656




1-28




0.975




0.926






665




1-25




0.977




0.776






670




1-41




0.986




0.847






679




1-24




0.915




0.578






682




1-24




0.950




0.737






683




1-28




0.987




0.785






684




1-19




0.890




0.552






685




1-22




0.968




0.934






686




1-22




0.968




0.934






687




1-23




0.965




0.883






688




1-23




0.965




0.883






690




1-26




0.896




0.615






693




1-30




0.956




0.665






696




1-39




0.971




0.694






698




1-44




0.992




0.576






701




1-45




0.964




0.657






702




1-45




0.964




0.657






705




1-17




0.968




0.947






707




1-28




0.960




0.607






709




1-31




0.977




0.720






714




1-28




0.956




0.604






718




1-47




0.985




0.646






719




1-19




0.990




0.946






729




1-47




0.996




0.556






736




1-18




0.930




0.679






739




1-25




0.992




0.948






741




1-26




0.947




0.594






745




1-22




0.963




0.859






747




1-26




0.956




0.830






755




1-26




0.990




0.959






758




1-40




0.987




0.917






759




1-26




0.985




0.917






762




1-36




0.991




0.868






765




1-24




0.887




0.553






766




1-23




0.995




0.974






767




1-18




0.997




0.977






768




1-22




0.968




0.934






770




1-26




0.974




0.730






773




1-18




0.983




0.939






783




1-24




0.988




0.919






795




1-30




0.939




0.639






796




1-26




0.984




0.746






807




1-23




0.965




0.693






810




1-22




0.962




0.919






811




1-21




0.988




0.911






812




1-36




0.980




0.559






821




1-10




0.880




0.780






823




1-14




0.922




0.678






825




1-39




0.982




0.829






830




1-34




0.973




0.817






831




1-36




0.976




0.794






838




1-18




0.918




0.651






839




1-35




0.991




0.834






841




1-22




0.947




0.677






847




1-24




0.963




0.865






848




1-30




0.967




0.758






849




1-33




0.926




0.807






852




1-25




0.889




0.718






857




1-20




0.995




0.968






859




1-27




0.887




0.642






862




1-41




0.975




0.875






865




1-26




0.921




0.620






878




1-36




0.951




0.782






884




1-19




0.983




0.888






885




1-23




0.971




0.941






886




1-40




0.964




0.560






891




1-39




0.942




0.587






898




1-16




0.945




0.737






899




1-24




0.946




0.593






900




1-44




0.974




0.662






902




1-26




0.974




0.730






903




1-27




0.952




0.832






908




1-25




0.960




0.642






918




1-31




0.956




0.846






921




1-16




0.968




0.921






936




1-19




0.984




0.936






944




1-35




0.954




0.699
























SEQUENCE LISTING











The patent contains a lengthy “Sequence Listing” section. A copy of the “Sequence Listing” is available in electronic form from the USPTO






web site (http://seqdata.uspto.gov/sequence.html?DocID=06783969B1). An electronic copy of the “Sequence Listing” will also be available from the






USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).












Claims
  • 1. An isolated polypeptide encoded by the polynucleotide of SEQ ID NO: 380.
  • 2. A composition comprising the polypeptide of claim 1 and a carrier.
  • 3. The polypeptide of claim 1 wherein the polypeptide is provided on a polypeptide array.
Non-Patent Literature Citations (1)
Entry
Santamaria et al. Cancer Research. Apr. 1998. 58:1624-1630.