Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof

Shao, Wei ;   et al.

Patent Application Summary

U.S. patent application number 10/760470 was filed with the patent office on 2004-07-08 for isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof. This patent application is currently assigned to APPLERA CORPORATION. Invention is credited to Beasley, Ellen M., Di Francesco, Valentina, Merkulov, Gennady V., Shao, Wei.

Application Number20040132152 10/760470
Document ID /
Family ID25231732
Filed Date2004-07-08

United States Patent Application 20040132152
Kind Code A1
Shao, Wei ;   et al. July 8, 2004

Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof

Abstract

The present invention provides amino acid sequences of peptides that are encoded by genes within the human genome, the kinase peptides of the present invention. The present invention specifically provides isolated peptide and nucleic acid molecules, methods of identifying orthologs and paralogs of the kinase peptides, and methods of identifying modulators of the kinase peptides.


Inventors: Shao, Wei; (Frederick, MD) ; Merkulov, Gennady V.; (Baltimore, MD) ; Di Francesco, Valentina; (Rockville, MD) ; Beasley, Ellen M.; (Darnestown, MD)
Correspondence Address:
    CELERA GENOMICS CORP.
    ATTN: WAYNE MONTGOMERY, VICE PRES, INTEL PROPERTY
    45 WEST GUDE DRIVE
    C2-4#20
    ROCKVILLE
    MD
    20850
    US
Assignee: APPLERA CORPORATION
Norwalk
CT

Family ID: 25231732
Appl. No.: 10/760470
Filed: January 21, 2004

Related U.S. Patent Documents

Application Number Filing Date Patent Number
10760470 Jan 21, 2004
09820790 Mar 30, 2001
6716604

Current U.S. Class: 435/194 ; 435/320.1; 435/325; 435/6.18; 435/69.1; 536/23.2
Current CPC Class: C12N 9/1205 20130101
Class at Publication: 435/194 ; 435/006; 435/069.1; 435/320.1; 435/325; 536/023.2
International Class: C12N 009/12; C12Q 001/68; C07H 021/04

Claims



That which is claimed is:

1. An isolated peptide consisting of an amino acid sequence selected from the group consisting of: (a) an amino acid sequence shown in SEQ ID NO:2; (b) an amino acid sequence of an allelic variant of an amino acid sequence shown in SEQ ID NO:2, wherein said allelic variant is encoded by a nucleic acid molecule that hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (c) an amino acid sequence of an ortholog of an amino acid sequence shown in SEQ ID NO:2, wherein said ortholog is encoded by a nucleic acid molecule that hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; and (d) a fragment of an amino acid sequence shown in SEQ ID NO:2, wherein said fragment comprises at least 10 contiguous amino acids.

2. An isolated peptide comprising an amino acid sequence selected from the group consisting of: (a) an amino acid sequence shown in SEQ ID NO:2; (b) an amino acid sequence of an allelic variant of an amino acid sequence shown in SEQ ID NO:2, wherein said allelic variant is encoded by a nucleic acid molecule that hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (c) an amino acid sequence of an ortholog of an amino acid sequence shown in SEQ ID NO:2, wherein said ortholog is encoded by a nucleic acid molecule that hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; and (d) a fragment of an amino acid sequence shown in SEQ ID NO:2, wherein said fragment comprises at least 10 contiguous amino acids.

3. An isolated antibody that selectively binds to a peptide of claim 2.

4. An isolated nucleic acid molecule consisting of a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence that encodes an amino acid sequence shown in SEQ ID NO:2; (b) a nucleotide sequence that encodes of an allelic variant of an amino acid sequence shown in SEQ ID NO:2, wherein said nucleotide sequence hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (c) a nucleotide sequence that encodes an ortholog of an amino acid sequence shown in SEQ ID NO:2, wherein said nucleotide sequence hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (d) a nucleotide sequence that encodes a fragment of an amino acid sequence shown in SEQ ID NO:2, wherein said fragment comprises at least 10 contiguous amino acids; and (e) a nucleotide sequence that is the complement of a nucleotide sequence of (a)-(d).

5. An isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence that encodes an amino acid sequence shown in SEQ ID NO:2; (b) a nucleotide sequence that encodes of an allelic variant of an amino acid sequence shown in SEQ ID NO:2, wherein said nucleotide sequence hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (c) a nucleotide sequence that encodes an ortholog of an amino acid sequence shown in SEQ ID NO:2, wherein said nucleotide sequence hybridizes under stringent conditions to the opposite strand of a nucleic acid molecule shown in SEQ ID NOS:1 or 3; (d) a nucleotide sequence that encodes a fragment of an amino acid sequence shown in SEQ ID NO:2, wherein said fragment comprises at least 10 contiguous amino acids; and (e) a nucleotide sequence that is the complement of a nucleotide sequence of (a)-(d).

6. A gene chip comprising a nucleic acid molecule of claim 5.

7. A transgenic non-human animal comprising a nucleic acid molecule of claim 5.

8. A nucleic acid vector comprising a nucleic acid molecule of claim 5.

9. A host cell containing the vector of claim 8.

10. A method for producing any of the peptides of claim 1 comprising introducing a nucleotide sequence encoding any of the amino acid sequences in (a)-(d) into a host cell, and culturing the host cell under conditions in which the peptides are expressed from the nucleotide sequence.

11. A method for producing any of the peptides of claim 2 comprising introducing a nucleotide sequence encoding any of the amino acid sequences in (a)-(d) into a host cell, and culturing the host cell under conditions in which the peptides are expressed from the nucleotide sequence.

12. A method for detecting the presence of any of the peptides of claim 2 in a sample, said method comprising contacting said sample with a detection agent that specifically allows detection of the presence of the peptide in the sample and then detecting the presence of the peptide.

13. A method for detecting the presence of a nucleic acid molecule of claim 5 in a sample, said method comprising contacting the sample with an oligonucleotide that hybridizes to said nucleic acid molecule under stringent conditions and determining whether the oligonucleotide binds to said nucleic acid molecule in the sample.

14. A method for identifying a modulator of a peptide of claim 2, said method comprising contacting said peptide with an agent and determining if said agent has modulated the function or activity of said peptide.

15. The method of claim 14, wherein said agent is administered to a host cell comprising an expression vector that expresses said peptide.

16. A method for identifying an agent that binds to any of the peptides of claim 2, said method comprising contacting the peptide with an agent and assaying the contacted mixture to determine whether a complex is formed with the agent bound to the peptide.

17. A pharmaceutical composition comprising an agent identified by the method of claim 16 and a pharmaceutically acceptable carrier therefor.

18. A method for treating a disease or condition mediated by a human kinase protein, said method comprising administering to a patient a pharmaceutically effective amount of an agent identified by the method of claim 16.

19. A method for identifying a modulator of the expression of a peptide of claim 2, said method comprising contacting a cell expressing said peptide with an agent, and determining if said agent has modulated the expression of said peptide.

20. An isolated human kinase peptide having an amino acid sequence that shares at least 70% homology with an amino acid sequence shown in SEQ ID NO:2.

21. A peptide according to claim 20 that shares at least 90 percent homology with an amino acid sequence shown in SEQ ID NO:2.

22. An isolated nucleic acid molecule encoding a human kinase peptide, said nucleic acid molecule sharing at least 80 percent homology with a nucleic acid molecule shown in SEQ ID NOS:1 or 3.

23. A nucleic acid molecule according to claim 22 that shares at least 90 percent homology with a nucleic acid molecule shown in SEQ ID NOS:1 or 3.
Description



FIELD OF THE INVENTION

[0001] The present invention is in the field of kinase proteins that are related to the calcium/calmodulin-dependent protein kinase subfamily, recombinant DNA molecules, and protein production. The present invention specifically provides novel peptides and proteins that effect protein phosphorylation and nucleic acid molecules encoding such peptide and protein molecules, all of which are useful in the development of human therapeutics and diagnostic compositions and methods.

BACKGROUND OF THE INVENTION

[0002] Protein Kinases

[0003] Kinases regulate many different cell proliferation, differentiation, and signaling processes by adding phosphate groups to proteins. Uncontrolled signaling has been implicated in a variety of disease conditions including inflammation, cancer, arteriosclerosis, and psoriasis. Reversible protein phosphorylation is the main strategy for controlling activities of eukaryotic cells. It is estimated that more than 1000 of the 10,000 proteins active in a typical mammalian cell are phosphorylated. The high energy phosphate, which drives activation, is generally transferred from adenosine triphosphate molecules (ATP) to a particular protein by protein kinases and removed from that protein by protein phosphatases. Phosphorylation occurs in response to extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc), cell cycle checkpoints, and environmental or nutritional stresses and is roughly analogous to turning on a molecular switch. When the switch goes on, the appropriate protein kinase activates a metabolic enzyme, regulatory protein, receptor, cytoskeletal protein, ion channel or pump, or transcription factor.

[0004] The kinases comprise the largest known protein group, a superfamily of enzymes with widely varied functions and specificities. They are usually named after their substrate, their regulatory molecules, or some aspect of a mutant phenotype. With regard to substrates, the protein kinases may be roughly divided into two groups; those that phosphorylate tyrosine residues (protein tyrosine kinases, PTK) and those that phosphorylate serine or threonine residues (serine/threonine kinases, STK). A few protein kinases have dual specificity and phosphorylate threonine and tyrosine residues. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The N-terminal domain, which contains subdomains I-IV, generally folds into a two-lobed structure, which binds and orients the ATP (or GTP) donor molecule. The larger C terminal lobe, which contains subdomains VI A-XI, binds the protein substrate and carries out the transfer of the gamma phosphate from ATP to the hydroxyl group of a serine, threonine, or tyrosine residue. Subdomain V spans the two lobes.

[0005] The kinases may be categorized into families by the different amino acid sequences (generally between 5 and 100 residues) located on either side of, or inserted into loops of, the kinase domain. These added amino acid sequences allow the regulation of each kinase as it recognizes and interacts with its target protein. The primary structure of the kinase domains is conserved and can be further subdivided into 11 subdomains. Each of the 11 subdomains contains specific residues and motifs or patterns of amino acids that are characteristic of that subdomain and are highly conserved (Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Books, Vol 1:7-20 Academic Press, San Diego, Calif.).

[0006] The second messenger dependent protein kinases primarily mediate the effects of second messengers such as cyclic AMP (cAMP), cyclic GMP, inositol triphosphate, phosphatidylinositol, 3,4,5-triphosphate, cyclic-ADPribose, arachidonic acid, diacylglycerol and calcium-calmodulin. The cyclic-AMP dependent protein kinases (PKA) are important members of the STK family. Cyclic-AMP is an intracellular mediator of hormone action in all prokaryotic and animal cells that have been studied. Such hormone-induced cellular responses include thyroid hormone secretion, cortisol secretion, progesterone secretion, glycogen breakdown, bone resorption, and regulation of heart rate and force of heart muscle contraction. PKA is found in all animal cells and is thought to account for the effects of cyclic-AMP in most of these cells. Altered PKA expression is implicated in a variety of disorders and diseases including cancer, thyroid disorders, diabetes, atherosclerosis, and cardiovascular disease (Isselbacher, K. J. et al. (1994) Harrison's Principles of Internal Medicine, McGraw-Hill, New York, N.Y., pp. 416431, 1887).

[0007] Calcium-calmodulin (CaM) dependent protein kinases are also members of STK family. Calmodulin is a calcium receptor that mediates many calcium regulated processes by binding to target proteins in response to the binding of calcium. The principle target protein in these processes is CaM dependent protein kinases. CaM-kinases are involved in regulation of smooth muscle contraction (MLC kinase), glycogen breakdown (phosphorylase kinase), and neurotransmission (CaM kinase I and CaM kinase II). CaM kinase I phosphorylates a variety of substrates including the neurotransmitter related proteins synapsin I and II, the gene transcription regulator, CREB, and the cystic fibrosis conductance regulator protein, CFTR (Haribabu, B. et al. (1995) EMBO Journal 14:3679-86). CaM II kinase also phosphorylates synapsin at different sites, and controls the synthesis of catecholamines in the brain through phosphorylation and activation of tyrosine hydroxylase. Many of the CaM kinases are activated by phosphorylation in addition to binding to CaM. The kinase may autophosphorylate itself, or be phosphorylated by another kinase as part of a "kinase cascade".

[0008] Another ligand-activated protein kinase is 5'-AMP-activated protein kinase (AMPK) (Gao, G. et al. (1996) J. Biol Chem. 15:8675-81). Mammalian AMPK is a regulator of fatty acid and sterol synthesis through phosphorylation of the enzymes acetyl-CoA carboxylase and hydroxymethylglutaryl-CoA reductase and mediates responses of these pathways to cellular stresses such as heat shock and depletion of glucose and ATP. AMPK is a heterotrimeric complex comprised of a catalytic alpha subunit and two non-catalytic beta and gamma subunits that are believed to regulate the activity of the alpha subunit. Subunits of AMPK have a much wider distribution in non-lipogenic tissues such as brain, heart, spleen, and lung than expected. This distribution suggests that its role may extend beyond regulation of lipid metabolism alone.

[0009] The mitogen-activated protein kinases (MAP) are also members of the STK family. MAP kinases also regulate intracellular signaling pathways. They mediate signal transduction from the cell surface to the nucleus via phosphorylation cascades. Several subgroups have been identified, and each manifests different substrate specificities and responds to distinct extracellular stimuli (Egan, S. E. and Weinberg, R. A. (1993) Nature 365:781-783). MAP kinase signaling pathways are present in mammalian cells as well as in yeast. The extracellular stimuli that activate mammalian pathways include epidermal growth factor (EGF), ultraviolet light, hyperosmolar medium, heat shock, endotoxic lipopolysaccharide (LPS), and pro-inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin-1 (IL-1).

[0010] PRK (proliferation-related kinase) is a serum/cytokine inducible STK that is involved in regulation of the cell cycle and cell proliferation in human megakaroytic cells (Li, B. et al. (1996) J. Biol. Chem. 271:19402-8). PRK is related to the polo (derived from humans polo gene) family of STKs implicated in cell division. PRK is downregulated in lung tumor tissue and may be a proto-oncogene whose deregulated expression in normal tissue leads to oncogenic transformation. Altered MAP kinase expression is implicated in a variety of disease conditions including cancer, inflammation, immune disorders, and disorders affecting growth and development.

[0011] The cyclin-dependent protein kinases (CDKs) are another group of STKs that control the progression of cells through the cell cycle. Cyclins are small regulatory proteins that act by binding to and activating CDKs that then trigger various phases of the cell cycle by phosphorylating and activating selected proteins involved in the mitotic process. CDKs are unique in that they require multiple inputs to become activated. In addition to the binding of cyclin, CDK activation requires the phosphorylation of a specific threonine residue and the dephosphorylation of a specific tyrosine residue.

[0012] Protein tyrosine kinases, PTKs, specifically phosphorylate tyrosine residues on their target proteins and may be divided into transmembrane, receptor PTKs and nontransmembrane, non-receptor PTKs. Transmembrane protein-tyrosine kinases are receptors for most growth factors. Binding of growth factor to the receptor activates the transfer of a phosphate group from ATP to selected tyrosine side chains of the receptor and other specific proteins. Growth factors (GF) associated with receptor PTKs include; epidermal GF, platelet-derived GF, fibroblast GF, hepatocyte GF, insulin and insulin-like GFs, nerve GF, vascular endothelial GF, and macrophage colony stimulating factor.

[0013] Non-receptor PTKs lack transmembrane regions and, instead, form complexes with the intracellular regions of cell surface receptors. Such receptors that function through non-receptor PTKs include those for cytokines, hormones (growth hormone and prolactin) and antigen-specific receptors on T and B lymphocytes.

[0014] Many of these PTKs were first identified as the products of mutant oncogenes in cancer cells where their activation was no longer subject to normal cellular controls. In fact, about one third of the known oncogenes encode PTKs, and it is well known that cellular transformation (oncogenesis) is often accompanied by increased tyrosine phosphorylation activity (Carbonneau H and Tonks N K (1992) Annu. Rev. Cell. Biol. 8:463-93). Regulation of PTK activity may therefore be an important strategy in controlling some types of cancer.

[0015] Calcium/Calmodulin-Dependent Protein Kinases

[0016] The novel human protein, and encoding gene, provided by the present invention is related to the family of calcium/calmodulin-dependent protein kinases, which are serine/threonine kinases. The protein of the present invention shows a high degree of similarity to calcium/calmodulin-dependent protein kinase II (CaM II), and the CaM II beta subunit in particular. Furthermore, the protein/cDNA of the present invention may be an alternative splice form of a protein provided in Genbank gi5326757 (see the amino acid sequence alignment in FIG. 2).

[0017] CaM II is comprised of alpha, beta, gamma, and delta subunits. Each subunit is encoded by a separate gene and alternatively splice forms of each subunit have been found (Breen et al., Biochem. Biophys. Res. Commun. 236 (2), 473-478 (1997)). CaM II exerts important effects on hormones and neurotransmitters that utilize calcium as a second messenger and has been implicated in a wide variety of neuronal and non-neuronal functions, including cell growth control (Tombes et al., Biochim Biophys Acta 1997 Mar. 1;1355(3):281-92). It has been found that certain CaM II isozymes are preferentially expressed in tumor cells and thus certain tumor cells express a completely different spectrum of CaM II isozymes compared with normal cells/tissues (Tombes et al., Biochim Biophys Acta 1997 Mar. 1;1355(3):281-92). Therefore, CaM II plays a key role in cell growth control and tumor proliferation and, importantly, novel human CaM II variants are valuable as potential diagnostic markers and therapeutic targets for cancer.

[0018] Expression of CaM II beta mRNA is elevated in the frontal cortex in schizophrenia and CaM II is known to play a key role in the amplified action of amphetamine induced-dopamine release, which is observed in schizophrenics (Novak et al., Brain Res. Mol. Brain Res. 82 (1-2), 95-100 (2000)). Thus, CaM II, and CaM II beta in particular, may play important roles in schizophrenia.

[0019] Beta-cell CaM II activity is associated with insulin secretion, and multiple isoforms of CaM II are expressed in human islets of Langerhans (Breen et al., Biochem. Biophys. Res. Commun. 236 (2), 473-478 (1997)). It has been suggested that CaM II controls activation-induced cellular differentiation, and is important for imparting antigen-dependent memory to T cells (Bui et al., Cell 100: 457-467, 2000). For a further review of CaM II and CaM II beta, see Wang et al., FEBS Lett. 475 (2), 107-110 (2000) and Li et al., Cytogenet. Cell Genet. 66: 113-116, 1994.

[0020] Calmodulin is a major Ca(2+)-binding protein in the brain, where it modulates numerous Ca(2+)-dependent enzymes and cellular functions. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is particularly important in the brain and is involved in a variety of neuronal functions (Sola et al., Prog Neurobiol 1999 June;58(3):207-32), such as postsynaptic responses (such as long-term potentiation), neurotransmitter synthesis and exocytosis, cytoskeletal interactions and gene transcription (Colbran, Neurochem Int 1992 December;21(4):469-97). Ca2+ and calmodulin antagonists inhibit seizures induced by convulsant agents, indicating that the Ca2+/calmodulin signaling system plays an important role in the onset of seizures. Changes in CaMKII expression has been observed following seizures and, furthermore, expression of calmodulin and CaMKII in microglial cells in the brain increases following seizures (Sola et al., Prog Neurobiol 1999 June;58(3):207-32). CaMKII levels are also altered in pathological states such as Alzheimer's disease and ischemia (Colbran, Neurochem Int 1992 December;21(4):469-97), suggesting a role of CaMKII in these disorders.

[0021] Calmodulin is also important for regulating the plasma membrane calcium pump, which transports Ca2+ out of cells. The pump is inactive in the absence of calmodulin, but is activated by calmodulin binding (Penniston et al., J Membr Biol 1998 Sep. 15;165(2):101-9).

[0022] Due to their importance in cell growth control, novel human CaM II proteins/genes, such as provided by the present invention, are valuable as potential targets for the development of therapeutics to treat cancer and other disorders. Furthermore, SNPs in CaM II genes, such as provided by the present invention, may serve as valuable markers for the diagnosis, prognosis, prevention, and/or treatment of cancer and other disorders.

[0023] Using the information provided by the present invention, reagents such as probes/primers for detecting the SNPs or the expression of the protein/gene provided herein may be readily developed and, if desired, incorporated into kit formats such as nucleic acid arrays, primer extension reactions coupled with mass spec detection (for SNP detection), or TaqMan PCR assays (Applied Biosystems, Foster City, Calif.).

[0024] Kinase proteins, particularly members of the calcium/calmodulin-dependent protein kinase subfamily, are a major target for drug action and development. Accordingly, it is valuable to the field of pharmaceutical development to identify and characterize previously unknown members of this subfamily of kinase proteins. The present invention advances the state of the art by providing previously unidentified human kinase proteins that have homology to members of the calcium/calmodulin-dependent protein kinase subfamily.

SUMMARY OF THE INVENTION

[0025] The present invention is based in part on the identification of amino acid sequences of human kinase peptides and proteins that are related to the calcium/calmodulin-dependent protein kinase subfamily, as well as allelic variants and other mammalian orthologs thereof. These unique peptide sequences, and nucleic acid sequences that encode these peptides, can be used as models for the development of human therapeutic targets, aid in the identification of therapeutic proteins, and serve as targets for the development of human therapeutic agents that modulate kinase activity in cells and tissues that express the kinase. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma.

DESCRIPTION OF THE FIGURE SHEETS

[0026] FIG. 1 provides the nucleotide sequence of a cDNA molecule that encodes the kinase protein of the present invention. (SEQ ID NO:1) In addition, structure and functional information is provided, such as ATG start, stop and tissue distribution, where available, that allows one to readily determine specific uses of inventions based on this molecular sequence. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma.

[0027] FIG. 2 provides the predicted amino acid sequence of the kinase of the present invention. (SEQ ID NO:2) In addition structure and functional information such as protein family, function, and modification sites is provided where available, allowing one to readily determine specific uses of inventions based on this molecular sequence.

[0028] FIG. 3 provides genomic sequences that span the gene encoding the kinase protein of the present invention. (SEQ ID NO:3) In addition structure and functional information, such as intron/exon structure, promoter location, etc., is provided where available, allowing one to readily determine specific uses of inventions based on this molecular sequence. As illustrated in FIG. 3, SNPs were identified at 26 different nucleotide positions.

DETAILED DESCRIPTION OF THE INVENTION

[0029] General Description

[0030] The present invention is based on the sequencing of the human genome. During the sequencing and assembly of the human genome, analysis of the sequence information revealed previously unidentified fragments of the human genome that encode peptides that share structural and/or sequence homology to protein/peptide/domains identified and characterized within the art as being a kinase protein or part of a kinase protein and are related to the calcium/calmodulin-dependent protein kinase subfamily. Utilizing these sequences, additional genomic sequences were assembled and transcript and/or cDNA sequences were isolated and characterized. Based on this analysis, the present invention provides amino acid sequences of human kinase peptides and proteins that are related to the calcium/calmodulin-dependent protein kinase subfamily, nucleic acid sequences in the form of transcript sequences, cDNA sequences and/or genomic sequences that encode these kinase peptides and proteins, nucleic acid variation (allelic information), tissue distribution of expression, and information about the closest art known protein/peptide/domain that has structural or sequence homology to the kinase of the present invention.

[0031] In addition to being previously unknown, the peptides that are provided in the present invention are selected based on their ability to be used for the development of commercially important products and services. Specifically, the present peptides are selected based on homology and/or structural relatedness to known kinase proteins of the calcium/calmodulin-dependent protein kinase subfamily and the expression pattern observed. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. The art has clearly established the commercial importance of members of this family of proteins and proteins that have expression patterns similar to that of the present gene. Some of the more specific features of the peptides of the present invention, and the uses thereof, are described herein, particularly in the Background of the Invention and in the annotation provided in the Figures, and/or are known within the art for each of the known calcium/calmodulin-dependent protein kinase family or subfamily of kinase proteins.

[0032] Specific Embodiments

[0033] Peptide Molecules

[0034] The present invention provides nucleic acid sequences that encode protein molecules that have been identified as being members of the kinase family of proteins and are related to the calcium/calmodulin-depen- dent protein kinase subfamily (protein sequences are provided in FIG. 2, transcript/cDNA sequences are provided in FIG. 1 and genomic sequences are provided in FIG. 3). The peptide sequences provided in FIG. 2, as well as the obvious variants described herein, particularly allelic variants as identified herein and using the information in FIG. 3, will be referred herein as the kinase peptides of the present invention, kinase peptides, or peptides/proteins of the present invention.

[0035] The present invention provides isolated peptide and protein molecules that consist of, consist essentially of, or comprise the amino acid sequences of the kinase peptides disclosed in the FIG. 2, (encoded by the nucleic acid molecule shown in FIG. 1, transcript/cDNA or FIG. 3, genomic sequence), as well as all obvious variants of these peptides that are within the art to make and use. Some of these variants are described in detail below.

[0036] As used herein, a peptide is said to be "isolated" or "purified" when it is substantially free of cellular material or free of chemical precursors or other chemicals. The peptides of the present invention can be purified to homogeneity or other degrees of purity. The level of purification will be based on the intended use. The critical feature is that the preparation allows for the desired function of the peptide, even if in the presence of considerable amounts of other components (the features of an isolated nucleic acid molecule is discussed below).

[0037] In some uses, "substantially free of cellular material" includes preparations of the peptide having less than about 30% (by dry weight) other proteins (i.e., contaminating protein), less than about 20% other proteins, less than about 10% other proteins, or less than about 5% other proteins. When the peptide is recombinantly produced, it can also be substantially free of culture medium, i.e., culture medium represents less than about 20% of the volume of the protein preparation.

[0038] The language "substantially free of chemical precursors or other chemicals" includes preparations of the peptide in which it is separated from chemical precursors or other chemicals that are involved in its synthesis. In one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of the kinase peptide having less than about 30% (by dry weight) chemical precursors or other chemicals, less than about 20% chemical precursors or other chemicals, less than about 10% chemical precursors or other chemicals, or less than about 5% chemical precursors or other chemicals.

[0039] The isolated kinase peptide can be purified from cells that naturally express it, purified from cells that have been altered to express it (recombinant), or synthesized using known protein synthesis methods. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. For example, a nucleic acid molecule encoding the kinase peptide is cloned into an expression vector, the expression vector introduced into a host cell and the protein expressed in the host cell. The protein can then be isolated from the cells by an appropriate purification scheme using standard protein purification techniques. Many of these techniques are described in detail below.

[0040] Accordingly, the present invention provides proteins that consist of the amino acid sequences provided in FIG. 2 (SEQ ID NO:2), for example, proteins encoded by the transcript/cDNA nucleic acid sequences shown in FIG. 1 (SEQ ID NO:1) and the genomic sequences provided in FIG. 3 (SEQ ID NO:3). The amino acid sequence of such a protein is provided in FIG. 2. A protein consists of an amino acid sequence when the amino acid sequence is the final amino acid sequence of the protein.

[0041] The present invention further provides proteins that consist essentially of the amino acid sequences provided in FIG. 2 (SEQ ID NO:2), for example, proteins encoded by the transcript/cDNA nucleic acid sequences shown in FIG. 1 (SEQ ID NO:1) and the genomic sequences provided in FIG. 3 (SEQ ID NO:3). A protein consists essentially of an amino acid sequence when such an amino acid sequence is present with only a few additional amino acid residues, for example from about 1 to about 100 or so additional residues, typically from 1 to about 20 additional residues in the final protein.

[0042] The present invention further provides proteins that comprise the amino acid sequences provided in FIG. 2 (SEQ ID NO:2), for example, proteins encoded by the transcript/cDNA nucleic acid sequences shown in FIG. 1 (SEQ ID NO:1) and the genomic sequences provided in FIG. 3 (SEQ ID NO:3). A protein comprises an amino acid sequence when the amino acid sequence is at least part of the final amino acid sequence of the protein. In such fashion, the protein can be only the peptide or have additional amino acid molecules such as amino acid residues (contiguous encoded sequence) that are naturally associated with it or heterologous amino acid residues/peptide sequences. Such a protein can have a few additional amino acid residues or can comprise several hundred or more additional amino acids. The preferred classes of proteins that are comprised of the kinase peptides of the present invention are the naturally occurring mature proteins. A brief description of how various types of these proteins can be made/isolated is provided below.

[0043] The kinase peptides of the present invention can be attached to heterologous sequences to form chimeric or fusion proteins. Such chimeric and fusion proteins comprise a kinase peptide operatively linked to a heterologous protein having an amino acid sequence not substantially homologous to the kinase peptide. "Operatively linked" indicates that the kinase peptide and the heterologous protein are fused in-frame. The heterologous protein can be fused to the N-terminus or C-terminus of the kinase peptide.

[0044] In some uses, the fusion protein does not affect the activity of the kinase peptide per se. For example, the fusion protein can include, but is not limited to, enzymatic fusion proteins, for example beta-galactosidase fusions, yeast two-hybrid GAL fusions, poly-His fusions, MYC-tagged, HI-tagged and Ig fusions. Such fusion proteins, particularly poly-His fusions, can facilitate the purification of recombinant kinase peptide. In certain host cells (e.g., mammalian host cells), expression and/or secretion of a protein can be increased by using a heterologous signal sequence.

[0045] A chimeric or fusion protein can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different protein sequences are ligated together in-frame in accordance with conventional techniques. 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 which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and re-amplified to generate a chimeric gene sequence (see Ausubel et al., Current Protocols in Molecular Biology, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST protein). A kinase peptide-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the kinase peptide.

[0046] As mentioned above, the present invention also provides and enables obvious variants of the amino acid sequence of the proteins of the present invention, such as naturally occurring mature forms of the peptide, allelic/sequence variants of the peptides, non-naturally occurring recombinantly derived variants of the peptides, and orthologs and paralogs of the peptides. Such variants can readily be generated using art-known techniques in the fields of recombinant nucleic acid technology and protein biochemistry. It is understood, however, that variants exclude any amino acid sequences disclosed prior to the invention.

[0047] Such variants can readily be identified/made using molecular techniques and the sequence information disclosed herein. Further, such variants can readily be distinguished from other peptides based on sequence and/or structural homology to the kinase peptides of the present invention. The degree of homology/identity present will be based primarily on whether the peptide is a functional variant or non-functional variant, the amount of divergence present in the paralog family and the evolutionary distance between the orthologs.

[0048] To determine the percent identity of two amino acid sequences or two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, at least 30%, 40%, 50%, 60%, 70%, 80%, or 90% or more of the length of a reference sequence is aligned for comparison purposes. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid "identity" is equivalent to amino acid or nucleic acid "homology"). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.

[0049] The comparison of sequences and determination of percent identity and similarity between two sequences can be accomplished using a mathematical algorithm. (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991). In a preferred embodiment, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (Devereux, J., et al., Nucleic Acids Res. 12(1):387 (1984)) (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. In another embodiment, the percent identity between two amino acid or nucleotide sequences is determined using the algorithm of E. Myers and W. Miller (CABIOS, 4:11-17 (1989)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.

[0050] The nucleic acid and protein sequences of the present invention can further be used as a "query sequence" to perform a search against sequence databases to, for example, identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (J. Mol. Biol. 215:403-10 (1990)). BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to the nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to the proteins of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (Nucleic Acids Res. 25(17):3389-3402 (1997)). When utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.

[0051] Full-length pre-processed forms, as well as mature processed forms, of proteins that comprise one of the peptides of the present invention can readily be identified as having complete sequence identity to one of the kinase peptides of the present invention as well as being encoded by the same genetic locus as the kinase peptide provided herein. The gene encoding the novel kinase protein of the present invention is located on a genome component that has been mapped to human chromosome 7 (as indicated in FIG. 3), which is supported by multiple lines of evidence, such as STS and BAC map data.

[0052] Allelic variants of a kinase peptide can readily be identified as being a human protein having a high degree (significant) of sequence homology/identity to at least a portion of the kinase peptide as well as being encoded by the same genetic locus as the kinase peptide provided herein. Genetic locus can readily be determined based on the genomic information provided in FIG. 3, such as the genomic sequence mapped to the reference human. The gene encoding the novel kinase protein of the present invention is located on a genome component that has been mapped to human chromosome 7 (as indicated in FIG. 3), which is supported by multiple lines of evidence, such as STS and BAC map data. As used herein, two proteins (or a region of the proteins) have significant homology when: the amino acid sequences are typically at least about 70-80%, 80-90%, and more typically at least about 90-95% or more homologous. A significantly homologous amino acid sequence, according to the present invention, will be encoded by a nucleic acid sequence that will hybridize to a kinase peptide encoding nucleic acid molecule under stringent conditions as more fully described below.

[0053] FIG. 3 provides information on SNPs that have been found in the gene encoding the kinase protein of the present invention. SNPs were identified at 26 different nucleotide positions. Some of these SNPs that are located outside the ORF and in introns may affect gene transcription.

[0054] Paralogs of a kinase peptide can readily be identified as having some degree of significant sequence homology/identity to at least a portion of the kinase peptide, as being encoded by a gene from humans, and as having similar activity or function. Two proteins will typically be considered paralogs when the amino acid sequences are typically at least about 60% or greater, and more typically at least about 70% or greater homology through a given region or domain. Such paralogs will be encoded by a nucleic acid sequence that will hybridize to a kinase peptide encoding nucleic acid molecule under moderate to stringent conditions as more fully described below.

[0055] Orthologs of a kinase peptide can readily be identified as having some degree of significant sequence homology/identity to at least a portion of the kinase peptide as well as being encoded by a gene from another organism. Preferred orthologs will be isolated from mammals, preferably primates, for the development of human therapeutic targets and agents. Such orthologs will be encoded by a nucleic acid sequence that will hybridize to a kinase peptide encoding nucleic acid molecule under moderate to stringent conditions, as more fully described below, depending on the degree of relatedness of the two organisms yielding the proteins.

[0056] Non-naturally occurring variants of the kinase peptides of the present invention can readily be generated using recombinant techniques. Such variants include, but are not limited to deletions, additions and substitutions in the amino acid sequence of the kinase peptide. For example, one class of substitutions are conserved amino acid substitution. Such substitutions are those that substitute a given amino acid in a kinase peptide by another amino acid of like characteristics. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu, and Ile; interchange of the hydroxyl residues Ser and Thr; exchange of the acidic residues Asp and Glu; substitution between the amide residues Asn and Gln; exchange of the basic residues Lys and Arg; and replacements among the aromatic residues Phe and Tyr. Guidance concerning which amino acid changes are likely to be phenotypically silent are found in Bowie et al., Science 247:1306-1310 (1990).

[0057] Variant kinase peptides can be fully functional or can lack function in one or more activities, e.g. ability to bind substrate, ability to phosphorylate substrate, ability to mediate signaling, etc. Fully functional variants typically contain only conservative variation or variation in non-critical residues or in non-critical regions. FIG. 2 provides the result of protein analysis and can be used to identify critical domains/regions. Functional variants can also contain substitution of similar amino acids that result in no change or an insignificant change in function. Alternatively, such substitutions may positively or negatively affect function to some degree.

[0058] Non-functional variants typically contain one or more non-conservative amino acid substitutions, deletions, insertions, inversions, or truncation or a substitution, insertion, inversion, or deletion in a critical residue or critical region.

[0059] Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al., Science 244:1081-1085 (1989)), particularly using the results provided in FIG. 2. The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as kinase activity or in assays such as an in vitro proliferative activity. Sites that are critical for binding partner/substrate binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith et al., J. Mol. Biol. 224:899-904 (1992); de Vos et al. Science 255:306-312 (1992)).

[0060] The present invention further provides fragments of the kinase peptides, in addition to proteins and peptides that comprise and consist of such fragments, particularly those comprising the residues identified in FIG. 2. The fragments to which the invention pertains, however, are not to be construed as encompassing fragments that may be disclosed publicly prior to the present invention.

[0061] As used herein, a fragment comprises at least 8, 10, 12, 14, 16, or more contiguous amino acid residues from a kinase peptide. Such fragments can be chosen based on the ability to retain one or more of the biological activities of the kinase peptide or could be chosen for the ability to perform a function, e.g. bind a substrate or act as an immunogen. Particularly important fragments are biologically active fragments, peptides that are, for example, about 8 or more amino acids in length. Such fragments will typically comprise a domain or motif of the kinase peptide, e.g., active site, a transmembrane domain or a substrate-binding domain. Further, possible fragments include, but are not limited to, domain or motif containing fragments, soluble peptide fragments, and fragments containing immunogenic structures. Predicted domains and functional sites are readily identifiable by computer programs well known and readily available to those of skill in the art (e.g., PROSITE analysis). The results of one such analysis are provided in FIG. 2.

[0062] Polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the 20 naturally occurring amino acids. Further, many amino acids, including the terminal amino acids, may be modified by natural processes, such as processing and other post-translational modifications, or by chemical modification techniques well known in the art. Common modifications that occur naturally in kinase peptides are described in basic texts, detailed monographs, and the research literature, and they are well known to those of skill in the art (some of these features are identified in FIG. 2).

[0063] Known modifications include, but are not limited to, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent crosslinks, formation of cystine, formation of pyroglutamate, formylation, gamma carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.

[0064] Such modifications are well known to those of skill in the art and have been described in great detail in the scientific literature. Several particularly common modifications, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, for instance, are described in most basic texts, such as Proteins--Structure and Molecular Properties, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993). Many detailed reviews are available on this subject, such as by Wold, F., Posttranslational Covalent Modification of Proteins, B. C. Johnson, Ed., Academic Press, New York 1-12 (1983); Seifter et al. (Meth. Enzymol. 182: 626-646 (1990)) and Rattan et al. (Ann. N.Y. Acad. Sci. 663:48-62 (1992)).

[0065] Accordingly, the kinase peptides of the present invention also encompass derivatives or analogs in which a substituted amino acid residue is not one encoded by the genetic code, in which a substituent group is included, in which the mature kinase peptide is fused with another compound, such as a compound to increase the half-life of the kinase peptide (for example, polyethylene glycol), or in which the additional amino acids are fused to the mature kinase peptide, such as a leader or secretory sequence or a sequence for purification of the mature kinase peptide or a pro-protein sequence.

[0066] Protein/Peptide Uses

[0067] The proteins of the present invention can be used in substantial and specific assays related to the functional information provided in the Figures; 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 binding partner or ligand) in biological fluids; and 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 a disease state). Where the protein binds or potentially binds to another protein or ligand (such as, for example, in a kinase-effector protein interaction or kinase-ligand interaction), the protein can be used to identify the binding partner/ligand so as to develop a system to identify inhibitors of the binding interaction. Any or all of these uses are capable of being developed into reagent grade or kit format for commercialization as commercial products.

[0068] Methods for performing the uses listed above are well known to those skilled in the art. References disclosing such methods include "Molecular Cloning: A Laboratory Manual", 2d ed., Cold Spring Harbor Laboratory Press, 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.

[0069] The potential uses of the peptides of the present invention are based primarily on the source of the protein as well as the class/action of the protein. For example, kinases isolated from humans and their human/mammalian orthologs serve as targets for identifying agents for use in mammalian therapeutic applications, e.g. a human drug, particularly in modulating a biological or pathological response in a cell or tissue that expresses the kinase. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis. A large percentage of pharmaceutical agents are being developed that modulate the activity of kinase proteins, particularly members of the calcium/calmodulin-dependent protein kinase subfamily (see Background of the Invention). The structural and functional information provided in the Background and Figures provide specific and substantial uses for the molecules of the present invention, particularly in combination with the expression information provided in FIG. 1. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. Such uses can readily be determined using the information provided herein, that which is known in the art, and routine experimentation.

[0070] The proteins of the present invention (including variants and fragments that may have been disclosed prior to the present invention) are useful for biological assays related to kinases that are related to members of the calcium/calmodulin-dependent protein kinase subfamily. Such assays involve any of the known kinase functions or activities or properties useful for diagnosis and treatment of kinase-related conditions that are specific for the subfamily of kinases that the one of the present invention belongs to, particularly in cells and tissues that express the kinase. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis.

[0071] The proteins of the present invention are also useful in drug screening assays, in cell-based or cell-free systems. Cell-based systems can be native, i.e., cells that normally express the kinase, as a biopsy or expanded in cell culture. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. In an alternate embodiment, cell-based assays involve recombinant host cells expressing the kinase protein.

[0072] The polypeptides can be used to identify compounds that modulate kinase activity of the protein in its natural state or an altered form that causes a specific disease or pathology associated with the kinase. Both the kinases of the present invention and appropriate variants and fragments can be used in high-throughput screens to assay candidate compounds for the ability to bind to the kinase. These compounds can be further screened against a functional kinase to determine the effect of the compound on the kinase activity. Further, these compounds can be tested in animal or invertebrate systems to determine activity/effectiveness. Compounds can be identified that activate (agonist) or inactivate (antagonist) the kinase to a desired degree.

[0073] Further, the proteins of the present invention can be used to screen a compound for the ability to stimulate or inhibit interaction between the kinase protein and a molecule that normally interacts with the kinase protein, e.g. a substrate or a component of the signal pathway that the kinase protein normally interacts (for example, another kinase). Such assays typically include the steps of combining the kinase protein with a candidate compound under conditions that allow the kinase protein, or fragment, to interact with the target molecule, and to detect the formation of a complex between the protein and the target or to detect the biochemical consequence of the interaction with the kinase protein and the target, such as any of the associated effects of signal transduction such as protein phosphorylation, cAMP turnover, and adenylate cyclase activation, etc.

[0074] Candidate compounds include, for example, 1) peptides such as soluble peptides, including Ig-tailed fusion peptides and members of random peptide libraries (see, e.g., Lam et al., Nature 354:82-84 (1991); Houghten et al., Nature 354:84-86 (1991)) and combinatorial chemistry-derived molecular libraries made of D- and/or L-configuration amino acids; 2) phosphopeptides (e.g., members of random and partially degenerate, directed phosphopeptide libraries, see, e.g., Songyang et al., Cell 72:767-778 (1993)); 3) antibodies (e.g., polyclonal, monoclonal, humanized, anti-idiotypic, chimeric, and single chain antibodies as well as Fab, F(ab).sub.2, Fab expression library fragments, and epitope-binding fragments of antibodies); and 4) small-organic and inorganic molecules (e.g., molecules obtained from combinatorial and natural product libraries).

[0075] One candidate compound is a soluble fragment of the receptor that competes for substrate binding. Other candidate compounds include mutant kinases or appropriate fragments containing mutations that affect kinase function and thus compete for substrate. Accordingly, a fragment that competes for substrate, for example with a higher affinity, or a fragment that binds substrate but does not allow release, is encompassed by the invention.

[0076] The invention further includes other end point assays to identify compounds that modulate (stimulate or inhibit) kinase activity. The assays typically involve an assay of events in the signal transduction pathway that indicate kinase activity. Thus, the phosphorylation of a substrate, activation of a protein, a change in the expression of genes that are up- or down-regulated in response to the kinase protein dependent signal cascade can be assayed.

[0077] Any of the biological or biochemical functions mediated by the kinase can be used as an endpoint assay. These include all of the biochemical or biochemical/biological events described herein, in the references cited herein, incorporated by reference for these endpoint assay targets, and other functions known to those of ordinary skill in the art or that can be readily identified using the information provided in the Figures, particularly FIG. 2. Specifically, a biological function of a cell or tissues that expresses the kinase can be assayed. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis.

[0078] Binding and/or activating compounds can also be screened by using chimeric kinase proteins in which the amino terminal extracellular domain, or parts thereof, the entire transmembrane domain or subregions, such as any of the seven transmembrane segments or any of the intracellular or extracellular loops and the carboxy terminal intracellular domain, or parts thereof, can be replaced by heterologous domains or subregions. For example, a substrate-binding region can be used that interacts with a different substrate then that which is recognized by the native kinase. Accordingly, a different set of signal transduction components is available as an end-point assay for activation. This allows for assays to be performed in other than the specific host cell from which the kinase is derived.

[0079] The proteins of the present invention are also useful in competition binding assays in methods designed to discover compounds that interact with the kinase (e.g. binding partners and/or ligands). Thus, a compound is exposed to a kinase polypeptide under conditions that allow the compound to bind or to otherwise interact with the polypeptide. Soluble kinase polypeptide is also added to the mixture. If the test compound interacts with the soluble kinase polypeptide, it decreases the amount of complex formed or activity from the kinase target. This type of assay is particularly useful in cases in which compounds are sought that interact with specific regions of the kinase. Thus, the soluble polypeptide that competes with the target kinase region is designed to contain peptide sequences corresponding to the region of interest.

[0080] To perform cell free drug screening assays, it is sometimes desirable to immobilize either the kinase protein, or fragment, or its target molecule to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay.

[0081] Techniques for immobilizing proteins on matrices can be used in the drug screening assays. In one embodiment, a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix. For example, glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, Mo.) or glutathione derivatized microtitre plates, which are then combined with the cell lysates (e.g., .sup.35S-labeled) and the candidate compound, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly, or in the supernatant after the complexes are dissociated. Alternatively, the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of kinase-binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques. For example, either the polypeptide or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin using techniques well known in the art. Alternatively, antibodies reactive with the protein but which do not interfere with binding of the protein to its target molecule can be derivatized to the wells of the plate, and the protein trapped in the wells by antibody conjugation. Preparations of a kinase-binding protein and a candidate compound are incubated in the kinase protein-presenting wells and the amount of complex trapped in the well can be quantitated. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the kinase protein target molecule, or which are reactive with kinase protein and compete with the target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.

[0082] Agents that modulate one of the kinases of the present invention can be identified using one or more of the above assays, alone or in combination. It is generally preferable to use a cell-based or cell free system first and then confirm activity in an animal or other model system. Such model systems are well known in the art and can readily be employed in this context.

[0083] Modulators of kinase protein activity identified according to these drug screening assays can be used to treat a subject with a disorder mediated by the kinase pathway, by treating cells or tissues that express the kinase. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. These methods of treatment include the steps of administering a modulator of kinase activity in a pharmaceutical composition to a subject in need of such treatment, the modulator being identified as described herein.

[0084] In yet another aspect of the invention, the kinase proteins can be used as "bait proteins" in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Pat. No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J. Biol. Chem. 268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and Brent WO94/10300), to identify other proteins, which bind to or interact with the kinase and are involved in kinase activity. Such kinase-binding proteins are also likely to be involved in the propagation of signals by the kinase proteins or kinase targets as, for example, downstream elements of a kinase-mediated signaling pathway. Alternatively, such kinase-binding proteins are likely to be kinase inhibitors.

[0085] The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for a kinase protein is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GALA). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming a kinase-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene which encodes the protein which interacts with the kinase protein.

[0086] This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model. For example, an agent identified as described herein (e.g., a kinase modulating agent, an antisense kinase nucleic acid molecule, a kinase-specific antibody, or a kinase-binding partner) can be used in an animal or other model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent identified as described herein can be used in an animal or other model to determine the mechanism of action of such an agent. Furthermore, this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.

[0087] The kinase proteins of the present invention are also useful to provide a target for diagnosing a disease or predisposition to disease mediated by the peptide. Accordingly, the invention provides methods for detecting the presence, or levels of, the protein (or encoding mRNA) in a cell, tissue, or organism. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. The method involves contacting a biological sample with a compound capable of interacting with the kinase protein such that the interaction can be detected. Such an assay can be provided in a single detection format or a multi-detection format such as an antibody chip array.

[0088] One agent for detecting a protein in a sample is an antibody capable of selectively binding to protein. A biological sample includes tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject.

[0089] The peptides of the present invention also provide targets for diagnosing active protein activity, disease, or predisposition to disease, in a patient having a variant peptide, particularly activities and conditions that are known for other members of the family of proteins to which the present one belongs. Thus, the peptide can be isolated from a biological sample and assayed for the presence of a genetic mutation that results in aberrant peptide. This includes amino acid substitution, deletion, insertion, rearrangement, (as the result of aberrant splicing events), and inappropriate post-translational modification. Analytic methods include altered electrophoretic mobility, altered tryptic peptide digest, altered kinase activity in cell-based or cell-free assay, alteration in substrate or antibody-binding pattern, altered isoelectric point, direct amino acid sequencing, and any other of the known assay techniques useful for detecting mutations in a protein. Such an assay can be provided in a single detection format or a multi-detection format such as an antibody chip array.

[0090] In vitro techniques for detection of peptide include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence using a detection reagent, such as an antibody or protein binding agent. Alternatively, the peptide can be detected in vivo in a subject by introducing into the subject a labeled anti-peptide antibody or other types of detection agent. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques. Particularly useful are methods that detect the allelic variant of a peptide expressed in a subject and methods which detect fragments of a peptide in a sample.

[0091] The peptides are also useful in pharmacogenomic analysis. Pharmacogenomics deal with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See, e.g., Eichelbaum, M. (Clin. Exp. Pharmacol. Physiol. 23(10-11):983-985 (1996)), and Linder, M. W. (Clin. Chem. 43(2):254-266 (1997)). The clinical outcomes of these variations result in severe toxicity of therapeutic drugs in certain individuals or therapeutic failure of drugs in certain individuals as a result of individual variation in metabolism. Thus, the genotype of the individual can determine the way a therapeutic compound acts on the body or the way the body metabolizes the compound. Further, the activity of drug metabolizing enzymes effects both the intensity and duration of drug action. Thus, the pharmacogenomics of the individual permit the selection of effective compounds and effective dosages of such compounds for prophylactic or therapeutic treatment based on the individual's genotype. The discovery of genetic polymorphisms in some drug metabolizing enzymes has explained why some patients do not obtain the expected drug effects, show an exaggerated drug effect, or experience serious toxicity from standard drug dosages. Polymorphisms can be expressed in the phenotype of the extensive metabolizer and the phenotype of the poor metabolizer. Accordingly, genetic polymorphism may lead to allelic protein variants of the kinase protein in which one or more of the kinase functions in one population is different from those in another population. The peptides thus allow a target to ascertain a genetic predisposition that can affect treatment modality. Thus, in a ligand-based treatment, polymorphism may give rise to amino terminal extracellular domains and/or other substrate-binding regions that are more or less active in substrate binding, and kinase activation. Accordingly, substrate dosage would necessarily be modified to maximize the therapeutic effect within a given population containing a polymorphism. As an alternative to genotyping, specific polymorphic peptides could be identified.

[0092] The peptides are also useful for treating a disorder characterized by an absence of, inappropriate, or unwanted expression of the protein. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. Accordingly, methods for treatment include the use of the kinase protein or fragments.

[0093] Antibodies

[0094] The invention also provides antibodies that selectively bind to one of the peptides of the present invention, a protein comprising such a peptide, as well as variants and fragments thereof. As used herein, an antibody selectively binds a target peptide when it binds the target peptide and does not significantly bind to unrelated proteins. An antibody is still considered to selectively bind a peptide even if it also binds to other proteins that are not substantially homologous with the target peptide so long as such proteins share homology with a fragment or domain of the peptide target of the antibody. In this case, it would be understood that antibody binding to the peptide is still selective despite some degree of cross-reactivity.

[0095] As used herein, an antibody is defined in terms consistent with that recognized within the art: they are multi-subunit proteins produced by a mammalian organism in response to an antigen challenge. The antibodies of the present invention include polyclonal antibodies and monoclonal antibodies, as well as fragments of such antibodies, including, but not limited to, Fab or F(ab').sub.2, and Fv fragments.

[0096] Many methods are known for generating and/or identifying antibodies to a given target peptide. Several such methods are described by Harlow, Antibodies, Cold Spring Harbor Press, (1989).

[0097] In general, to generate antibodies, an isolated peptide is used as an immunogen and is administered to a mammalian organism, such as a rat, rabbit or mouse. The full-length protein, an antigenic peptide fragment or a fusion protein can be used. Particularly important fragments are those covering functional domains, such as the domains identified in FIG. 2, and domain of sequence homology or divergence amongst the family, such as those that can readily be identified using protein alignment methods and as presented in the Figures.

[0098] Antibodies are preferably prepared from regions or discrete fragments of the kinase proteins. Antibodies can be prepared from any region of the peptide as described herein. However, preferred regions will include those involved in function/activity and/or kinase/binding partner interaction. FIG. 2 can be used to identify particularly important regions while sequence alignment can be used to identify conserved and unique sequence fragments.

[0099] An antigenic fragment will typically comprise at least 8 contiguous amino acid residues. The antigenic peptide can comprise, however, at least 10, 12, 14, 16 or more amino acid residues. Such fragments can be selected on a physical property, such as fragments correspond to regions that are located on the surface of the protein, e.g., hydrophilic regions or can be selected based on sequence uniqueness (see FIG. 2).

[0100] Detection on an antibody of the present invention can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, .beta.-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include .sup.125I, .sup.131I, .sup.35S or .sup.3H.

[0101] Antibody Uses

[0102] The antibodies can be used to isolate one of the proteins of the present invention by standard techniques, such as affinity chromatography or immunoprecipitation. The antibodies can facilitate the purification of the natural protein from cells and recombinantly produced protein expressed in host cells. In addition, such antibodies are useful to detect the presence of one of the proteins of the present invention in cells or tissues to determine the pattern of expression of the protein among various tissues in an organism and over the course of normal development. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis. Further, such antibodies can be used to detect protein in situ, in vitro, or in a cell lysate or supernatant in order to evaluate the abundance and pattern of expression. Also, such antibodies can be used to assess abnormal tissue distribution or abnormal expression during development or progression of a biological condition. Antibody detection of circulating fragments of the full length protein can be used to identify turnover.

[0103] Further, the antibodies can be used to assess expression in disease states such as in active stages of the disease or in an individual with a predisposition toward disease related to the protein's function. When a disorder is caused by an inappropriate tissue distribution, developmental expression, level of expression of the protein, or expressed/processed form, the antibody can be prepared against the normal protein. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. If a disorder is characterized by a specific mutation in the protein, antibodies specific for this mutant protein can be used to assay for the presence of the specific mutant protein.

[0104] The antibodies can also be used to assess normal and aberrant subcellular localization of cells in the various tissues in an organism. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. The diagnostic uses can be applied, not only in genetic testing, but also in monitoring a treatment modality. Accordingly, where treatment is ultimately aimed at correcting expression level or the presence of aberrant sequence and aberrant tissue distribution or developmental expression, antibodies directed against the protein or relevant fragments can be used to monitor therapeutic efficacy.

[0105] Additionally, antibodies are useful in pharmacogenomic analysis. Thus, antibodies prepared against polymorphic proteins can be used to identify individuals that require modified treatment modalities. The antibodies are also useful as diagnostic tools as an immunological marker for aberrant protein analyzed by electrophoretic mobility, isoelectric point, tryptic peptide digest, and other physical assays known to those in the art.

[0106] The antibodies are also useful for tissue typing. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. Thus, where a specific protein has been correlated with expression in a specific tissue, antibodies that are specific for this protein can be used to identify a tissue type.

[0107] The antibodies are also useful for inhibiting protein function, for example, blocking the binding of the kinase peptide to a binding partner such as a substrate. These uses can also be applied in a therapeutic context in which treatment involves inhibiting the protein's function. An antibody can be used, for example, to block binding, thus modulating (agonizing or antagonizing) the peptides activity. Antibodies can be prepared against specific fragments containing sites required for function or against intact protein that is associated with a cell or cell membrane. See FIG. 2 for structural information relating to the proteins of the present invention.

[0108] The invention also encompasses kits for using antibodies to detect the presence of a protein in a biological sample. The kit can comprise antibodies such as a labeled or labelable antibody and a compound or agent for detecting protein in a biological sample; means for determining the amount of protein in the sample; means for comparing the amount of protein in the sample with a standard; and instructions for use. Such a kit can be supplied to detect a single protein or epitope or can be configured to detect one of a multitude of epitopes, such as in an antibody detection array. Arrays are described in detail below for nuleic acid arrays and similar methods have been developed for antibody arrays.

[0109] Nucleic Acid Molecules

[0110] The present invention further provides isolated nucleic acid molecules that encode a kinase peptide or protein of the present invention (cDNA, transcript and genomic sequence). Such nucleic acid molecules will consist of, consist essentially of, or comprise a nucleotide sequence that encodes one of the kinase peptides of the present invention, an allelic variant thereof, or an ortholog or paralog thereof.

[0111] As used herein, an "isolated" nucleic acid molecule is one that is separated from other nucleic acid present in the natural source of the nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. However, there can be some flanking nucleotide sequences, for example up to about 5 KB, 4 KB, 3 KB, 2 KB, or 1 KB or less, particularly contiguous peptide encoding sequences and peptide encoding sequences within the same gene but separated by introns in the genomic sequence. The important point is that the nucleic acid is isolated from remote and unimportant flanking sequences such that it can be subjected to the specific manipulations described herein such as recombinant expression, preparation of probes and primers, and other uses specific to the nucleic acid sequences.

[0112] Moreover, an "isolated" nucleic acid molecule, such as a transcript/cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized. However, the nucleic acid molecule can be fused to other coding or regulatory sequences and still be considered isolated.

[0113] For example, recombinant DNA molecules contained in a vector are considered isolated. Further examples of isolated DNA molecules include recombinant DNA molecules maintained in heterologous host cells or purified (partially or substantially) DNA molecules in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the isolated DNA molecules of the present invention. Isolated nucleic acid molecules according to the present invention further include such molecules produced synthetically.

[0114] Accordingly, the present invention provides nucleic acid molecules that consist of the nucleotide sequence shown in FIG. 1 or 3 (SEQ ID NO:1, transcript sequence and SEQ ID NO:3, genomic sequence), or any nucleic acid molecule that encodes the protein provided in FIG. 2, SEQ ID NO:2. A nucleic acid molecule consists of a nucleotide sequence when the nucleotide sequence is the complete nucleotide sequence of the nucleic acid molecule.

[0115] The present invention further provides nucleic acid molecules that consist essentially of the nucleotide sequence shown in FIG. 1 or 3 (SEQ ID NO:1, transcript sequence and SEQ ID NO:3, genomic sequence), or any nucleic acid molecule that encodes the protein provided in FIG. 2, SEQ ID NO:2. A nucleic acid molecule consists essentially of a nucleotide sequence when such a nucleotide sequence is present with only a few additional nucleic acid residues in the final nucleic acid molecule.

[0116] The present invention further provides nucleic acid molecules that comprise the nucleotide sequences shown in FIG. 1 or 3 (SEQ ID NO:1, transcript sequence and SEQ ID NO:3, genomic sequence), or any nucleic acid molecule that encodes the protein provided in FIG. 2, SEQ ID NO:2. A nucleic acid molecule comprises a nucleotide sequence when the nucleotide sequence is at least part of the final nucleotide sequence of the nucleic acid molecule. In such a fashion, the nucleic acid molecule can be only the nucleotide sequence or have additional nucleic acid residues, such as nucleic acid residues that are naturally associated with it or heterologous nucleotide sequences. Such a nucleic acid molecule can have a few additional nucleotides or can comprises several hundred or more additional nucleotides. A brief description of how various types of these nucleic acid molecules can be readily made/isolated is provided below.

[0117] In FIGS. 1 and 3, both coding and non-coding sequences are provided. Because of the source of the present invention, humans genomic sequence (FIG. 3) and cDNA/transcript sequences (FIG. 1), the nucleic acid molecules in the Figures will contain genomic intronic sequences, 5' and 3' non-coding sequences, gene regulatory regions and non-coding intergenic sequences. In general such sequence features are either noted in FIGS. 1 and 3 or can readily be identified using computational tools known in the art. As discussed below, some of the non-coding regions, particularly gene regulatory elements such as promoters, are useful for a variety of purposes, e.g. control of heterologous gene expression, target for identifying gene activity modulating compounds, and are particularly claimed as fragments of the genomic sequence provided herein.

[0118] The isolated nucleic acid molecules can encode the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature peptide (when the mature form has more than one peptide chain, for instance). Such sequences may play a role in processing of a protein from precursor to a mature form, facilitate protein trafficking, prolong or shorten protein half-life or facilitate manipulation of a protein for assay or production, among other things. As generally is the case in situ, the additional amino acids may be processed away from the mature protein by cellular enzymes.

[0119] As mentioned above, the isolated nucleic acid molecules include, but are not limited to, the sequence encoding the kinase peptide alone, the sequence encoding the mature peptide and additional coding sequences, such as a leader or secretory sequence (e.g., a pre-pro or pro-protein sequence), the sequence encoding the mature peptide, with or without the additional coding sequences, plus additional non-coding sequences, for example introns and non-coding 5' and 3' sequences such as transcribed but non-translated sequences that play a role in transcription, mRNA processing (including splicing and polyadenylation signals), ribosome binding and stability of mRNA. In addition, the nucleic acid molecule may be fused to a marker sequence encoding, for example, a peptide that facilitates purification.

[0120] Isolated nucleic acid molecules can be in the form of RNA, such as mRNA, or in the form DNA, including cDNA and genomic DNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof. The nucleic acid, especially DNA, can be double-stranded or single-stranded. Single-stranded nucleic acid can be the coding strand (sense strand) or the non-coding strand (anti-sense strand).

[0121] The invention further provides nucleic acid molecules that encode fragments of the peptides of the present invention as well as nucleic acid molecules that encode obvious variants of the kinase proteins of the present invention that are described above. Such nucleic acid molecules may be naturally occurring, such as allelic variants (same locus); paralogs (different locus), and orthologs (different organism), or may be constructed by recombinant DNA methods or by chemical synthesis. Such non-naturally occurring variants may be made by mutagenesis techniques, including those applied to nucleic acid molecules, cells, or organisms. Accordingly, as discussed above, the variants can contain nucleotide substitutions, deletions, inversions and insertions. Variation can occur in either or both the coding and non-coding regions. The variations can produce both conservative and non-conservative amino acid substitutions.

[0122] The present invention further provides non-coding fragments of the nucleic acid molecules provided in FIGS. 1 and 3. Preferred non-coding fragments include, but are not limited to, promoter sequences, enhancer sequences, gene modulating sequences and gene termination sequences. Such fragments are useful in controlling heterologous gene expression and in developing screens to identify gene-modulating agents. A promoter can readily be identified as being 5' to the ATG start site in the genomic sequence provided in FIG. 3.

[0123] A fragment comprises a contiguous nucleotide sequence greater than 12 or more nucleotides. Further, a fragment could at least 30, 40, 50, 100, 250 or 500 nucleotides in length. The length of the fragment will be based on its intended use. For example, the fragment can encode epitope bearing regions of the peptide, or can be useful as DNA probes and primers. Such fragments can be isolated using the known nucleotide sequence to synthesize an oligonucleotide probe. A labeled probe can then be used to screen a cDNA library, genomic DNA library, or mRNA to isolate nucleic acid corresponding to the coding region. Further, primers can be used in PCR reactions to clone specific regions of gene.

[0124] A probe/primer typically comprises substantially a purified oligonucleotide or oligonucleotide pair. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 20, 25, 40, 50 or more consecutive nucleotides.

[0125] Orthologs, homologs, and allelic variants can be identified using methods well known in the art. As described in the Peptide Section, these variants comprise a nucleotide sequence encoding a peptide that is typically 60-70%, 70-80%, 0.80-90%, and more typically at least about 90-95% or more homologous to the nucleotide sequence shown in the Figure sheets or a fragment of this sequence. Such nucleic acid molecules can readily be identified as being able to hybridize under moderate to stringent conditions to the nucleotide sequence shown in the Figure sheets or a fragment of the sequence. Allelic variants can readily be determined by genetic locus of the encoding gene. The gene encoding the novel kinase protein of the present invention is located on a genome component that has been mapped to human chromosome 7 (as indicated in FIG. 3), which is supported by multiple lines of evidence, such as STS and BAC map data.

[0126] FIG. 3 provides information on SNPs that have been found in the gene encoding the kinase protein of the present invention. SNPs were identified at 26 different nucleotide positions. Some of these SNPs that are located outside the ORF and in introns may affect gene transcription.

[0127] As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences encoding a peptide at least 60-70% homologous to each other typically remain hybridized to each other. The conditions can be such that sequences at least about 60%, at least about 70%, or at least about 80% or more homologous to each other typically remain hybridized to each other. Such stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. One example of stringent hybridization conditions are hybridization in 6.times. sodium chloride/sodium citrate (SSC) at about 45C, followed by one or more washes in 0.2.times.SSC, 0.1% SDS at 50-65C. Examples of moderate to low stringency hybridization conditions are well known in the art.

[0128] Nucleic Acid Molecule Uses

[0129] The nucleic acid molecules of the present invention are useful for probes, primers, chemical intermediates, and in biological assays. The nucleic acid molecules are useful as a hybridization probe for messenger RNA, transcript/cDNA and genomic DNA to isolate full-length cDNA and genomic clones encoding the peptide described in FIG. 2 and to isolate cDNA and genomic clones that correspond to variants (alleles, orthologs, etc.) producing the same or related peptides shown in FIG. 2. As illustrated in FIG. 3, SNPs were identified at 26 different nucleotide positions.

[0130] The probe can correspond to any sequence along the entire length of the nucleic acid molecules provided in the Figures. Accordingly, it could be derived from 5' noncoding regions, the coding region, and 3' noncoding regions. However, as discussed, fragments are not to be construed as encompassing fragments disclosed prior to the present invention.

[0131] The nucleic acid molecules are also useful as primers for PCR to amplify any given region of a nucleic acid molecule and are useful to synthesize antisense molecules of desired length and sequence.

[0132] The nucleic acid molecules are also useful for constructing recombinant vectors. Such vectors include expression vectors that express a portion of, or all of, the peptide sequences. Vectors also include insertion vectors, used to integrate into another nucleic acid molecule sequence, such as into the cellular genome, to alter in situ expression of a gene and/or gene product. For example, an endogenous coding sequence can be replaced via homologous recombination with all or part of the coding region containing one or more specifically introduced mutations.

[0133] The nucleic acid molecules are also useful for expressing antigenic portions of the proteins.

[0134] The nucleic acid molecules are also useful as probes for determining the chromosomal positions of the nucleic acid molecules by means of in situ hybridization methods. The gene encoding the novel kinase protein of the present invention is located on a genome component that has been mapped to human chromosome 7 (as indicated in FIG. 3), which is supported by multiple lines of evidence, such as STS and BAC map data.

[0135] The nucleic acid molecules are also useful in making vectors containing the gene regulatory regions of the nucleic acid molecules of the present invention.

[0136] The nucleic acid molecules are also useful for designing ribozymes corresponding to all, or a part, of the mRNA produced from the nucleic acid molecules described herein.

[0137] The nucleic acid molecules are also useful for making vectors that express part, or all, of the peptides.

[0138] The nucleic acid molecules are also useful for constructing host cells expressing a part, or all, of the nucleic acid molecules and peptides.

[0139] The nucleic acid molecules are also useful for constructing transgenic animals expressing all, or a part, of the nucleic acid molecules and peptides.

[0140] The nucleic acid molecules are also useful as hybridization probes for determining the presence, level, form and distribution of nucleic acid expression. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis. Accordingly, the probes can be used to detect the presence of, or to determine levels of, a specific nucleic acid molecule in cells, tissues, and in organisms. The nucleic acid whose level is determined can be DNA or RNA. Accordingly, probes corresponding to the peptides described herein can be used to assess expression and/or gene copy number in a given cell, tissue, or organism. These uses are relevant for diagnosis of disorders involving an increase or decrease in kinase protein expression relative to normal results.

[0141] In vitro techniques for detection of mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detecting DNA includes Southern hybridizations and in situ hybridization.

[0142] Probes can be used as a part of a diagnostic test kit for identifying cells or tissues that express a kinase protein, such as by measuring a level of a kinase-encoding nucleic acid in a sample of cells from a subject e.g., mRNA or genomic DNA, or determining if a kinase gene has been mutated. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis.

[0143] Nucleic acid expression assays are useful for drug screening to identify compounds that modulate kinase nucleic acid expression.

[0144] The invention thus provides a method for identifying a compound that can be used to treat a disorder associated with nucleic acid expression of the kinase gene, particularly biological and pathological processes that are mediated by the kinase in cells and tissues that express it. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma. The method typically includes assaying the ability of the compound to modulate the expression of the kinase nucleic acid and thus identifying a compound that can be used to treat a disorder characterized by undesired kinase nucleic acid expression. The assays can be performed in cell-based and cell-free systems. Cell-based assays include cells naturally expressing the kinase nucleic acid or recombinant cells genetically engineered to express specific nucleic acid sequences.

[0145] The assay for kinase nucleic acid expression can involve direct assay of nucleic acid levels, such as mRNA levels, or on collateral compounds involved in the signal pathway. Further, the expression of genes that are up- or down-regulated in response to the kinase protein signal pathway can also be assayed. In this embodiment the regulatory regions of these genes can be operably linked to a reporter gene such as luciferase.

[0146] Thus, modulators of kinase gene expression can be identified in a method wherein a cell is contacted with a candidate compound and the expression of mRNA determined. The level of expression of kinase mRNA in the presence of the candidate compound is compared to the level of expression of kinase mRNA in the absence of the candidate compound. The candidate compound can then be identified as a modulator of nucleic acid expression based on this comparison and be used, for example to treat a disorder characterized by aberrant nucleic acid expression. When expression of mRNA is statistically significantly greater in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of nucleic acid expression. When nucleic acid expression is statistically significantly less in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of nucleic acid expression.

[0147] The invention further provides methods of treatment, with the nucleic acid as a target, using a compound identified through drug screening as a gene modulator to modulate kinase nucleic acid expression in cells and tissues that express the kinase. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis. Modulation includes both up-regulation (i.e. activation or agonization) or down-regulation (suppression or antagonization) or nucleic acid expression.

[0148] Alternatively, a modulator for kinase nucleic acid expression can be a small molecule or drug identified using the screening assays described herein as long as the drug or small molecule inhibits the kinase nucleic acid expression in the cells and tissues that express the protein. Experimental data as provided in FIG. 1 indicates expression in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma.

[0149] The nucleic acid molecules are also useful for monitoring the effectiveness of modulating compounds on the expression or activity of the kinase gene in clinical trials or in a treatment regimen. Thus, the gene expression pattern can serve as a barometer for the continuing effectiveness of treatment with the compound, particularly with compounds to which a patient can develop resistance. The gene expression pattern can also serve as a marker indicative of a physiological response of the affected cells to the compound. Accordingly, such monitoring would allow either increased administration of the compound or the administration of alternative compounds to which the patient has not become resistant. Similarly, if the level of nucleic acid expression falls below a desirable level, administration of the compound could be commensurately decreased.

[0150] The nucleic acid molecules are also useful in diagnostic assays for qualitative changes in kinase nucleic acid expression, and particularly in qualitative changes that lead to pathology. The nucleic acid molecules can be used to detect mutations in kinase genes and gene expression products such as mRNA. The nucleic acid molecules can be used as hybridization probes to detect naturally occurring genetic mutations in the kinase gene and thereby to determine whether a subject with the mutation is at risk for a disorder caused by the mutation. Mutations include deletion, addition, or substitution of one or more nucleotides in the gene, chromosomal rearrangement, such as inversion or transposition, modification of genomic DNA, such as aberrant methylation patterns or changes in gene copy number, such as amplification. Detection of a mutated form of the kinase gene associated with a dysfunction provides a diagnostic tool for an active disease or susceptibility to disease when the disease results from overexpression, underexpression, or altered expression of a kinase protein.

[0151] Individuals carrying mutations in the kinase gene can be detected at the nucleic acid level by a variety of techniques. FIG. 3 provides information on SNPs that have been found in the gene encoding the kinase protein of the present invention. SNPs were identified at 26 different nucleotide positions. Some of these SNPs that are located outside the ORF and in introns may affect gene transcription. The gene encoding the novel kinase protein of the present invention is located on a genome component that has been mapped to human chromosome 7 (as indicated in FIG. 3), which is supported by multiple lines of evidence, such as STS and BAC map data. Genomic DNA can be analyzed directly or can be amplified by using PCR prior to analysis. RNA or cDNA can be used in the same way. In some uses, detection of the mutation involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g. U.S. Pat. Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al., Science 241:1077-1080 (1988); and Nakazawa et al., PNAS 91:360-364 (1994)), the latter of which can be particularly useful for detecting point mutations in the gene (see Abravaya et al., Nucleic Acids Res. 23:675-682 (1995)). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to a gene under conditions such that hybridization and amplification of the gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. Deletions and insertions can be detected by a change in size of the amplified product compared to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to normal RNA or antisense DNA sequences.

[0152] Alternatively, mutations in a kinase gene can be directly identified, for example, by alterations in restriction enzyme digestion patterns determined by gel electrophoresis.

[0153] Further, sequence-specific ribozymes (U.S. Pat. No. 5,498,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site. Perfectly matched sequences can be distinguished from mismatched sequences by nuclease cleavage digestion assays or by differences in melting temperature.

[0154] Sequence changes at specific locations can also be assessed by nuclease protection assays such as RNase and SI protection or the chemical cleavage method. Furthermore, sequence differences between a mutant kinase gene and a wild-type gene can be determined by direct DNA sequencing. A variety of automated sequencing procedures can be utilized when performing the diagnostic assays (Naeve, C. W., (1995) Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al., Adv. Chromatogr. 36:127-162 (1996); and Griffin et al., Appl. Biochem. Biotechnol. 38:147-159 (1993)).

[0155] Other methods for detecting mutations in the gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA duplexes (Myers et al., Science 230:1242 (1985)); Cotton et al., PNAS 85:4397 (1988); Saleeba et al., Meth. Enzymol. 217:286-295 (1992)), electrophoretic mobility of mutant and wild type nucleic acid is compared (Orita et al., PNAS 86:2766 (1989); Cotton et al., Mutat. Res. 285:125-144 (1993); and Hayashi et al., Genet. Anal. Tech. Appl. 9:73-79 (1992)), and movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (Myers et al., Nature 313:495 (1985)). Examples of other techniques for detecting point mutations include selective oligonucleotide hybridization, selective amplification, and selective primer extension.

[0156] The nucleic acid molecules are also useful for testing an individual for a genotype that while not necessarily causing the disease, nevertheless affects the treatment modality. Thus, the nucleic acid molecules can be used to study the relationship between an individual's genotype and the individual's response to a compound used for treatment (pharmacogenomic relationship). Accordingly, the nucleic acid molecules described herein can be used to assess the mutation content of the kinase gene in an individual in order to select an appropriate compound or dosage regimen for treatment. FIG. 3 provides information on SNPs that have been found in the gene encoding the kinase protein of the present invention. SNPs were identified at 26 different nucleotide positions. Some of these SNPs that are located outside the ORF and in introns may affect gene transcription.

[0157] Thus nucleic acid molecules displaying genetic variations that affect treatment provide a diagnostic target that can be used to tailor treatment in an individual. Accordingly, the production of recombinant cells and animals containing these polymorphisms allow effective clinical design of treatment compounds and dosage regimens.

[0158] The nucleic acid molecules are thus useful as antisense constructs to control kinase gene expression in cells, tissues, and organisms. A DNA antisense nucleic acid molecule is designed to be complementary to a region of the gene involved in transcription, preventing transcription and hence production of kinase protein. An antisense RNA or DNA nucleic acid molecule would hybridize to the mRNA and thus block translation of mRNA into kinase protein.

[0159] Alternatively, a class of antisense molecules can be used to inactivate mRNA in order to decrease expression of kinase nucleic acid. Accordingly, these molecules can treat a disorder characterized by abnormal or undesired kinase nucleic acid expression. This technique involves cleavage by means of ribozymes containing nucleotide sequences complementary to one or more regions in the mRNA that attenuate the ability of the mRNA to be translated. Possible regions include coding regions and particularly coding regions corresponding to the catalytic and other functional activities of the kinase protein, such as substrate binding.

[0160] The nucleic acid molecules also provide vectors for gene therapy in patients containing cells that are aberrant in kinase gene expression. Thus, recombinant cells, which include the patient's cells that have been engineered ex vivo and returned to the patient, are introduced into an individual where the cells produce the desired kinase protein to treat the individual.

[0161] The invention also encompasses kits for detecting the presence of a kinase nucleic acid in a biological sample. Experimental data as provided in FIG. 1 indicates that kinase proteins of the present invention are expressed in fetal brain, testis, lung small cell carcinoma, and uterus endometrium adenocarcinoma, as indicated by virtual northern blot analysis. For example, the kit can comprise reagents such as a labeled or labelable nucleic acid or agent capable of detecting kinase nucleic acid in a biological sample; means for determining the amount of kinase nucleic acid in the sample; and means for comparing the amount of kinase nucleic acid in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect kinase protein mRNA or DNA.

[0162] Nucleic Acid Arrays

[0163] The present invention further provides nucleic acid detection kits, such as arrays or microarrays of nucleic acid molecules that are based on the sequence information provided in FIGS. 1 and 3 (SEQ ID NOS:1 and 3).

[0164] As used herein "Arrays" or "Microarrays" refers to an array of distinct polynucleotides or oligonucleotides synthesized on a substrate, such as paper, nylon or other type of membrane, filter, chip, glass slide, or any other suitable solid support. In one embodiment, the microarray is prepared and used according to the methods described in U.S. Pat. No. 5,837,832, Chee et al., PCT application WO95/11995 (Chee et al.), Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their entirety by reference. In other embodiments, such arrays are produced by the methods described by Brown et al., U.S. Pat. No. 5,807,522.

[0165] The microarray or detection kit is preferably composed of a large number of unique, single-stranded nucleic acid sequences, usually either synthetic antisense oligonucleotides or fragments of cDNAs, fixed to a solid support. The oligonucleotides are preferably about 6-60 nucleotides in length, more preferably 15-30 nucleotides in length, and most preferably about 20-25 nucleotides in length. For a certain type of microarray or detection kit, it may be preferable to use oligonucleotides that are only 7-20 nucleotides in length. The microarray or detection kit may contain oligonucleotides that cover the known 5', or 3', sequence, sequential oligonucleotides which cover the full length sequence; or unique oligonucleotides selected from particular areas along the length of the sequence. Polynucleotides used in the microarray or detection kit may be oligonucleotides that are specific to a gene or genes of interest.

[0166] In order to produce oligonucleotides to a known sequence for a microarray or detection kit, the gene(s) of interest (or an ORF identified from the contigs of the present invention) is typically examined using a computer algorithm which starts at the 5' or at the 3' end of the nucleotide sequence. Typical algorithms will then identify oligomers of defined length that are unique to the gene, have a GC content within a range suitable for hybridization, and lack predicted secondary structure that may interfere with hybridization. In certain situations it may be appropriate to use pairs of oligonucleotides on a microarray or detection kit. The "pairs" will be identical, except for one nucleotide that preferably is located in the center of the sequence. The second oligonucleotide in the pair (mismatched by one) serves as a control. The number of oligonucleotide pairs may range from two to one million. The oligomers are synthesized at designated areas on a substrate using a light-directed chemical process. The substrate may be paper, nylon or other type of membrane, filter, chip, glass slide or any other suitable solid support.

[0167] In another aspect, an oligonucleotide may be synthesized on the surface of the substrate by using a chemical coupling procedure and an ink jet application apparatus, as described in PCT application WO95/251116 (Baldeschweiler et al.) which is incorporated herein in its entirety by reference. In another aspect, a "gridded" array analogous to a dot (or slot) blot may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical bonding procedures. An array, such as those described above, may be produced by hand or by using available devices (slot blot or dot blot apparatus), materials (any suitable solid support), and machines (including robotic instruments), and may contain 8, 24, 96, 384, 1536, 6144 or more oligonucleotides, or any other number between two and one million which lends itself to the efficient use of commercially available instrumentation.

[0168] In order to conduct sample analysis using a microarray or detection kit, the RNA or DNA from a biological sample is made into hybridization probes. The mRNA is isolated, and cDNA is produced and used as a template to make antisense RNA (aRNA). The aRNA is amplified in the presence of fluorescent nucleotides, and labeled probes are incubated with the microarray or detection kit so that the probe sequences hybridize to complementary oligonucleotides of the microarray or detection kit. Incubation conditions are adjusted so that hybridization occurs with precise complementary matches or with various degrees of less complementarity. After removal of nonhybridized probes, a scanner is used to determine the levels and patterns of fluorescence. The scanned images are examined to determine degree of complementarity and the relative abundance of each oligonucleotide sequence on the microarray or detection kit. The biological samples may be obtained from any bodily fluids (such as blood, urine, saliva, phlegm, gastric juices, etc.), cultured cells, biopsies, or other tissue preparations. A detection system may be used to measure the absence, presence, and amount of hybridization for all of the distinct sequences simultaneously. This data may be used for large-scale correlation studies on the sequences, expression patterns, mutations, variants, or polymorphisms among samples.

[0169] Using such arrays, the present invention provides methods to identify the expression of the kinase proteins/peptides of the present invention. In detail, such methods comprise incubating a test sample with one or more nucleic acid molecules and assaying for binding of the nucleic acid molecule with components within the test sample. Such assays will typically involve arrays comprising many genes, at least one of which is a gene of the present invention and or alleles of the kinase gene of the present invention. FIG. 3 provides information on SNPs that have been found in the gene encoding the kinase protein of the present invention. SNPs were identified at 26 different nucleotide positions. Some of these SNPs that are located outside the ORF and in introns may affect gene transcription.

[0170] Conditions for incubating a nucleic acid molecule 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 molecule used in the assay. One skilled in the art will recognize that any one of the commonly available hybridization, amplification or array assay formats can readily be adapted to employ the novel fragments of the Human genome disclosed herein. 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 Enzyme Immunoassays: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1985).

[0171] The test samples of the present invention include cells, protein or membrane extracts of cells. 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 nucleic acid extracts or of cells are well known in the art and can be readily be adapted in order to obtain a sample that is compatible with the system utilized.

[0172] In another embodiment of the present invention, kits are provided which contain the necessary reagents to carry out the assays of the present invention.

[0173] Specifically, the invention provides a compartmentalized kit to receive, in close confinement, one or more containers which comprises: (a) a first container comprising one of the nucleic acid molecules that can bind to a fragment of the Human genome disclosed herein; and (b) one or more other containers comprising one or more of the following: wash reagents, reagents capable of detecting presence of a bound nucleic acid.

[0174] In detail, a compartmentalized kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers, strips of plastic, glass or paper, or arraying material such as silica. 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 nucleic acid probe, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents used to detect the bound probe. One skilled in the art will readily recognize that the previously unidentified kinase gene of the present invention can be routinely identified using the sequence information disclosed herein can be readily incorporated into one of the established kit formats which are well known in the art, particularly expression arrays.

[0175] Vectors/Host Cells

[0176] The invention also provides vectors containing the nucleic acid molecules described herein. The term "vector" refers to a vehicle, preferably a nucleic acid molecule, which can transport the nucleic acid molecules. When the vector is a nucleic acid molecule, the nucleic acid molecules are covalently linked to the vector nucleic acid. With this aspect of the invention, the vector includes a plasmid, single or double stranded phage, a single or double stranded RNA or DNA viral vector, or artificial chromosome, such as a BAC, PAC, YAC, OR MAC.

[0177] A vector can be maintained in the host cell as an extrachromosomal element where it replicates and produces additional copies of the nucleic acid molecules. Alternatively, the vector may integrate into the host cell genome and produce additional copies of the nucleic acid molecules when the host cell replicates.

[0178] The invention provides vectors for the maintenance (cloning vectors) or vectors for expression (expression vectors) of the nucleic acid molecules. The vectors can function in prokaryotic or eukaryotic cells or in both (shuttle vectors).

[0179] Expression vectors contain cis-acting regulatory regions that are operably linked in the vector to the nucleic acid molecules such that transcription of the nucleic acid molecules is allowed in a host cell. The nucleic acid molecules can be introduced into the host cell with a separate nucleic acid molecule capable of affecting transcription. Thus, the second nucleic acid molecule may provide a trans-acting factor interacting with the cis-regulatory control region to allow transcription of the nucleic acid molecules from the vector. Alternatively, a trans-acting factor may be supplied by the host cell. Finally, a trans-acting factor can be produced from the vector itself. It is understood, however, that in some embodiments, transcription and/or translation of the nucleic acid molecules can occur in a cell-free system.

[0180] The regulatory sequence to which the nucleic acid molecules described herein can be operably linked include promoters for directing mRNA transcription. These include, but are not limited to, the left promoter from bacteriophage .lambda., the lac, TRP, and TAC promoters from E. coli, the early and late promoters from SV40, the CMV immediate early promoter, the adenovirus early and late promoters, and retrovirus long-terminal repeats.

[0181] In addition to control regions that promote transcription, expression vectors may also include regions that modulate transcription, such as repressor binding sites and enhancers. Examples include the SV40 enhancer, the cytomegalovirus immediate early enhancer, polyoma enhancer, adenovirus enhancers, and retrovirus LTR enhancers.

[0182] In addition to containing sites for transcription initiation and control, expression vectors can also contain sequences necessary for transcription termination and, in the transcribed region a ribosome binding site for translation. Other regulatory control elements for expression include initiation and termination codons as well as polyadenylation signals. The person of ordinary skill in the art would be aware of the numerous regulatory sequences that are useful in expression vectors. Such regulatory sequences are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual. 2nd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1989).

[0183] A variety of expression vectors can be used to express a nucleic acid molecule. Such vectors include chromosomal, episomal, and virus-derived vectors, for example vectors derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast chromosomal elements, including yeast artificial chromosomes, from viruses such as baculoviruses, papovaviruses such as SV40, Vaccinia viruses, adenoviruses, poxviruses, pseudorabies viruses, and retroviruses. Vectors may also be derived from combinations of these sources such as those derived from plasmid and bacteriophage genetic elements, e.g. cosmids and phagemids. Appropriate cloning and expression vectors for prokaryotic and eukaryotic hosts are described in Sambrook et al., Molecular Cloning: A Laboratory Manual. 2nd. ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1989).

[0184] The regulatory sequence may provide constitutive expression in one or more host cells (i.e. tissue specific) or may provide for inducible expression in one or more cell types such as by temperature, nutrient additive, or exogenous factor such as a hormone or other ligand. A variety of vectors providing for constitutive and inducible expression in prokaryotic and eukaryotic hosts are well known to those of ordinary skill in the art.

[0185] The nucleic acid molecules can be inserted into the vector nucleic acid by well-known methodology. Generally, the DNA sequence that will ultimately be expressed is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction enzymes and then ligating the fragments together. Procedures for restriction enzyme digestion and ligation are well known to those of ordinary skill in the art.

[0186] The vector containing the appropriate nucleic acid molecule can be introduced into an appropriate host cell for propagation or expression using well-known techniques. Bacterial cells include, but are not limited to, E. coli, Streptomyces, and Salmonella typhimurium. Eukaryotic cells include, but are not limited to, yeast, insect cells such as Drosophila, animal cells such as COS and CHO cells, and plant cells.

[0187] As described herein, it may be desirable to express the peptide as a fusion protein. Accordingly, the invention provides fusion vectors that allow for the production of the peptides. Fusion vectors can increase the expression of a recombinant protein, increase the solubility of the recombinant protein, and aid in the purification of the protein by acting for example as a ligand for affinity purification. A proteolytic cleavage site may be introduced at the junction of the fusion moiety so that the desired peptide can ultimately be separated from the fusion moiety. Proteolytic enzymes include, but are not limited to, factor Xa, thrombin, and enterokinase. Typical fusion expression vectors include pGEX (Smith et al., Gene 67:31-40 (1988)), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein. Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., Gene 69:301-315 (1988)) and pET 11d (Studier et al., Gene Expression Technology: Methods in Enzymology 185:60-89 (1990)).

[0188] Recombinant protein expression can be maximized in host bacteria by providing a genetic background wherein the host cell has an impaired capacity to proteolytically cleave the recombinant protein. (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990) 119-128). Alternatively, the sequence of the nucleic acid molecule of interest can be altered to provide preferential codon usage for a specific host cell, for example E. coli. (Wada et al., Nucleic Acids Res. 20:2111-2118 (1992)).

[0189] The nucleic acid molecules can also be expressed by expression vectors that are operative in yeast. Examples of vectors for expression in yeast e.g., S. cerevisiae include pYepSec1 (Baldari, et al., EMBO J. 6:229-234 (1987)), pMFa (Kurjan et al., Cell 30:933-943(1982)), pJRY88 (Schultz et al., Gene 54:113-123 (1987)), and pYES2 (Invitrogen Corporation, San Diego, Calif.).

[0190] The nucleic acid molecules can also be expressed in insect cells using, for example, baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf 9 cells) include the pAc series (Smith et al., Mol. Cell Biol. 3:2156-2165 (1983)) and the pVL series (Lucklow et al., Virology 170:31-39 (1989)).

[0191] In certain embodiments of the invention, the nucleic acid molecules described herein are expressed in mammalian cells using mammalian expression vectors. Examples of mammalian expression vectors include pCDM8 (Seed, B. Nature 329:840(1987)) and pMT2PC (Kaufman et al., EMBO J. 6:187-195 (1987)).

[0192] The expression vectors listed herein are provided by way of example only of the well-known vectors available to those of ordinary skill in the art that would be useful to express the nucleic acid molecules. The person of ordinary skill in the art would be aware of other vectors suitable for maintenance propagation or expression of the nucleic acid molecules described herein. These are found for example in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.

[0193] The invention also encompasses vectors in which the nucleic acid sequences described herein are cloned into the vector in reverse orientation, but operably linked to a regulatory sequence that permits transcription of antisense RNA. Thus, an antisense transcript can be produced to all, or to a portion, of the nucleic acid molecule sequences described herein, including both coding and non-coding regions. Expression of this antisense RNA is subject to each of the parameters described above in relation to expression of the sense RNA (regulatory sequences, constitutive or inducible expression, tissue-specific expression).

[0194] The invention also relates to recombinant host cells containing the vectors described herein. Host cells therefore include prokaryotic cells, lower eukaryotic cells such as yeast, other eukaryotic cells such as insect cells, and higher eukaryotic cells such as mammalian cells.

[0195] The recombinant host cells are prepared by introducing the vector constructs described herein into the cells by techniques readily available to the person of ordinary skill in the art. These include, but are not limited to, calcium phosphate transfection, DEAE-dextran-mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, lipofection, and other techniques such as those found in Sambrook, et al. (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).

[0196] Host cells can contain more than one vector. Thus, different nucleotide sequences can be introduced on different vectors of the same cell. Similarly, the nucleic acid molecules can be introduced either alone or with other nucleic acid molecules that are not related to the nucleic acid molecules such as those providing trans-acting factors for expression vectors. When more than one vector is introduced into a cell, the vectors can be introduced independently, co-introduced or joined to the nucleic acid molecule vector.

[0197] In the case of bacteriophage and viral vectors, these can be introduced into cells as packaged or encapsulated virus by standard procedures for infection and transduction. Viral vectors can be replication-competent or replication-defective. In the case in which viral replication is defective, replication will occur in host cells providing functions that complement the defects.

[0198] Vectors generally include selectable markers that enable the selection of the subpopulation of cells that contain the recombinant vector constructs. The marker can be contained in the same vector that contains the nucleic acid molecules described herein or may be on a separate vector. Markers include tetracycline or ampicillin-resistance genes for prokaryotic host cells and dihydrofolate reductase or neomycin resistance for eukaryotic host cells. However, any marker that provides selection for a phenotypic trait will be effective.

[0199] While the mature proteins can be produced in bacteria, yeast, mammalian cells, and other cells under the control of the appropriate regulatory sequences, cell-free transcription and translation systems can also be used to produce these proteins using RNA derived from the DNA constructs described herein.

[0200] Where secretion of the peptide is desired, which is difficult to achieve with multi-transmembrane domain containing proteins such as kinases, appropriate secretion signals are incorporated into the vector. The signal sequence can be endogenous to the peptides or heterologous to these peptides.

[0201] Where the peptide is not secreted into the medium, which is typically the case with kinases, the protein can be isolated from the host cell by standard disruption procedures, including freeze thaw, sonication, mechanical disruption, use of lysing agents and the like. The peptide can then be recovered and purified by well-known purification methods including ammonium sulfate precipitation, acid extraction, anion or cationic exchange chromatography, phosphocellulose chromatography, hydrophobic-interaction chromatography, affinity chromatography, hydroxylapatite chromatography, lectin chromatography, or high performance liquid chromatography.

[0202] It is also understood that depending upon the host cell in recombinant production of the peptides described herein, the peptides can have various glycosylation patterns, depending upon the cell, or maybe non-glycosylated as when produced in bacteria. In addition, the peptides may include an initial modified methionine in some cases as a result of a host-mediated process.

[0203] Uses of Vectors and Host Cells

[0204] The recombinant host cells expressing the peptides described herein have a variety of uses. First, the cells are useful for producing a kinase protein or peptide that can be further purified to produce desired amounts of kinase protein or fragments. Thus, host cells containing expression vectors are useful for peptide production.

[0205] Host cells are also useful for conducting cell-based assays involving the kinase protein or kinase protein fragments, such as those described above as well as other formats known in the art. Thus, a recombinant host cell expressing a native kinase protein is useful for assaying compounds that stimulate or inhibit kinase protein function.

[0206] Host cells are also useful for identifying kinase protein mutants in which these functions are affected. If the mutants naturally occur and give rise to a pathology, host cells containing the mutations are useful to assay compounds that have a desired effect on the mutant kinase protein (for example, stimulating or inhibiting function) which may not be indicated by their effect on the native kinase protein.

[0207] Genetically engineered host cells can be further used to produce non-human transgenic animals. A transgenic animal is preferably a mammal, for example a rodent, such as a rat or mouse, in which one or more of the cells of the animal include a transgene. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal in one or more cell types or tissues of the transgenic animal. These animals are useful for studying the function of a kinase protein and identifying and evaluating modulators of kinase protein activity. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, and amphibians.

[0208] A transgenic animal can be produced by introducing nucleic acid into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. Any of the kinase protein nucleotide sequences can be introduced as a transgene into the genome of a non-human animal, such as a mouse.

[0209] Any of the regulatory or other sequences useful in expression vectors can form part of the transgenic sequence. This includes intronic sequences and polyadenylation signals, if not already included. A tissue-specific regulatory sequence(s) can be operably linked to the transgene to direct expression of the kinase protein to particular cells.

[0210] Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 and 4,870,009, both by Leder et al., U.S. Pat. No. 4,873,191 by Wagner et al. and in Hogan, B., Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the transgene in its genome and/or expression of transgenic mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene can further be bred to other transgenic animals carrying other transgenes. A transgenic animal also includes animals in which the entire animal or tissues in the animal have been produced using the homologously recombinant host cells described herein.

[0211] In another embodiment, transgenic non-human animals can be produced which contain selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage P1. For a description of the cre/loxP recombinase system, see, e.g., Lakso et al. PNAS 89:6232-6236 (1992). Another example of a recombinase system is the FLP recombinase system of S. cerevisiae (O'Gorman et al. Science 251:1351-1355 (1991). If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein is required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.

[0212] Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, I. et al. Nature 385:810-813 (1997) and PCT International Publication Nos. WO 97/07668 and WO 97/07669. In brief, a cell, e.g., a somatic cell, from the transgenic animal can be isolated and induced to exit the growth cycle and enter Go phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyst and then transferred to pseudopregnant female foster animal. The offspring born of this female foster animal will be a clone of the animal from which the cell, e.g., the somatic cell, is isolated.

[0213] Transgenic animals containing recombinant cells that express the peptides described herein are useful to conduct the assays described herein in an in vivo context. Accordingly, the various physiological factors that are present in vivo and that could effect substrate binding, kinase protein activation, and signal transduction, may not be evident from in vitro cell-free or cell-based assays. Accordingly, it is useful to provide non-human transgenic animals to assay in vivo kinase protein function, including substrate interaction, the effect of specific mutant kinase proteins on kinase protein function and substrate interaction, and the effect of chimeric kinase proteins. It is also possible to assess the effect of null mutations, that is, mutations that substantially or completely eliminate one or more kinase protein functions.

[0214] All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the above-described modes for carrying out the invention which are obvious to those skilled in the field of molecular biology or related fields are intended to be within the scope of the following claims.

Sequence CWU 1

1

30 1 2218 DNA Homo sapiens 1 cgggcgcggc ggcggcggcg gtgacagcgg cgcccgcgcc tccccgcgcg taggtgtgcg 60 gcgcgctcct ggcgaggacg gagcgagcag atctcgcgtg cgctcgccgc ccggcgcagc 120 ccagcccggc ccccgcctgg cgccgcgagc cgaggtgtct cccgcgcccg cgcccgtgtc 180 gccgccgtgc ccgcgagcgg gagccggagt cgccgccgcc cgagcgcagc cgagcgcacg 240 ccgagcccgt ccgccgccgc catggccacc acggtgacct gcacccgctt caccgacgag 300 taccagctct acgaggatat tggcaagggg gctttctctg tggtccgacg ctgtgtcaag 360 ctctgcaccg gccatgagta tgcagccaag atcatcaaca ccaagaagct gtcagccaga 420 gatcaccaga agctggagag agaggctcgg atctgccgcc ttctgaagca ttccaacatc 480 gtgcgtctcc acgacagcat ctccgaggag ggcttccact acctggtctt cgatctggtc 540 actggtgggg agctctttga agacattgtg gcgagagagt actacagcga ggctgatgcc 600 agtcactgta tccagcagat cctggaggcc gttctccatt gtcaccaaat gggggtcgtc 660 cacagagacc tcaagccgga gaacctgctt ctggccagca agtgcaaagg ggctgcagtg 720 aagctggcag acttcggcct agctatcgag gtgcaggggg accagcaggc atggtttggt 780 ttcgctggca caccaggcta cctgtcccct gaggtccttc gcaaagaggc gtatggcaag 840 cctgtggaca tctgggcatg tggggtgatc ctgtacatcc tgctcgtggg ctacccaccc 900 ttctgggacg aggaccagca caagctgtac cagcagatca aggctggtgc ctatgacttc 960 ccgtcccctg agtgggacac cgtcactcct gaagccaaaa acctcatcaa ccagatgctg 1020 accatcaacc ctgccaagcg catcacagcc catgaggccc tgaagcaccc gtgggtctgc 1080 caacgctcca cggtagcatc catgatgcac agacaggaga ctgtggagtg tctgaaaaag 1140 ttcaatgcca ggagaaagct caagggagcc atcctcacca ccatgctggc cacacggaat 1200 ttctcagtgg gcagacagac caccgctccg gccacaatgt ccaccgcggc ctccggcacc 1260 accatggggc tggtggaaca agccaagagt ttactcaaca agaaagcaga tggagtcaag 1320 ccccagacga atagcaccaa aaacagtgca gccgccacca gccccaaagg gacgcttcct 1380 cctgccgccc tggagcctca aaccaccgtc atccataacc cagtggacgg gattaaggag 1440 tcttctgaca gtgccaatac caccatagag gatgaagacg ctaaagcccg gaagcaggag 1500 atcattaaga ccacggagca gctcatcgag gccgtcaaca acggtgactt tgaggcctac 1560 gcattctact tcgagaacct gctggccaag aacagcaagc cgatccacac gaccatcctg 1620 aacccacacg tgcacgtcat tggagaggat gccgcctgca tcgcttacat ccggctcacg 1680 cagtacattg acgggcaggg ccggccccgc accagccagt ctgaggagac ccgcgtgtgg 1740 caccgccgcg acggcaagtg gcagaacgtg cacttccact gctcgggcgc gcctgtggcc 1800 ccgctgcagt gaagccaagg gaggggcaca gaatggggaa caggacacag gatcctaaac 1860 tccaagggga ctgtccaccg atgaacactc agagtggaca ccatcttccg tccacgctgt 1920 gcccaggaca gctgtcccca tccatgaaca cagggtaaac atctgccggg ctccgcacca 1980 gtggctccct gggccatggg acagcggcag ggctcaccac ggacagcacg tggcccagca 2040 gccggccacc ctggcgtcct ggggcctcct cccctcctct ccctctcacc ttgtcacctc 2100 cacggagctg cctgtctggg ataatttggg gatttttttt tctgggggat aattcttttg 2160 catgacccct aaagagcaag ccacaccggt ctgctagcta ggtgtccgcg gtgtggtg 2218 2 516 PRT Homo sapiens 2 Met Ala Thr Thr Val Thr Cys Thr Arg Phe Thr Asp Glu Tyr Gln Leu 1 5 10 15 Tyr Glu Asp Ile Gly Lys Gly Ala Phe Ser Val Val Arg Arg Cys Val 20 25 30 Lys Leu Cys Thr Gly His Glu Tyr Ala Ala Lys Ile Ile Asn Thr Lys 35 40 45 Lys Leu Ser Ala Arg Asp His Gln Lys Leu Glu Arg Glu Ala Arg Ile 50 55 60 Cys Arg Leu Leu Lys His Ser Asn Ile Val Arg Leu His Asp Ser Ile 65 70 75 80 Ser Glu Glu Gly Phe His Tyr Leu Val Phe Asp Leu Val Thr Gly Gly 85 90 95 Glu Leu Phe Glu Asp Ile Val Ala Arg Glu Tyr Tyr Ser Glu Ala Asp 100 105 110 Ala Ser His Cys Ile Gln Gln Ile Leu Glu Ala Val Leu His Cys His 115 120 125 Gln Met Gly Val Val His Arg Asp Leu Lys Pro Glu Asn Leu Leu Leu 130 135 140 Ala Ser Lys Cys Lys Gly Ala Ala Val Lys Leu Ala Asp Phe Gly Leu 145 150 155 160 Ala Ile Glu Val Gln Gly Asp Gln Gln Ala Trp Phe Gly Phe Ala Gly 165 170 175 Thr Pro Gly Tyr Leu Ser Pro Glu Val Leu Arg Lys Glu Ala Tyr Gly 180 185 190 Lys Pro Val Asp Ile Trp Ala Cys Gly Val Ile Leu Tyr Ile Leu Leu 195 200 205 Val Gly Tyr Pro Pro Phe Trp Asp Glu Asp Gln His Lys Leu Tyr Gln 210 215 220 Gln Ile Lys Ala Gly Ala Tyr Asp Phe Pro Ser Pro Glu Trp Asp Thr 225 230 235 240 Val Thr Pro Glu Ala Lys Asn Leu Ile Asn Gln Met Leu Thr Ile Asn 245 250 255 Pro Ala Lys Arg Ile Thr Ala His Glu Ala Leu Lys His Pro Trp Val 260 265 270 Cys Gln Arg Ser Thr Val Ala Ser Met Met His Arg Gln Glu Thr Val 275 280 285 Glu Cys Leu Lys Lys Phe Asn Ala Arg Arg Lys Leu Lys Gly Ala Ile 290 295 300 Leu Thr Thr Met Leu Ala Thr Arg Asn Phe Ser Val Gly Arg Gln Thr 305 310 315 320 Thr Ala Pro Ala Thr Met Ser Thr Ala Ala Ser Gly Thr Thr Met Gly 325 330 335 Leu Val Glu Gln Ala Lys Ser Leu Leu Asn Lys Lys Ala Asp Gly Val 340 345 350 Lys Pro Gln Thr Asn Ser Thr Lys Asn Ser Ala Ala Ala Thr Ser Pro 355 360 365 Lys Gly Thr Leu Pro Pro Ala Ala Leu Glu Pro Gln Thr Thr Val Ile 370 375 380 His Asn Pro Val Asp Gly Ile Lys Glu Ser Ser Asp Ser Ala Asn Thr 385 390 395 400 Thr Ile Glu Asp Glu Asp Ala Lys Ala Arg Lys Gln Glu Ile Ile Lys 405 410 415 Thr Thr Glu Gln Leu Ile Glu Ala Val Asn Asn Gly Asp Phe Glu Ala 420 425 430 Tyr Ala Phe Tyr Phe Glu Asn Leu Leu Ala Lys Asn Ser Lys Pro Ile 435 440 445 His Thr Thr Ile Leu Asn Pro His Val His Val Ile Gly Glu Asp Ala 450 455 460 Ala Cys Ile Ala Tyr Ile Arg Leu Thr Gln Tyr Ile Asp Gly Gln Gly 465 470 475 480 Arg Pro Arg Thr Ser Gln Ser Glu Glu Thr Arg Val Trp His Arg Arg 485 490 495 Asp Gly Lys Trp Gln Asn Val His Phe His Cys Ser Gly Ala Pro Val 500 505 510 Ala Pro Leu Gln 515 3 28438 DNA Homo sapiens 3 gagctgctgt gtctctgtcc ccaggggcag aggggctgtg gggttgcagg ctcagcgtct 60 gggactctgg ggtgaaggct cagccatgcc ctgcagacac catggggcag ggctcagacc 120 tgtgcacctg tctcttgcaa accactgttt tctctgtttt gtaacccccc acccaacccc 180 acataacacc tctgggttta aacaacatgc acccttgtgc cggtcacctc cctgcagccg 240 gagaacctgc ttctggccag caagtgcaaa ggggctgcag tgaagctggc agacttcggc 300 ctagctatcg aggtgcaggg ggaccagcag gcatggtttg gtgagtgcca ggggcagggt 360 gtgttggctg gcagttggca gggcaggagg tgatgctgac agccccttgt ggcctcttcc 420 cctctctcta ggtttcgctg gcacaccagg ctacctgtcc cctgaggtcc ttcgcaaaga 480 ggcgtatggc aagcctgtgg acatctgggc atgtggtgag gcctggcctg agttggtgcg 540 gggcagggcc tcgggtgttt caggacttcc cacctacatc ctggagtgtg cagtggccag 600 cacgtcttgc tctcatctgg gtttatctgt gtcagacctg cccttgagct gccctggcag 660 gggtctgccc acacagccaa gagccccctt tccacccaga ttagaattgc tcacatgaac 720 ctggcgcacc ccagtgctcg cctgcgctca gcagaggtct ggtccagaag tgtggtgggt 780 ggatgggagt ggagaagaga ggtcaggggc tgttgggcca tgggcagggc cacctccttg 840 ggtaggggtc tcctcccaca gaggtgggga gcagcagagg ggcttgacat caccctcatc 900 cctgtgatag tgtgggtgtg gggcagaggt cagggggccg gctgtgccct tctaccccag 960 tgtctgctgc acaggtgggg gcaaaggaat gctgaggacc ccaatgccct cccagggcca 1020 caggagctag gcagtgaggg tgcagggcat gggcttcatg gacggtggca ccctgcaagt 1080 ggctgcggtg ctcacaggcc ccatccgcag gggtgatcct gtacatcctg ctcgtgggct 1140 acccaccctt ctgggacgag gaccagcaca agctgtacca gcagatcaag gctggtgcct 1200 atgacgtgag tgcaccagcc cctctctgat gagctccctt cctccaggtg tggccgggtg 1260 agggcagcgt gggaagaggc taggagtggg gtgaagccac ctgtggccag gtcctgggtc 1320 ctgctctccc agattcgtgg ctggagatga agccccttgg agaattcttg cccctgcctg 1380 agagggagct tcaggcccgg ccggggcgct gtttccttct gcagttcccg tcccctgagt 1440 gggacaccgt cactcctgaa gccaaaaacc tcatcaacca gatgctgacc atcaaccctg 1500 ccaagcgcat cacagcccat gaggccctga agcacccgtg ggtctgcgtg agtcgccctt 1560 ggtgcccatg gtggggaggg ggctcctggt ggagatggcc tcagaccact cccctggcaa 1620 ggaccccaag agggtcctgt tcctgacatc caagagctcc cttgggtccc ctgggtgctc 1680 cttgtggcct ctggcttggg acataccagc acgtttgtga ggcctggggc ttggaaggca 1740 ttagagggta gaggtgatcc cttcctccca actgcagtcc tgtctgtgag gggcagagtg 1800 gacgaggcaa gggagagacg agtcttgaag tcccaggcgg gtggggacag acaacccttg 1860 ccgcaatggt ggccggtggc tcttggcaag tggggacccc agggtgccac aagccttgcc 1920 accctggcct ctcccctgtg cctcgggctc ggctgccata tgaccaccca tttccccaca 1980 gcaacgctcc acggtagcat ccatgatgca cagacaggag actgtggagt gtctgaaaaa 2040 gttcaatgcc aggagaaagc tcaaggtgag gccctggccc ctagtcccag gcacggccat 2100 gcttctctgt gtccctctgg gctggagcag gggggccttg gggggtctgg gcagacctag 2160 gggttactgc tgcccccaag actgactgtt agcaagtccc agactggatg catcaggtga 2220 actcaggcca gcttgggaat gagtccagag gggccctggg ccaggtgtgg ctcctcctag 2280 ttgtctgtgc cacctcctag cagcccttgg aggagctgtc ctgaagcgct cgctgtgggc 2340 tcctcacccg ggctctgcag gcagcactca ccctctggca gtcacactgt ttagtacaag 2400 caagtccgaa gcttccggct cagacaggtt tggtaaggag agcagagcca cacacactgg 2460 tcttgggtgg gctgggggag ttctgggagg gaggtgggtc ccagtagggt atccaacctg 2520 cctgctttgg tcagggctgg ctccggtgac cgcacactgg cagtccctct acttgtgggt 2580 tccgggatgg ggacttgttg cctgactgcc ctctgctggt ctctgagcag ttctccccgg 2640 aagccccagg actgttgccc tgtctgagcc tgtcaggaaa agaaggggct gtcagggagc 2700 tggaccccag aggagctgcc gtggtgacca gctgttctgg tgacccctga ggcttgaggg 2760 gtcttgaagc agctagaagc tgtagttggt caacaggttt aggcccaggg tgtgtgtagt 2820 tctggaaata ggtgatctgt ctcagtgcgg ctgctggctt cctggagctc ttgcctctct 2880 ggaaggctga ggtcatgtca gcctcatgac aatgaggctg agcatctggg caggaggaca 2940 ggggtcttat cctggccaga agccagcagg gaacactgat gggatagccc cggttttatc 3000 tgtgtctctc cccagggagc catcctcacc accatgctgg ccacacggaa tttctcaggt 3060 gagcctttct tctccaggga gacaggcgct gccccctccc tgctggccca cgcaggagag 3120 cgcctccttc ctcaccagcc tctccactcc tcctctgcgg caggcctgcc ctcggcgtct 3180 gccctcagct ctgagaccca ctgcccacct ggccccgctg ggctcccacc ttgggtgata 3240 ccacagggtc cagccccccg aggccatcac cttcgtgctg ggtctgtgtc cctccacccc 3300 ctgaacacga gcgtctgtgc tgccccactg gggctcacag catcgtgtgt gtctgtccag 3360 gcgtttgtcg ggcatctatg tggcctcctt gtcattttga gtgctctgaa cattgtgttt 3420 tgtgcgggag gtgggcagaa gggatgcggg gtgatgcggg aggctcgggg gcctccttcc 3480 aagttctgga tgagctgcag cctcctgtcc cggctgctca gggtgggtgg ttgggaagca 3540 agttctcttg gcaggggggt ggggtctgtt atagacccct gaggcccagg gcgctggcag 3600 acccatcggg gcatgatgtt agccccggag tggagccggc agcccaggtc tggacaagct 3660 gtacctgtgg cttctccgtc gtccgacact ccgtgtgcga gcgtctgtga tccgtctctc 3720 tcgttgtccg tttgcatctg gtgcccccca cccgccatcc tgttactttt gctgtgatgc 3780 tgtaatgccg ggaacgcgtg cacacggtca caccaacact aataggactg tcctgtctgc 3840 tgtgtgctca ccacaccctt tgggcatgag aagcccccac tggggttttc taaggagaaa 3900 ggaggcaaat gcttttccgt gtcaatcagt ccaatcttgt tttcactctc ttgagcaaag 3960 gattctggaa ccatctgtca cctaaacttt aactctaatc ttcttctgct tcctttgtct 4020 cttttcttcc cttacctcgc ccacccctcg tctgtgtccg cccacccctc ccttcccctc 4080 gtctctaacc cggtgctaac agtgggcaga cagaccaccg ctccggccac aatgtccacc 4140 gcggcctccg gcaccaccat ggggctggtg gaacaaggta gatgtgtctc gaccagcgtc 4200 ccgcccgctc ccgcccgtcc ctcctgccag catgcagccc cctgctgcac gcagccgctg 4260 gccgggctcc agagccgccc cagaggccgc caggcccccg ggagcccctg ctcccgtgtg 4320 gtcacatccc agcagagccc accacaaggg cagggaggca gcccccaagg ctcctcgcct 4380 gtaagaggag gggctgggct aggtggcccc tgggctacac caagcccttc tggtcctggc 4440 ccccgaggtc tgggggtccg gagaccccca ttaagaatgg cctgggcccc acagggagcc 4500 actgggcctg ctgctggggg gtctgaatcc tgaaaggaga gccttgagga gcagagccag 4560 agaggcagag gcccttgggg cagacacaca ccctgcccct ctggggccgc atggagacgg 4620 tggtctgtgc tgctgagtcc tacacatgca tgtctgccct gagcatcccc ccaggacaag 4680 ccgctctgga gtgggtgagg gttttatgca ccctgaggag actttcaagg cttcctcttg 4740 ggttgtttct gcaaagtcct cctcccctgg cctcaaaccc tgtgagggaa aaggccggca 4800 ctggccacct gctcctctgg gctgtgcggg gccagagccc agaggcccaa gttggcttct 4860 gcccacctgc tggcttgtga ccatgggcag accccatgag ggctaggcga ccccaagacc 4920 tccttgcagc tccagcctga gctgaaggct ggtgagagct tagggcaggc caagctgaca 4980 acgcctggcc acagaacaca gagggctaca ggggtgaccc cagatcctcc ctgggctgag 5040 ctgctgagtt ccctgtcggt gcctccaacg tgggctgggg acccggcaga ggttccaggg 5100 tgctggagac tgccttcccc aggcctcctc atgacccaca gggtgagcag cctggccttc 5160 ccagccagag aaccctcctt ctggggaggc ccagggcgtc ctcggggagg gcagtctatt 5220 ctcctcccat gagcccagtg gacgtgtcta gcaggcagca ccccgggaga gccctcccac 5280 gtcttctcca tttgacaggc ctttccagag cgcaggcggg agggggctgt gattagaaaa 5340 gagtgaggct agtggcttct ggggaggcac tgctgcccag gggacagtgc tgagagacag 5400 ctgcctctac gctgccctgt gcccggggct cccgctgcaa tgcccgcctg tctgcaagtg 5460 aacgtggggc gacggtgcat gaggccctgc atgtgtggct ccaccctggg cgccgagagc 5520 agctctgtcc tggagggtgg tcagtgcatg tggacagagc ccagcatggc tgtcctgggt 5580 gaccagctaa ggggacaagg cagaggcagg gctgagagga ccacccatcc tgctaggtca 5640 gcccagctca gccatatcac acggcagtga gcatggagct cagttctctg ccaatggcag 5700 ctgagtctag taccatccag tcagagtctg gtaccagccc atgtggcata gccccctcgg 5760 cccgcagaga gaccccgtct gtcgagtgtg cttcagtttg gcctctgtgg tctctcctgc 5820 attgatcagg tgtaagggca taggagaccc agtgtccggc cagctgcagg gtggcagcag 5880 ttgccccggc ctggagaccc gggaatgggc agtgccttcc caggatggag ggcagagggt 5940 ctctccttgt cccacagagg cctgcagaac ccccaaccca ggtgtctgag atgcctgtga 6000 ctgctccgcc taccctgggc tcctgcggca cctaacgcat gctttgaact tgagacacag 6060 aaaggaagtt cccgtgccct tgaatgctag tgtagatggg catcgacagg actctggcca 6120 cggtgaatct ggagttagtc ccaggcagag atgtgaaatg agcagccccc caaaaaatgg 6180 ttggccggga gccatgcact caggagggcc gggcccatgc accccacact gcgcccaagg 6240 cgtgcacaag cgattgtttt aaaagcgggt tcacaaggaa ggatgtttgg gaactgactg 6300 agacaacagg gacgtctgct gcagggcttc ccagagctct gatggcagcg tcggcctgag 6360 tccttcgagg agggctggtt tgtacgtggc atttgctgcc cactggactg tgaacttctg 6420 tctttttatt tcccactgct gctgtggtac atctccagta gcatagtttg gaaatgcagg 6480 ttttgataga ctcaaggatc taaatagaac cctcttagta ccaaggactg tccggggtct 6540 ctgccagccc cgccgatggg cctaactgtg gtgcctcctt tcctgtgaga atcttctgag 6600 gacatgcccg gggaaagagc tcagttctgc tgctgcctag ggtgccatgc tggccccggt 6660 tccaatgcag agcctagctg gaagtaccgc tgggttggcg gaggctacgt gcctgactgt 6720 cccctcgggg gtggggtgga actagccttc tgaaaccgcc tgcttcagtt ggccacagct 6780 ttttgaaatg tgtgtttctg gaagggactg ggtcccttcc ttgcctgttc agctccccac 6840 gacaaatgtc ctcaaggcga ggctggatgc ttccttcctc aggctcctag gaggagcccg 6900 tcccccagct gtgtcgggca gctggtcacc agcaaggaca ggatccctca gctgcagcct 6960 caggctggct ggcactgggc gggtgtttct gggatgagtt gtgtgtactg gagatgggag 7020 gggagctgag agggtgggat gcacagacag gagaggggac tgtgggggtc ctggaaccct 7080 gagttccaag tcttcaggac tctccctcca tagcaagtta cagggaagca gatttgagcc 7140 acagggaagc agatttgagc tgcagcgagg gggagggttt tcagtctgtg ctatagggaa 7200 gtgggcagtc ggcatttctg gtcctgggaa ctcactgggc agggctgcct tgggacatca 7260 gggaggtggc gctgtgctca gcttcaccag gaggggcctt aggcctgggg acggagagtg 7320 atgcctgagg cccctctact tctccatgga tcctgggagg gactcctggg ctggatacaa 7380 aattgttgag agttaagaga tctgtgagga aggggaggct gggaatagaa agtgtgtgcc 7440 cactgcacat ggggtccgca gggccacgtg cagccactgc gcaggcacaa ccccagtccc 7500 cacagagccc aggaggggcc agagccatgg aggaggcagc actgggcatt tggacaggga 7560 gggggtggtc agcaggcagc aggcccaggc ctgtctatgc cctgcggggt gcagcctcct 7620 gatctccacg gcaacctgga gcacccagcg tcagaaccac cgggagggct tatggaacag 7680 atgtccagcc ctgcagaagt tctggctcag gagggcgggg tgggcctggg aatttgcatt 7740 tctgactgta cagggcgatt ctgctgctgc tgctgctgct ggggttgggg gaggatccca 7800 tttgagaagc gctgcagtcc taggttgaaa cgtgcctgtc tgtccccacc caggcctgca 7860 tgggcagcac gggatcccca ggcaggagga cccaatttca tggcctggcc agccagggtc 7920 ctggagccag gcggtggggg agggatgggg gattgctgtg ccaccttcct tcccggcttg 7980 gcccgggggc aagcatcctc acacttccca tgtcgtcatc cccttggctc cagcctggct 8040 gcctctctaa ccctgctgta ccggctggcc gcatggccct ggctcttttt ggtgagcgtg 8100 gtccaggact ggtgacctgt gagtcctggg cccgcagtct tgcgcccctg cccgaaccaa 8160 cacaaatctt gttttctctc tctctcttcc ttcctcactc cctccccttc tcacctttcc 8220 ttttctgtaa ggtaagctga cttcctcttt tggtttttta tttattttta ttttttagtt 8280 ctgtaattaa aatcctaaca gccatggagg gtgtgggcac cgggggctgg ggccaggccc 8340 ctctgacctc tgagggggaa tgctgggtga ggcaggggcc ccgctgctgg gaccaagtat 8400 cctcaggggc ttgtgggcag aaaggcctgt gctggcccca gtcagtgcac agaagcggcc 8460 ccaaggccag ggctgctggg cagctcggaa tgagggcgag cagggctgcc cttggtgcct 8520 gagccaagga gccaatggga cagacctctg agcctgggtg ccaagtatga ggtctgagac 8580 agggtgagcg cctgggctgg gacaaggccc tctgagtggg cggccagctg cagcccaccc 8640 acccctaccc caggaaggca gggcccggga gggcatgacc tctggggtgc tggctcagct 8700 gcccccaccc caacctgaca ccgctagtcc tgagttccca tcagggagga agcagcatcc 8760 tgccttcctc taggaagagc ttgcatgtgg cccagaagcc aagggggctc cccagcaccc 8820 acgggcatct ctgggtctgg tcagaggaga aatctggatg cttgcaggag ccccagggtc 8880 atggaggagg ctggagacag ggctgtcctg gggtgatggg atggcccccc cacctgctca 8940 gagccagcct gggtgctgga accacacttg cctcaggacc ctgggcttgc tcctggggaa 9000 agagtggggt caggcaaagg ggtggggttg cgctgcagcg agacccaggc ccatcactca 9060 ccataccttc ttcctcccca tgcagcagcc aagagtttac tcaacaagaa agcagatgga 9120 gtcaaggtga ggctccagcc gggccctgtg gtgccgggga gcccagagcc tgcagcttca 9180 cccccacgcc ctggggctcc tgctctggag tccccctccc cccatgccct gagagacacg 9240 ggacagggaa tggcgagtga ggggcttctc ccacctaaga gttcctcttc cctctctcca 9300 cagccccaga cgaatagcac caaaaacagt gcagccgcca ccagccccaa agggacgctt 9360 cctcctgccg ccctggtact gagctcctca aattctgcct ctcagcccct cctacgcccc 9420 tggctgtgtg attgccgctg gtcagagggg gccgggtgaa ggtggggtct ggccccgcct 9480 ggcctgtctg acagcactcg catggccccc gcccctcatc cctcaccggt ggtgaagtgg 9540 agagaagagg ccactgttgt ggggggctcc aattcagaca ggtttaggac tgctctgggg 9600

agcccctggc tgagacccac agatgttggg gtgcagggga gaggcccagc ctcccaccca 9660 tgttgacttg tggatgtctc tccaggagtg ttcaggaagt cagtgaggca gaagataccc 9720 tctccccacc aggaccccac cctcagctcc tccaccatcc tcaacaggcc gacccacaga 9780 ccactccgaa ggtctggctt ggtggggctg ggccaggatc tgcaggggga acagcccata 9840 gtggcacatt ccacggccca tggggagacg gggccacggt ggtgcagtag agaggtgtct 9900 aagccagtgg cagccaaggg gagggcttgc cgtcacctct gtgttccctc agtgctgctc 9960 tgtggctgcc tgagaggcag ggcttagggg ctccctgccg gggaggggag gggtccccac 10020 catgctccgc tccaactgcg cccctcagtg ccccttgccc tgggggctcc tacaggtgaa 10080 ccctatagca gtactcccaa ggatgtaaag ttgtggctgg tgggtgccgg ccttcctgct 10140 ggggcgctgt gctgtgtccc ctcagctgtc ctaagagctt tggggcttgc tggcccgtag 10200 gtccccatat ttgctggaag caggcttggt gtcccctgag aaccccaggc caggcttcgg 10260 gagccagccc cagaccgccc acgggaatac tgggtttgcc aaatggccac cttgagaccc 10320 aggagaggag agcggtcctg ggaggggcga gctgctcaga gcagccaggc cgtggctgga 10380 gggtggcctg gtgcagccta cctagggcct tccagtggcc agggcagccc acgtgccagc 10440 ctcacagcca gccccatctc ggaccctgtc catcccatgt gccaccgcca cccccatgac 10500 atcttcaaac ctgtgccccc caccacgctg gggcacaggt tcaggcagta aagggtaggg 10560 agaacccctc aagaccgagc ctggcttctc tggctcccac acacattgtg cagcttgtcg 10620 gggccccaca cggtccatct cccaccctgg acagcagcac ctccgccagc ctggacagag 10680 ctcctgtcca ttccatccct gccggctgac ccaggctcct cccccagctg ctccacgccg 10740 cctccatccc tgtcccccac tctgctctgc acttctttct cgcaggctct ggccacccac 10800 acctcctctg tctccctgtt cccctcctgg tggtctccgc ttcctcctct tctcactttc 10860 cctctctttc cttcctctgt gtcttccttc ttctgtagga gcctcaaacc accgtcatcc 10920 ataacccagt ggacgggatt aaggtactgc cccactttcc tcctcccgct ttccccaggc 10980 aggaggctcc aggccaggag agaggtctgg ggcagcattt gtgccagagt ggagggcaga 11040 tgtcccatgg ccctggccgc ccctccccgc agtacggtag ggccccagtc cgtcttcgtg 11100 ggcaacaaca ggacagactg gctcaggccc caggcgcgcc cctggaggtg cttggcacag 11160 ttgcgcccgg tccccatgtg gccgacactc tcagaccagg gctctgcgtg tcccacctac 11220 ggcaggcagt agggcttcct gaggtctgga gcagggcctg catctcagga gctgcatcct 11280 tggccctcct ggctgtcctc caccccacct ccctcacgtg gcccccagtg cttcctgctg 11340 agcagaccct ccctcctctg ctcccctctc tgctctggcc atcagctccc atcacattgg 11400 catcatcact ctggggccag ggaaggggct ggctctctgg ggtggtggga gggatggggc 11460 cagcagccaa gccatttcca ggacttccaa aacagcgcca ctacacccaa cacggccctc 11520 cagcccagct cccacctagg cctgggctcc ttacagagcc cccagagtgc ctctgtgggg 11580 accccccact tccttctggc cagtgccacc acccagccca tcatcagaag acatctttct 11640 ccatggcagg gaccaggggg tccaaggggc acccatggtg ctaggcacca gggcctgggc 11700 attcttccca tctggcagct ggggatgggt gcccctggga cccgtgtgtg tctggggtgg 11760 gtcatgctct ctgcaggact cctaaacaac cttctgggct gtggtgaact ctgagcctgc 11820 acctaaaaga cctgtagttc tggtctaggg cctccaagca gtgtccaggc agtgtccaga 11880 ccagggggcg gtcccccagg gaccttgtaa gatgtttcct ctgaggagca gagcaggcct 11940 cctggggacc tgggggatgg tcttttgaag ggcagcagcc ctggagcagg gtgggagagt 12000 ctggggccac ctctgccctc taaggccacc tgagaggtga ggccggggcc tgactggacg 12060 tccagtccca gaggggcagg tgccctgagg gaatgtgggc gacaggaatg ctctgcctgg 12120 ggccaggcca aggttcctgg agccctgtgc ggatctgcag agctcctggg aacgcctcac 12180 cctgtatttt ggatgacacc ggctgctgct tcattggaac cagccagtcc cattgtgttt 12240 tacgtcttgg aatttcaaaa agcccatttt cctctcttgt taaagagtca gctgagcata 12300 ccagtctctc tgccaggctc atcttgctgg gagaagtgga gccctcatgt gttggggatg 12360 cagggtggcc acagcactag ggtggcaggg ccggcctcgg actccgtgcc agcctgtgct 12420 ggctgccgtg agaatgcacc ctggtgaggg gcgccctccc agggaccagc acagaactgg 12480 gtgtcttctc cggtcactgc cgcatgaggt ccacagagct ggggccctgc agccgccaga 12540 gggcatgtcc cctgagcccc tggcctttaa gccccgtgga agcagccgag gcagagatca 12600 gcttcagagc ctgggctggt cctgacacag gcccagccct gtccacctgc cctcagccac 12660 gtcccaccta tccttggccg catcctgacc cgctgcctcc cgtgtttcct caggagtctt 12720 ctgacagtgc caataccacc atagaggatg aagacgctaa aggtacctgc acttgagtcc 12780 ttgccccccc agcggccttg gcattgctgg gttgctcttt gaggtgggtg ggacttgggc 12840 agggtcaact ctcctgcgac gcctagttta tgcatgtgtt gaggggctca gggaccctgt 12900 agctgtaatc ctgctccaag cctgggtgtc aggcctgccc agagcggaga agcatggcag 12960 agatgaccga cagctgggca gtctcggtca ccgcatccaa gtgaggaagc cacggctttg 13020 catggaggca ggttctccac accaggaccc tcacggggaa acaggcccat gggtagaatt 13080 tgttccaaga tgctgtcctt gtcttaaagc tccttaagct tgcgtttctg tccagcatgc 13140 acttgccaag tggccgggca gctgggtgag tgtttccgtg tttgcctttg cttagccagg 13200 agtgtcctgc tgcggtgggt ttctgcacca cagattccag ggccccctcc cttgctcacc 13260 caggccaatg tcttgtgtgt tccccaagag gcccccaggg caccaggcac tggggcatgc 13320 tccatggatt ctgccgcctc cagaccaccc acatggggcc tcctgaccct catcgctcac 13380 acggtcacct aataagcctt atgctgttct cagggctacc ctggtgccca aaaagggtca 13440 gccactctgc cagtttaggg gagaaaactt ctcacctgtc caaagcatag ccttgctcct 13500 gcccggccta cccagctatg acactgtccc tgagcagaga tgagcacagg actttgggcc 13560 ctggatgccg gagagtgggt gtttgtgtga ttcccctgca gtctggaaca ggccccaaag 13620 gcaacagcat gaaggctgtc cagaggttct ccatcaccct cagccgagtg gggtgctgag 13680 cagtgaggga ggggacctgg gaggggggcc cagcctggat cctgcagggg agaagagaag 13740 acagccagaa gccagcagct gtggctcaga tctgagcccg agcagcctct cgaggtggag 13800 gcagacaccc cccaccccac cccgtgcaga aagaagcctt gccagcctgc cctgaggctg 13860 gtacagagtc caggcaggct cagtggccat catgccccta cgatgactgt cactccctct 13920 ccgtgcgcct ggcctctgct ggctctggcc aggggtggtc acagcactag ggtggcaggg 13980 tggcctctga ctctgcgcca gcctgcactg gcctgtgctg ccctggcctc tgctggctct 14040 ggctctggca ccggtcccgt gttggctcct tcagccttca catacctgct gcggccacca 14100 caggcccagg acccccacag ggtggccacc ccacctccac cccaggagcc ccaggtatcc 14160 agctgtcacc ccctccctcc ctcctggcct ccccctgtcc ttctccagtt gccttctttt 14220 cctgcgggcg caccacccac ctgcctgcct cacctgttcc gcctcagccc ccagggtccc 14280 cgacatcctg agctcagtga ggaggggctc gggagcccca gaagccgagg ggcccctgcc 14340 ctgcccatct ccggctccct ttagccccct gccagcccca tgtaagtagc ctgggtcctg 14400 ctgctgtggg ggtcatgttg gagggctggc aaccccctag aggggccact ccagagccga 14460 gggcaggctg agcgtggacc ctggctccag cctcatcacc ccacaatccc tcactggggc 14520 tttccagggt ggccccagcc catcgagccc cacctctttg tgaggagggc cctggaccac 14580 tttcctgctc aaggccactg ggcaggatgg gaggccctgg aggctcgggc ctcaattcca 14640 gtcttcaggg tcggtgcagg cctcactcca cctcagcttg cgggcggggg ggctccctgc 14700 tattgaggca ggctctgatt cagggcctga tcccagggcc caaggggtct agaacacggg 14760 acccctccca ctggcctcct ccgccttgcc gccgcctcgt gtgtctgtct gcctcatgtt 14820 cacgtctcat ctgttccacc ccagccccca gggatctctg acatcctgaa ctctgtgaga 14880 aggggttcag gaaccccaga agccgagggc cccctctcag cggggccccc gccctgcctg 14940 tctccggctc tcctaggccc cctgtcctcc ccgtgtaagt agtggccccc aggcctgccg 15000 cctctgctgc cggacagctc cctgcgaatg gccggcgctc agcagcttcc cacctgcatg 15060 cacggcccag ctaccctgcc ccggcgccgc agcctggagt cctgccctgg cggggcttcc 15120 tgtgggctcc catgctaacc agcagggcag ctcctggctt ctccctaagg ggcccagacc 15180 cctccacggc tcctgctccc actgccactc cccgctcgct gtccagcccc aggcccctct 15240 ccaaaatgtc tgtcccagcc ctgggcagcc ctggcccctc cgaggccccc catgccccta 15300 ggccctctct gctgatcact gtcccagccc cacagacttc acacccaccc aggggccctg 15360 cccatggtgc ccaggagctg cactcagggc caccctggtt cctgatgtgg ccccaacccc 15420 tgagcaccct ccctcagtct aggaggctga ggaaggtgcc aaaactggaa ccccgaccag 15480 ggtctctgga gctcaccaac aaggggatag tacggagaat cataagcctg gcctctgctg 15540 acctgggctg tcctcatggg gccaggccag gcctcctctg taacgcccgt gactccctcc 15600 tctccctgta accccgtcca gcgttcctca agggccactt acctgacagc ttcttgctgg 15660 ccagcagcct ctccctggag ggtgccctct gcccccagca gcttcagccc acgccacccg 15720 acagccagag catctgccct tcactcctgc agcctcctct ccacgcacca cgctgtccgc 15780 agcagcaccc tctgtccccc tgtctccctc cgtcccccca tatccccctc ggtcagccta 15840 caacctctcc acgtccccct aagtccacgc tctatcccta catccccctc tgtcccccaa 15900 attcccctct ttccctcatt tccattttcc tccccaaact ctgctctgcc cctcacattc 15960 tccctctgtc ccccacaccc tcctctgtcc cccacaccct cctgtgtccc ccacaccctc 16020 ctctgtcccc catatacccc tctgtccccc acacccacct tggtcccttg cacgcccttt 16080 tctgtccccc acaccccctc tgttccctac actctccctc tgtcctccag accctcctct 16140 gtcccccaca ctccctctgt cccccacacc ccctgtcccc cacactctcc ctctgccccc 16200 cagaccctcc tctgtcccct acactccctc tgtcccccat atccccctct gtcccccaca 16260 ccctcctctg tcctccaccc cctgcccccc ataccccctt ctgtccccca cacttcctct 16320 gtcttccaca ccccctcctg tcccccacac cccctctgtc ccccagactc tccctctgtc 16380 ccccacactc cgtctgtccc ccacacctcc tgtcttccac acccccttct gtcccccaca 16440 ccccctctgt cccccatact ctcctctgtc ccccacctcc cctctgttcc ccacaccgct 16500 tctgtccccc acaccccctc tgtcttccac ttcccctctg tcccccacat ccccctctgt 16560 cccctgcacc ctcctctgtc ccctgcaccc tcctctgtcc catgcacctc tctctgtccc 16620 ccacatcccc ctctgtcctc cacactccct ctgtccccca catccacctt ggtcccctca 16680 cgcaccccca tcccccatga ccccttctgt cccccacacc ccctctgtct tccacacccc 16740 cctctgtccc ccacacccac cttggtcccc tcatgccccc catcccctac acccccactt 16800 tgtcccccca catgcccctc tgtcccccac gttcccttct gtctcccacg tctcctccat 16860 ttcccgtttc cctctctgtc ccccaagctc ccctccatcc cccacatccc cttctttccc 16920 ctatatcccc tctgtcggcc caggtccacc atcttccccc cacacccccc cattctccct 16980 tcctcccctc tgtccccttg tgccccatcc cccacatctg cctctgtgcc cctcaatctc 17040 tggcttggct gtctgcccat ggtttctctc ctgcgtgccc cccgtgcctg ccttgtgttc 17100 acgtctcgtc tgttccgccc cagcccccag gatctctgac atcctgaact ctgtgaggag 17160 gggctcaggg accccagaag ccgagggccc ctcgccagtg gggcccccgc cctgcccatc 17220 tccgactatc cctggccccc tgcccacccc atgtaagtag caccttgagt ggccgtggca 17280 gcggctgcct ggaggggctc ggggcgtgcg agcctggcag tggtgctctg ggaagggcca 17340 ttcttgcgga ggagggcggg gcacaggatc cctctgctgg gtcccaggga attgctttga 17400 agcacatgaa ggtgccactg ggtctcagaa aatggaggtt atggttatga agtgtgtatg 17460 acatatgtgt ataggaagag cgtccgaaag agcaggtttg ttgccgaccc cagcattcgc 17520 aaccctgagg tccacagctt tctcctgatg ggaggggaat gggtggcaaa gggtctgcgc 17580 gtgtggcaag ggctagcacg ccaggagctg ctggcttggg tcaaggtgga cctgctgggc 17640 cgggacagaa aagtgtcagt cccggcctga gacgctctag cattagagct gtccaagtcc 17700 agacagcagg gagcaggtgg ggatcgggag gcgcggatct ggggggcagc tggggccagg 17760 ctgaaacaga gcgggcggga caggaagcac aggctgggca gcctccccgg ccagggagga 17820 gccaggctgg gccacctccc ggtctgtctg ccgactaccc gcagtatcac ttacagggat 17880 ggatgacatc ccagggctgc tgccaccccc acctgtgggg agacaccaga ctgggggtgg 17940 tgtggagata ctcttagaga agaggctgct gggccacggg ctcggcatgg cagggcagtg 18000 gctaggtaag tacttgaggg acaggtgggg tctgcttgcc accgtcccct ctgcaggctg 18060 ggcctggggg ctgctgcagg cggccagggc agaagggtgt ggggagagtg aacccacagg 18120 agcagcggct cgaggagggg gatgcaggct gcaggctcaa aggggcactg gatccaccct 18180 gggtgcccga gagagcaggg ggcagcccct ggaggggtac tcacccccag agcttctgtg 18240 gtcggctgag gacccccagc aggggttgac tgaggggatc agaggcaagc agctgagggg 18300 agaggccagg ttcttgatgc tgatagggtc ggggtgcctg ggcgaccaga actcaaggag 18360 ggaggcatgg ggaggggccg ccgtgcagct ggggtgggtg caccgcagag cctctgggag 18420 tggtcagaac ccccgacacc tgccacttct acagcagctc atctgatttt aaggggcttg 18480 ctgcccttgc agaagtggag gggtgtgccc aaaggagcct gcctggaagg tcaccccatc 18540 aggttggcat gaccccagcc caggactgca gcctgccctc aaggtctgtg cagtatctgg 18600 ggtgagtcct ctgaggacag ggcccagggt gggtgtggag tggccagctc ggggctcggt 18660 gtccaggctc accttcaggg gccacagcac agacctgccc ttccagagtc ttccctgagc 18720 ttggctgggg aggagggggc tgcaggaagg agctgtgagc agggcaggat ggagattcgt 18780 gtggccctcc tgggaggggc tgggcagggc tgggaaaggg gtgggtgaga tgttccggaa 18840 ctcagggaaa ggaagagtct gggtactgcc ctgggggcac ctgggcccag gtggcaggtg 18900 gccagctttc tgcctccttt ccacctcctt tctccagaag gcacccacca gctgtgtaaa 18960 tagggcaggt gcccacggcc cgcctcaggc cccgtctcct ccccacccac gctctctaat 19020 cgcggattat acacaatcca gcctgatccc tgggcagctg ccctccctcc cgcagccacc 19080 tctggctctg agagatgggc ttggggccag cctggggtcc caggagtcca ggccaggatg 19140 agaacctgct ctgaccccac ctggacgcat taggcctgcc tggacctgtt gcctcacccc 19200 aagagagcca caggcaatgc aaaggctcct gttcatgtca gggcacctgg aaggcctgac 19260 ttgcagaggc tcttggctcg tgcagacccc tccaagccca ggccctgccc accacctccc 19320 ctttgtctct ggaactgcca ggacagcttg tcctcagcca gcaggtttcc cgacccgggc 19380 acctcttcat gttgggcccc cctcctttcc ctccatcagg gatcatgccc ttcttcaggg 19440 gcctggatat caaggacaca aaagctccca tgtgctatgt ggggaggcag agtgggggct 19500 gggttgagct ggggtctggg cagcgccatt ccgcagggca ggggcagcct aggcttccca 19560 tctgtggaat gggtgggtgg gtctcacaac ggacctgctt cccgtacttc agcacggtta 19620 ccactcttga ttggaactct gaccatgcat ctcctcttct gtttacttca cgctttctct 19680 tcccatcaac tcccatttta attacaattt gtttaaaagc actgcatatt acttcattaa 19740 acagaagatt agtttcactt accattagtg taaggtgact atagaaccaa agcagactgg 19800 aaaccaaatg acataatgtc attctcttct ccattccagc tgcctgctgc tgtgcgcctg 19860 agaacccctg tggagtggga ggggcagctg tctctgtaca ttagaaaggg aggttaacta 19920 agtgacagga ggtgtttggg acatgtggac accagacttc tctcttgatg caaggagggc 19980 agagccaggc agcctagtgg gggctggctt gggggctgct ggaaggactg gctacaggtg 20040 gaagagaggt cagacctgaa gcttggggcc acctccagga aaggacaggt gaaagtggag 20100 gcatgaggca ggggagaggc aggtgccagg cagagggtgg agaggaggca ggaacatagc 20160 agctggggcg ggggcgggcc ctcaagtgtc atatgctact ttcctggggc ccaggggcaa 20220 ggacaggaac agccacagca tgtgttggga cagagccctg tgccttccta gagctgggca 20280 ggtggaatgg ggcaggaatg ggactcgtgg tggctgcagc aggaactgga ggggaagggg 20340 cttctggatc ctgcagccta ccttcctaga ggccagcttt ccggggtcca ccaggtgggt 20400 gggaactggg cttgtgtagc aagactgccc tgaggaccat ccatgacatg gtctagatga 20460 aagttaggaa agaaagggag acaagctggc agcagaagta cagctgggtc aggagcaagg 20520 gcctttccag atagggacaa cccaagagtg cacatgtgcc cacgccacac aacacaggca 20580 cacacgacac gtgcacgctc ataggcactg cacacacaca tgcacaggtg ctcatgcata 20640 tgtatgagct tcatctacac acattcacat gccgtcctgc ttatgtgcat gtttccatac 20700 atgcacatga atgcacaatc acgtgtacac acatgcatgt gatcacatac atgaacatgt 20760 gtgcacccca ctcctcaggt gccatcgggc tcctcctgct gtcactgtgc agcaggggac 20820 atgaggcccc agagcagaca ggtgcagcac aggcgttccc aggcagtgcc ccacacacat 20880 gcatgagcac acccgggcat gtggcgcctc ctttgtggac tcagtccacc tgccaggtgg 20940 gctccctggt ggtgtgagct cccagaggtc tggcgagaga gataaaggca accccaccac 21000 caggcgtgct gagaattccc tcttctggct gggcacagtg gctcatacct gtaatcccag 21060 cactttggga ggccgaggtg ggcagatcac ttgaggttag gagtttgaga ccagcctggc 21120 caatatggtg aaacctcatc tccactaaaa atatacacac acaaaaatta gctgggtgtg 21180 gtggtgtgca cctgtagttc cagctactcg ggaggctgag gcaggagaat cgcttgaacc 21240 tgggagtcag agactgcagt gagccgagat catgtcactg cactccagcc cgggtgacag 21300 agtgagactc catctaaaaa aaaaaaagaa ttccctcctc tgggaattta gaccacagac 21360 aggttgcatg tatgtggccg ttggaggcag cactcacagc aaagagtgga aacgtcacca 21420 cagggcctgc cttctggtga aaatggtgtc ctgcagggcg ggcagctgtt tgagggcagg 21480 tgtcccaggt gcggcctgca gcagcctgag ggtcacagag cgcagtgctg ggagtgcaga 21540 gacttccccc acagggagag ttcccaggaa cctgcttccg gtgcacttct gggggtttga 21600 gttttttcca cggacgaatt actttgagaa accactgtta ctcgtgtgta taggtgagcg 21660 tgcgtgtgca tgtgtgttct gtgtgtgagt gtgcatgtat gtgcgtgcct gcgtatatat 21720 cctcgcagat acggctaggg acctcactca ggacagtagt tctgcctgag gagagtgaat 21780 gcggcaagat tgaggagaac acaggcatct tcaaactaca tgtgcggtgc tttatttctt 21840 taaaaatgcg tctaaagcaa ataggaaaat gttaagattt gaatccgtag agtgtgggtt 21900 ctattattct ctccacatct tccatacgtt taaaatcttt tgcaatgaaa ataagctgta 21960 gttaaagcag caatgcaggc tgccagtgag cgccccggag gccagtgagg accagcatgg 22020 ctgggtggcc tgttggaatc caaggggggc gggcaggagc tgcaggcagg cgcccgggag 22080 tagcccgggc atgggggtgc ggggcaacag ggatgtctgc aggggtagca tgtgggcccc 22140 ggactgcaag caggtggagc cagccggatg cggctcctat gagaaaagcg gggaacaaga 22200 gaccacgctc gttcttcctg ctgcggggac agccctggtc atcgctccgg ggaaccctgc 22260 agcctgcgcc gcacgtggcc gccccctgct gcttcctcct ccccggcctc cgggtggcct 22320 tgctgacggc tccttctctg aggcaggtct ctgccttctc gcctggtgcc tgcactcagt 22380 agccccctca ccagagctgc tgggtgaagg aagcactaag aacccaaggc tcgggaggag 22440 agtggggccg ggaagctgca gggaagcgca gggccaggcc tggtgggccc aggggctggc 22500 tcacgggagg gcaggaggga gactgtggcg gacagcacgt ggggccagga ggtgacctcc 22560 aagtggattg tgggtgggtt ttttgtcctc tttctgcatt ttccaggcat tttgtaatgt 22620 ggatagaata tttctgttct tcaaaaatac tttagttaag aaaaataaga tggaagctgt 22680 tgcacttgaa aatgaggaag ccactggtga tgcagggggg gcggcggaga ggacctcttc 22740 tgcaaatagc ggcaggaaca cggcatggat gcagctcgcg ctcccccagg ccctcccctg 22800 ggctgtgtgg aggggtccgg ggggaatggg ccagcgccca gtggtcacct ggccatgtct 22860 ccccacagcc cggaagcagg agatcattaa gaccacggag cagctcatcg aggccgtcaa 22920 caacggtgac tttgaggcct acgcgtgagt ccctggggct gggggggggc tgtgcaggac 22980 aaggatgtgg gacccttggg ggggcctgct cagagtcagg ggtccacggg gcccctcctc 23040 acttggattt ggcccccagg aaaatctgtg acccagggct gacctcgttt gagcctgaag 23100 cactgggcaa cctggttgaa gggatggact tccacagatt ctacttcgag aaccgtgagt 23160 gaggaagccc gggtgggcat gagggggcgg tgcccccagg agagcctctc ggcccctccc 23220 agggacagca tggtggctgc ctatggaagc cctgtcccct ctgtgcccag ggttggccag 23280 ccacctctcc cccgccagag gccataccca gcccccagaa tcccactctt ggaggggccc 23340 atgctgctcc caggagagcc gagcctcccc aataagggga gttgagagag ggaaaggatt 23400 aggctggtgg ggtggaagac gggcaccagg gcagtcatgg taacccgaga cccccgcccc 23460 gcctgctgtc cacagtgctg gccaagaaca gcaagccgat ccacacgacc atcctgaacc 23520 cacacgtgca cgtcattgga gaggatgccg cctgcatcgc ttacatccgg ctcacgcagt 23580 acattgacgg gcagggccgg ccccgcacca gccagtctga ggagacccgc gtgtggcacc 23640 gccgcgacgg caagtggcag aacgtgcact tccactgctc gggcgcgcct gtggccccgc 23700 tgcagtgaag gtgagtgttc tgtgctaagt gacagctggg gcagaggggt ggcggtggtg 23760 tgagtggctg cagcctgggg aggcgatggg gagcggtggg gcctgtggca gagcccatgc 23820 ctgggaagtc cctgagcttt cctggtgagg ccacaggaat gatgtcaaat tagggaccac 23880 ggcaggctgg gtgtggcagg cctccccaga ggactgggga gctggtgagg gcctgagcag 23940 tccacactgg ccagagctgg gtgggttgca ggtggatggg ccccgggcag cacagtcctg 24000 ggcaccatgc cctgtttgtg aggactgtta gagccccaga tgggcgttcc ccaggtggtg 24060 ggtgcagcgg gcccagagcc cagttttaca gggatagtag taattgggtt gggcaccttg 24120 aacctctctc ccgagtgggc ccttttctgg actttaaccc tctctgcagt gccgcatggc 24180 agacagcaga gcctgggggt ggatgggaga gggggctgct gaggagctga cccacccgcc 24240 ccatttcaga gctgcgccct ggtttcgccg gacagagttg gtgtttggag cccgactgcc 24300 ctcgggcaca cggcctgcct gtcgcatgtt tgtgtctgcc tcgttccctc ccctggtgcc 24360 tgtgtctgca gaaaaacaag accagatgtg atttgttaaa aaaaaaaaaa aaaaaaaaaa 24420 aaaaaacaag atgacgacga caaccacaaa aaaaattgac atcagatgaa atgaaaaaaa 24480 aaaaaaacaa aaaaaactaa aggaaggaaa aagctgtaaa aatcactggc attcgtgggg 24540 ccactcccca cccaagctcc acgtgtgtcc gtctgtgctc ctggcctctg ggggaccagc 24600 tgggacatga acttgtctgc caggcccccg tcgcgtgctg aacggtgtta gtttgtaggt 24660

aacgcacaca ccccacacct aaggtgtctg catcctcctg ccaacgcatg ggctccacgt 24720 ggtgtgctcg ctggctgtcg tgactgtcag ctgtctcttg ggaggggctg tgggggcccg 24780 ctgggctgcc tcctttcccg ctagttgtgc ctgagagttg ctgttgttcc tgctttccct 24840 tcccttcctt tcatcccctg aagggctagg tgtgggtttt ccgtgcccgg tatccccaca 24900 cacccagcac ggacaaccct tcggcagagc ccaggccggc ccctcacccc ctggagtatt 24960 gaaactggag tcccgtcccc aaggccttca gagatgcccc tacacaccca gggctccagc 25020 tctggtcctt ctgggggagt aaagtgcaaa gaggggcaca gcttagtttt gggcctctcg 25080 ccgagcaaga gacagcactg ctggctacag ctccaacaca gccagctgtg gcaagaggac 25140 tctgcctggg ctggcccccc tcctgtgtga ggtgtctgtc ccttctctgc tggccagcag 25200 cagatgcact ggcagctccc aaccctgttt ccgcccctcg gccctccccc agcctgttcg 25260 gcttctctgc agcccgcaag ggggagcaga cttttgacaa aggactgcgg gcctcgctca 25320 agtccctgag cccccagctg aagctgggag gggaggccag gctttgtgtc tgggcatatt 25380 cgtctgctga tggggtttgg ggaagcctgg ggcttggggt ttggtcgggt ggtgcagcta 25440 gtggcagagc gggatcagag gtggtggctg cccagcttct gggctgagac aagggtctgt 25500 gcaggggttt actgaagtgg gagtgccttt ggaatctggg ccgggagcag aagggagcaa 25560 aagctacagt gggagccagc ctagggcaca tgggaggcgt gagggcagtg ctgcccgtgc 25620 agtgtcaggt gtgccagtgc cttggcgggc tgcagtgcgt gtgagggcac cttctaggtg 25680 ggccagggat gcagctatgg agataaggcg ggctggggac agaaacaggt gggcacaggg 25740 cccaggacac cagcggatgg agggcagggt ctagccctgt gctcctgagc gtcggctgcc 25800 tgggttcgag gcggtgggtc cccggcccct tgtgatggtg tgtaccatgg gggagctcgg 25860 ggacagggca agcccgagca tggtggggct gcagggtggg tctgaagcca ggttgggtgg 25920 gggtggtcac aagccctgac tgcagagggt caggggctcc tgccccagtg cctgcccact 25980 ttcaattcac attgttttca acaaggattt tctttatctt cccctacaaa tcaagccaag 26040 ggaggggcac agaatgggga acaggacaca ggatcctaaa ctccaagggg actgtccacc 26100 gatgaacact cagagtggac accatcttcc gtccacgctg tgcccaggac agctgtcccc 26160 atccatgaac acagggtaaa catctgccgg gctccgcacc agtggctccc tgggccatgg 26220 gacagcggca gggctcacca cggacagcac gtggcccagc agccggccac cctggcgtcc 26280 tggggcctcc tcccctcctc tccctctcac cttgtcacct ccacggagct gcctgtctgg 26340 gataatttgg ggattttttt tctgggggat aattcttttg catgacccct aaagagcaag 26400 ccacaccggt ctgctagcta ggtgtccgcg gtgtggtggt ggcggccgct ggccagcgct 26460 gcaaggggtc ggctgcccac ggtgctggct ggcctcccct cctctctctt tttgctgagt 26520 ttcattgtct tttctttctg agccttgtaa gtgtacaaaa attattctta ttttgttctg 26580 tctcgggaaa ctgcaaataa aagaaaaaca ggacaaactg cttcaagtgc agctgggtgc 26640 tttagctgga atcctgccga cctcctgcgc caaaatacag actcaagccc ggtccctggc 26700 caagacccta cttgggcccc tcctccaatg aaaggtagtg ctatgggagc cctgagctgg 26760 ccctgacagt cctgagcccc tctagggtga acggctcacc ccaggtaggg cactagtcat 26820 agatcatagc tctaccagct gtctccacct cttcctctgg tcctctgaag tcttctgggc 26880 ccagcgctgt ccaccctgaa tgctggaact gaaactggat cccagccccc aacacccctg 26940 acctctccat tcacccccgg tggccgctaa ggatgtggcc agggcagcct ctgggcagga 27000 aggagcccca ggaccaagac ctctggctgt cctgctgttt ccttccgccc ctgctacatg 27060 tattggctat tctggatgct gaggacacac agtgaccaca gagccgggct ccaccccagt 27120 ggattatgca gacagatggc acgcaggcct gtgtggacat cagcctcggg caccagacat 27180 aggcaaggcg caaggtgata cagtaggcag ccaccatggg ggccaggagg ctccagcaga 27240 ggccacacaa ccagcccaga atccaggaca gagagctgga atggagacag ggaagccaga 27300 taccaggcca gactggccag gtgctacagg cctgtgggcc aggccaggct tggggacttc 27360 gtcctgggtg tgaaggagac aggcacccct gaggccttcc ctctgcatct ccagcccaag 27420 ctaagcgcaa actcttaggt tggagtaagg agtaaccccc tgccaagttt ctcctgtcct 27480 caggctccac ccaccaccta tgctgcctgg ccccatgggg cacacgctca ggcccagcct 27540 gggaaagcaa ctgcacctgc ctgtgctatg ctggcccttc tcagcctcaa tgccctcctc 27600 cctccccgac gcaccctcgt ggcccccgct gggccccctg atgcaccctc atgtctccat 27660 ggcaacctgc tcagagtgtg gccctgccct tggctcccct ccacacctgt gtcccaggca 27720 gtgccacggc actttcctaa acagaaggat gggcttcaaa acagtcccag acactaaaca 27780 cacctgcatt ttgggtccaa gtaacttctg acaagacgag tgcccctaca caccctcagt 27840 cctatccact atgggcaagg agcctgaagg atcccccaga actggctaaa gccctcagtc 27900 tcctcctcca ccctgagcac cttcacgcgg cagagtggcc ctggatgtca gcttcttgct 27960 ccccatggtc tgcacctgga caggtgctct caggtgtgtg ggtgggcagg tggcaggtcc 28020 caagagccag gtgcaaagaa tctaggccag tgcccacgag tgctgcagtg tctgtcccca 28080 gcatggtatc tagggctcca cttgcctatc agctgtaatc ggaggaggct ttccaggcca 28140 ggcctccccc aggaaggctg caggcactgc ggatcgtgcg ccctcacatg cattattcct 28200 gaggcccttc tgcagatgcc atcagggcag caactctgat gaggtattag ggcacagcac 28260 acagggctaa gccaccctgt actgggccaa gcgctacagg caaaaaggac accaccgacg 28320 ggcatttcat tcatcgcttt tatttttata tatttttgag agggagcctc actctgtcgc 28380 ccaggctgga gtgcagtggc gcgatcttgg ctcactgcaa cttctccctc ctgggttc 28438 4 542 PRT Homo sapiens 4 Met Ala Thr Thr Val Thr Cys Thr Arg Phe Thr Asp Glu Tyr Gln Leu 1 5 10 15 Tyr Glu Asp Ile Gly Lys Gly Ala Phe Ser Val Val Arg Arg Cys Val 20 25 30 Lys Leu Cys Thr Gly His Glu Tyr Ala Ala Lys Ile Ile Asn Thr Lys 35 40 45 Lys Leu Ser Ala Arg Asp His Gln Lys Leu Glu Arg Glu Ala Arg Ile 50 55 60 Cys Arg Leu Leu Lys His Ser Asn Ile Val Arg Leu His Asp Ser Ile 65 70 75 80 Ser Glu Glu Gly Phe His Tyr Leu Val Phe Asp Leu Val Thr Gly Gly 85 90 95 Glu Leu Phe Glu Asp Ile Val Ala Arg Glu Tyr Tyr Ser Glu Ala Asp 100 105 110 Ala Ser His Cys Ile Gln Gln Ile Leu Glu Ala Val Leu His Cys His 115 120 125 Gln Met Gly Val Val His Arg Asp Leu Lys Pro Glu Asn Leu Leu Leu 130 135 140 Ala Ser Lys Cys Lys Gly Ala Ala Val Lys Leu Ala Asp Phe Gly Leu 145 150 155 160 Ala Ile Glu Val Gln Gly Asp Gln Gln Ala Trp Phe Gly Phe Ala Gly 165 170 175 Thr Pro Gly Tyr Leu Ser Pro Glu Val Leu Arg Lys Glu Ala Tyr Gly 180 185 190 Lys Pro Val Asp Ile Trp Ala Cys Gly Val Ile Leu Tyr Ile Leu Leu 195 200 205 Val Gly Tyr Pro Pro Phe Trp Asp Glu Asp Gln His Lys Leu Tyr Gln 210 215 220 Gln Ile Lys Ala Gly Ala Tyr Asp Phe Pro Ser Pro Glu Trp Asp Thr 225 230 235 240 Val Thr Pro Glu Ala Lys Asn Leu Ile Asn Gln Met Leu Thr Ile Asn 245 250 255 Pro Ala Lys Arg Ile Thr Ala His Glu Ala Leu Lys His Pro Trp Val 260 265 270 Cys Gln Arg Ser Thr Val Ala Ser Met Met His Arg Gln Glu Thr Val 275 280 285 Glu Cys Leu Lys Lys Phe Asn Ala Arg Arg Lys Leu Lys Gly Ala Ile 290 295 300 Leu Thr Thr Met Leu Ala Thr Arg Asn Phe Ser Val Gly Arg Gln Thr 305 310 315 320 Thr Ala Pro Ala Thr Met Ser Thr Ala Ala Ser Gly Thr Thr Met Gly 325 330 335 Leu Val Glu Gln Ala Lys Ser Leu Leu Asn Lys Lys Ala Asp Gly Val 340 345 350 Lys Pro Gln Thr Asn Ser Thr Lys Asn Ser Ala Ala Ala Thr Ser Pro 355 360 365 Lys Gly Thr Leu Pro Pro Ala Ala Leu Glu Pro Gln Thr Thr Val Ile 370 375 380 His Asn Pro Val Asp Gly Ile Lys Glu Ser Ser Asp Ser Ala Asn Thr 385 390 395 400 Thr Ile Glu Asp Glu Asp Ala Lys Ala Arg Lys Gln Glu Ile Ile Lys 405 410 415 Thr Thr Glu Gln Leu Ile Glu Ala Val Asn Asn Gly Asp Phe Glu Ala 420 425 430 Tyr Ala Lys Ile Cys Asp Pro Gly Leu Thr Ser Phe Glu Pro Glu Ala 435 440 445 Leu Gly Asn Leu Val Glu Gly Met Asp Phe His Arg Phe Tyr Phe Glu 450 455 460 Asn Leu Leu Ala Lys Asn Ser Lys Pro Ile His Thr Thr Ile Leu Asn 465 470 475 480 Pro His Val His Val Ile Gly Glu Asp Ala Ala Cys Ile Ala Tyr Ile 485 490 495 Arg Leu Thr Gln Tyr Ile Asp Gly Gln Gly Arg Pro Arg Thr Ser Gln 500 505 510 Ser Glu Glu Thr Arg Val Trp His Arg Arg Asp Gly Lys Trp Gln Asn 515 520 525 Val His Phe His Cys Ser Gly Ala Pro Val Ala Pro Leu Gln 530 535 540 5 601 DNA Homo sapiens 5 cacctctggg tttaaacaac atgcaccctt gtgccggtca cctccctgca gccggagaac 60 ctgcttctgg ccagcaagtg caaaggggct gcagtgaagc tggcagactt cggcctagct 120 atcgaggtgc agggggacca gcaggcatgg tttggtgagt gccaggggca gggtgtgttg 180 gctggcagtt ggcagggcag gaggtgatgc tgacagcccc ttgtggcctc ttcccctctc 240 tctaggtttc gctggcacac caggctacct gtcccctgag gtccttcgca aagaggcgta 300 yggcaagcct gtggacatct gggcatgtgg tgaggcctgg cctgagttgg tgcggggcag 360 ggcctcgggt gtttcaggac ttcccaccta catcctggag tgtgcagtgg ccagcacgtc 420 ttgctctcat ctgggtttat ctgtgtcaga cctgcccttg agctgccctg gcaggggtct 480 gcccacacag ccaagagccc cctttccacc cagattagaa ttgctcacat gaacctggcg 540 caccccagtg ctcgcctgcg ctcagcagag gtctggtcca gaagtgtggt gggtggatgg 600 g 601 6 601 DNA Homo sapiens 6 gtttaaacaa catgcaccct tgtgccggtc acctccctgc agccggagaa cctgcttctg 60 gccagcaagt gcaaaggggc tgcagtgaag ctggcagact tcggcctagc tatcgaggtg 120 cagggggacc agcaggcatg gtttggtgag tgccaggggc agggtgtgtt ggctggcagt 180 tggcagggca ggaggtgatg ctgacagccc cttgtggcct cttcccctct ctctaggttt 240 cgctggcaca ccaggctacc tgtcccctga ggtccttcgc aaagaggcgt atggcaagcc 300 ygtggacatc tgggcatgtg gtgaggcctg gcctgagttg gtgcggggca gggcctcggg 360 tgtttcagga cttcccacct acatcctgga gtgtgcagtg gccagcacgt cttgctctca 420 tctgggttta tctgtgtcag acctgccctt gagctgccct ggcaggggtc tgcccacaca 480 gccaagagcc ccctttccac ccagattaga attgctcaca tgaacctggc gcaccccagt 540 gctcgcctgc gctcagcaga ggtctggtcc agaagtgtgg tgggtggatg ggagtggaga 600 a 601 7 601 DNA Homo sapiens 7 gaattcttgc ccctgcctga gagggagctt caggcccggc cggggcgctg tttccttctg 60 cagttcccgt cccctgagtg ggacaccgtc actcctgaag ccaaaaacct catcaaccag 120 atgctgacca tcaaccctgc caagcgcatc acagcccatg aggccctgaa gcacccgtgg 180 gtctgcgtga gtcgcccttg gtgcccatgg tggggagggg gctcctggtg gagatggcct 240 cagaccactc ccctggcaag gaccccaaga gggtcctgtt cctgacatcc aagagctccc 300 ytgggtcccc tgggtgctcc ttgtggcctc tggcttggga cataccagca cgtttgtgag 360 gcctggggct tggaaggcat tagagggtag aggtgatccc ttcctcccaa ctgcagtcct 420 gtctgtgagg ggcagagtgg acgaggcaag ggagagacga gtcttgaagt cccaggcggg 480 tggggacaga caacccttgc cgcaatggtg gccggtggct cttggcaagt ggggacccca 540 gggtgccaca agccttgcca ccctggcctc tcccctgtgc ctcgggctcg gctgccatat 600 g 601 8 601 DNA Homo sapiens 8 ctgaccatca accctgccaa gcgcatcaca gcccatgagg ccctgaagca cccgtgggtc 60 tgcgtgagtc gcccttggtg cccatggtgg ggagggggct cctggtggag atggcctcag 120 accactcccc tggcaaggac cccaagaggg tcctgttcct gacatccaag agctcccttg 180 ggtcccctgg gtgctccttg tggcctctgg cttgggacat accagcacgt ttgtgaggcc 240 tggggcttgg aaggcattag agggtagagg tgatcccttc ctcccaactg cagtcctgtc 300 wgtgaggggc agagtggacg aggcaaggga gagacgagtc ttgaagtccc aggcgggtgg 360 ggacagacaa cccttgccgc aatggtggcc ggtggctctt ggcaagtggg gaccccaggg 420 tgccacaagc cttgccaccc tggcctctcc cctgtgcctc gggctcggct gccatatgac 480 cacccatttc cccacagcaa cgctccacgg tagcatccat gatgcacaga caggagactg 540 tggagtgtct gaaaaagttc aatgccagga gaaagctcaa ggtgaggccc tggcccctag 600 t 601 9 601 DNA Homo sapiens 9 gtggagatgg cctcagacca ctcccctggc aaggacccca agagggtcct gttcctgaca 60 tccaagagct cccttgggtc ccctgggtgc tccttgtggc ctctggcttg ggacatacca 120 gcacgtttgt gaggcctggg gcttggaagg cattagaggg tagaggtgat cccttcctcc 180 caactgcagt cctgtctgtg aggggcagag tggacgaggc aagggagaga cgagtcttga 240 agtcccaggc gggtggggac agacaaccct tgccgcaatg gtggccggtg gctcttggca 300 wgtggggacc ccagggtgcc acaagccttg ccaccctggc ctctcccctg tgcctcgggc 360 tcggctgcca tatgaccacc catttcccca cagcaacgct ccacggtagc atccatgatg 420 cacagacagg agactgtgga gtgtctgaaa aagttcaatg ccaggagaaa gctcaaggtg 480 aggccctggc ccctagtccc aggcacggcc atgcttctct gtgtccctct gggctggagc 540 aggggggcct tggggggtct gggcagacct aggggttact gctgccccca agactgactg 600 t 601 10 601 DNA Homo sapiens 10 tctgggctgg agcagggggg ccttgggggg tctgggcaga cctaggggtt actgctgccc 60 ccaagactga ctgttagcaa gtcccagact ggatgcatca ggtgaactca ggccagcttg 120 ggaatgagtc cagaggggcc ctgggccagg tgtggctcct cctagttgtc tgtgccacct 180 cctagcagcc cttggaggag ctgtcctgaa gcgctcgctg tgggctcctc acccgggctc 240 tgcaggcagc actcaccctc tggcagtcac actgtttagt acaagcaagt ccgaagcttc 300 yggctcagac aggtttggta aggagagcag agccacacac actggtcttg ggtgggctgg 360 gggagttctg ggagggaggt gggtcccagt agggtatcca acctgcctgc tttggtcagg 420 gctggctccg gtgaccgcac actggcagtc cctctacttg tgggttccgg gatggggact 480 tgttgcctga ctgccctctg ctggtctctg agcagttctc cccggaagcc ccaggactgt 540 tgccctgtct gagcctgtca ggaaaagaag gggctgtcag ggagctggac cccagaggag 600 c 601 11 487 DNA Homo sapiens 11 gctaggtggc ccctgggcta caccaagccc ttctggtcct ggcccccgag gtctgggggt 60 ccggagaccc ccattaagaa tggcctgggc cccacaggga gccactgggc ctgctgctgg 120 ggggtctgaa tcctgaaagg agagccttga ggagcagagc cagagaggca gaggcccttg 180 gggcagacac acaccctgcc cctctggggc cgcatggaga cggtggtctg tgctgctgag 240 tcctacacat gcatgtctgc cctgagcatc cccccaggac aagccgctct ggagtgggtg 300 rgggttttat gcaccctgag gagactttca aggcttcctc ttgggttgtt tctgcaaagt 360 cctcctcccc tggcctcaaa ccctgtgagg gaaaaggccg gcactggcca cctgctcctc 420 tgggctgtgc ggggccagag cccagaggcc caagttggct tctgcccacc tgctggcttg 480 tgaccat 487 12 601 DNA Homo sapiens 12 cctcctcatg acccacaggg tgagcagcct ggccttccca gccagagaac cctccttctg 60 gggaggccca gggcgtcctc ggggagggca gtctattctc ctcccatgag cccagtggac 120 gtgtctagca ggcagcaccc cgggagagcc ctcccacgtc ttctccattt gacaggcctt 180 tccagagcgc aggcgggagg gggctgtgat tagaaaagag tgaggctagt ggcttctggg 240 gaggcactgc tgcccagggg acagtgctga gagacagctg cctctacgct gccctgtgcc 300 yggggctccc gctgcaatgc ccgcctgtct gcaagtgaac gtggggcgac ggtgcatgag 360 gccctgcatg tgtggctcca ccctgggcgc cgagagcagc tctgtcctgg agggtggtca 420 gtgcatgtgg acagagccca gcatggctgt cctgggtgac cagctaaggg gacaaggcag 480 aggcagggct gagaggacca cccatcctgc taggtcagcc cagctcagcc atatcacacg 540 gcagtgagca tggagctcag ttctctgcca atggcagctg agtctagtac catccagtca 600 g 601 13 601 DNA Homo sapiens 13 aaggcctgtg ctggccccag tcagtgcaca gaagcggccc caaggccagg gctgctgggc 60 agctcggaat gagggcgagc agggctgccc ttggtgcctg agccaaggag ccaatgggac 120 agacctctga gcctgggtgc caagtatgag gtctgagaca gggtgagcgc ctgggctggg 180 acaaggccct ctgagtgggc ggccagctgc agcccaccca cccctacccc aggaaggcag 240 ggcccgggag ggcatgacct ctggggtgct ggctcagctg cccccacccc aacctgacac 300 mgctagtcct gagttcccat cagggaggaa gcagcatcct gccttcctct aggaagagct 360 tgcatgtggc ccagaagcca agggggctcc ccagcaccca cgggcatctc tgggtctggt 420 cagaggagaa atctggatgc ttgcaggagc cccagggtca tggaggaggc tggagacagg 480 gctgtcctgg ggtgatggga tggccccccc acctgctcag agccagcctg ggtgctggaa 540 ccacacttgc ctcaggaccc tgggcttgct cctggggaaa gagtggggtc aggcaaaggg 600 g 601 14 601 DNA Homo sapiens 14 ccaggagtgt tcaggaagtc agtgaggcag aagataccct ctccccacca ggaccccacc 60 ctcagctcct ccaccatcct caacaggccg acccacagac cactccgaag gtctggcttg 120 gtggggctgg gccaggatct gcagggggaa cagcccatag tggcacattc cacggcccat 180 ggggagacgg ggccacggtg gtgcagtaga gaggtgtcta agccagtggc agccaagggg 240 agggcttgcc gtcacctctg tgttccctca gtgctgctct gtggctgcct gagaggcagg 300 rcttaggggc tccctgccgg ggaggggagg ggtccccacc atgctccgct ccaactgcgc 360 ccctcagtgc cccttgccct gggggctcct acaggtgaac cctatagcag tactcccaag 420 gatgtaaagt tgtggctggt gggtgccggc cttcctgctg gggcgctgtg ctgtgtcccc 480 tcagctgtcc taagagcttt ggggcttgct ggcccgtagg tccccatatt tgctggaagc 540 aggcttggtg tcccctgaga accccaggcc aggcttcggg agccagcccc agaccgccca 600 c 601 15 601 DNA Homo sapiens 15 acagcagcac ctccgccagc ctggacagag ctcctgtcca ttccatccct gccggctgac 60 ccaggctcct cccccagctg ctccacgccg cctccatccc tgtcccccac tctgctctgc 120 acttctttct cgcaggctct ggccacccac acctcctctg tctccctgtt cccctcctgg 180 tggtctccgc ttcctcctct tctcactttc cctctctttc cttcctctgt gtcttccttc 240 ttctgtagga gcctcaaacc accgtcatcc ataacccagt ggacgggatt aaggtactgc 300 yccactttcc tcctcccgct ttccccaggc aggaggctcc aggccaggag agaggtctgg 360 ggcagcattt gtgccagagt ggagggcaga tgtcccatgg ccctggccgc ccctccccgc 420 agtacggtag ggccccagtc cgtcttcgtg ggcaacaaca ggacagactg gctcaggccc 480 caggcgcgcc cctggaggtg cttggcacag ttgcgcccgg tccccatgtg gccgacactc 540 tcagaccagg gctctgcgtg tcccacctac ggcaggcagt agggcttcct gaggtctgga 600 g 601 16 601 DNA Homo sapiens 16 agtctctctg ccaggctcat cttgctggga gaagtggagc cctcatgtgt tggggatgca 60 gggtggccac agcactaggg tggcagggcc ggcctcggac tccgtgccag cctgtgctgg 120 ctgccgtgag aatgcaccct ggtgaggggc gccctcccag ggaccagcac agaactgggt 180 gtcttctccg gtcactgccg catgaggtcc acagagctgg ggccctgcag ccgccagagg 240 gcatgtcccc tgagcccctg gcctttaagc cccgtggaag cagccgaggc agagatcagc 300 ytcagagcct gggctggtcc tgacacaggc ccagccctgt ccacctgccc tcagccacgt 360 cccacctatc cttggccgca tcctgacccg ctgcctcccg tgtttcctca ggagtcttct 420 gacagtgcca ataccaccat agaggatgaa gacgctaaag gtacctgcac ttgagtcctt 480 gcccccccag

cggccttggc attgctgggt tgctctttga ggtgggtggg acttgggcag 540 ggtcaactct cctgcgacgc ctagtttatg catgtgttga ggggctcagg gaccctgtag 600 c 601 17 601 DNA Homo sapiens 17 acatcctgag ctcagtgagg aggggctcgg gagccccaga agccgagggg cccctgccct 60 gcccatctcc ggctcccttt agccccctgc cagccccatg taagtagcct gggtcctgct 120 gctgtggggg tcatgttgga gggctggcaa ccccctagag gggccactcc agagccgagg 180 gcaggctgag cgtggaccct ggctccagcc tcatcacccc acaatccctc actggggctt 240 tccagggtgg ccccagccca tcgagcccca cctctttgtg aggagggccc tggaccactt 300 ycctgctcaa ggccactggg caggatggga ggccctggag gctcgggcct caattccagt 360 cttcagggtc ggtgcaggcc tcactccacc tcagcttgcg ggcggggggg ctccctgcta 420 ttgaggcagg ctctgattca gggcctgatc ccagggccca aggggtctag aacacgggac 480 ccctcccact ggcctcctcc gccttgccgc cgcctcgtgt gtctgtctgc ctcatgttca 540 cgtctcatct gttccacccc agcccccagg gatctctgac atcctgaact ctgtgagaag 600 g 601 18 601 DNA Homo sapiens 18 ctgtcccctt gtgccccatc ccccacatct gcctctgtgc ccctcaatct ctggcttggc 60 tgtctgccca tggtttctct cctgcgtgcc ccccgtgcct gccttgtgtt cacgtctcgt 120 ctgttccgcc ccagccccca ggatctctga catcctgaac tctgtgagga ggggctcagg 180 gaccccagaa gccgagggcc cctcgccagt ggggcccccg ccctgcccat ctccgactat 240 ccctggcccc ctgcccaccc catgtaagta gcaccttgag tggccgtggc agcggctgcc 300 yggaggggct cggggcgtgc gagcctggca gtggtgctct gggaagggcc attcttgcgg 360 aggagggcgg ggcacaggat ccctctgctg ggtcccaggg aattgctttg aagcacatga 420 aggtgccact gggtctcaga aaatggaggt tatggttatg aagtgtgtat gacatatgtg 480 tataggaaga gcgtccgaaa gagcaggttt gttgccgacc ccagcattcg caaccctgag 540 gtccacagct ttctcctgat gggaggggaa tgggtggcaa agggtctgcg cgtgtggcaa 600 g 601 19 601 DNA Homo sapiens 19 atcccagggc tgctgccacc cccacctgtg gggagacacc agactggggg tggtgtggag 60 atactcttag agaagaggct gctgggccac gggctcggca tggcagggca gtggctaggt 120 aagtacttga gggacaggtg gggtctgctt gccaccgtcc cctctgcagg ctgggcctgg 180 gggctgctgc aggcggccag ggcagaaggg tgtggggaga gtgaacccac aggagcagcg 240 gctcgaggag ggggatgcag gctgcaggct caaaggggca ctggatccac cctgggtgcc 300 ygagagagca gggggcagcc cctggagggg tactcacccc cagagcttct gtggtcggct 360 gaggaccccc agcaggggtt gactgagggg atcagaggca agcagctgag gggagaggcc 420 aggttcttga tgctgatagg gtcggggtgc ctgggcgacc agaactcaag gagggaggca 480 tggggagggg ccgccgtgca gctggggtgg gtgcaccgca gagcctctgg gagtggtcag 540 aacccccgac acctgccact tctacagcag ctcatctgat tttaaggggc ttgctgccct 600 t 601 20 601 DNA Homo sapiens 20 agcacggtta ccactcttga ttggaactct gaccatgcat ctcctcttct gtttacttca 60 cgctttctct tcccatcaac tcccatttta attacaattt gtttaaaagc actgcatatt 120 acttcattaa acagaagatt agtttcactt accattagtg taaggtgact atagaaccaa 180 agcagactgg aaaccaaatg acataatgtc attctcttct ccattccagc tgcctgctgc 240 tgtgcgcctg agaacccctg tggagtggga ggggcagctg tctctgtaca ttagaaaggg 300 rggttaacta agtgacagga ggtgtttggg acatgtggac accagacttc tctcttgatg 360 caaggagggc agagccaggc agcctagtgg gggctggctt gggggctgct ggaaggactg 420 gctacaggtg gaagagaggt cagacctgaa gcttggggcc acctccagga aaggacaggt 480 gaaagtggag gcatgaggca ggggagaggc aggtgccagg cagagggtgg agaggaggca 540 ggaacatagc agctggggcg ggggcgggcc ctcaagtgtc atatgctact ttcctggggc 600 c 601 21 601 DNA Homo sapiens 21 gctgggcaca gtggctcata cctgtaatcc cagcactttg ggaggccgag gtgggcagat 60 cacttgaggt taggagtttg agaccagcct ggccaatatg gtgaaacctc atctccacta 120 aaaatataca cacacaaaaa ttagctgggt gtggtggtgt gcacctgtag ttccagctac 180 tcgggaggct gaggcaggag aatcgcttga acctgggagt cagagactgc agtgagccga 240 gatcatgtca ctgcactcca gcccgggtga cagagtgaga ctccatctaa aaaaaaaaaa 300 vaattccctc ctctgggaat ttagaccaca gacaggttgc atgtatgtgg ccgttggagg 360 cagcactcac agcaaagagt ggaaacgtca ccacagggcc tgccttctgg tgaaaatggt 420 gtcctgcagg gcgggcagct gtttgagggc aggtgtccca ggtgcggcct gcagcagcct 480 gagggtcaca gagcgcagtg ctgggagtgc agagacttcc cccacaggga gagttcccag 540 gaacctgctt ccggtgcact tctgggggtt tgagtttttt ccacggacga attactttga 600 g 601 22 601 DNA Homo sapiens 22 ttgaggttag gagtttgaga ccagcctggc caatatggtg aaacctcatc tccactaaaa 60 atatacacac acaaaaatta gctgggtgtg gtggtgtgca cctgtagttc cagctactcg 120 ggaggctgag gcaggagaat cgcttgaacc tgggagtcag agactgcagt gagccgagat 180 catgtcactg cactccagcc cgggtgacag agtgagactc catctaaaaa aaaaaaagaa 240 ttccctcctc tgggaattta gaccacagac aggttgcatg tatgtggccg ttggaggcag 300 yactcacagc aaagagtgga aacgtcacca cagggcctgc cttctggtga aaatggtgtc 360 ctgcagggcg ggcagctgtt tgagggcagg tgtcccaggt gcggcctgca gcagcctgag 420 ggtcacagag cgcagtgctg ggagtgcaga gacttccccc acagggagag ttcccaggaa 480 cctgcttccg gtgcacttct gggggtttga gttttttcca cggacgaatt actttgagaa 540 accactgtta ctcgtgtgta taggtgagcg tgcgtgtgca tgtgtgttct gtgtgtgagt 600 g 601 23 601 DNA Homo sapiens 23 gctgcttcct cctccccggc ctccgggtgg ccttgctgac ggctccttct ctgaggcagg 60 tctctgcctt ctcgcctggt gcctgcactc agtagccccc tcaccagagc tgctgggtga 120 aggaagcact aagaacccaa ggctcgggag gagagtgggg ccgggaagct gcagggaagc 180 gcagggccag gcctggtggg cccaggggct ggctcacggg agggcaggag ggagactgtg 240 gcggacagca cgtggggcca ggaggtgacc tccaagtgga ttgtgggtgg gttttttgtc 300 ytctttctgc attttccagg cattttgtaa tgtggataga atatttctgt tcttcaaaaa 360 tactttagtt aagaaaaata agatggaagc tgttgcactt gaaaatgagg aagccactgg 420 tgatgcaggg ggggcggcgg agaggacctc ttctgcaaat agcggcagga acacggcatg 480 gatgcagctc gcgctccccc aggccctccc ctgggctgtg tggaggggtc cggggggaat 540 gggccagcgc ccagtggtca cctggccatg tctccccaca gcccggaagc aggagatcat 600 t 601 24 601 DNA Homo sapiens variation (301)...(301) 'G' may be either present or absent (single nucleotide insertion/deletion polymorphism) 24 ataagatgga agctgttgca cttgaaaatg aggaagccac tggtgatgca gggggggcgg 60 cggagaggac ctcttctgca aatagcggca ggaacacggc atggatgcag ctcgcgctcc 120 cccaggccct cccctgggct gtgtggaggg gtccgggggg aatgggccag cgcccagtgg 180 tcacctggcc atgtctcccc acagcccgga agcaggagat cattaagacc acggagcagc 240 tcatcgaggc cgtcaacaac ggtgactttg aggcctacgc gtgagtccct ggggctgggg 300 gggggctgtg caggacaagg atgtgggacc cttggggggg cctgctcaga gtcaggggtc 360 cacggggccc ctcctcactt ggatttggcc cccaggaaaa tctgtgaccc agggctgacc 420 tcgtttgagc ctgaagcact gggcaacctg gttgaaggga tggacttcca cagattctac 480 ttcgagaacc gtgagtgagg aagcccgggt gggcatgagg gggcggtgcc cccaggagag 540 cctctcggcc cctcccaggg acagcatggt ggctgcctat ggaagccctg tcccctctgt 600 g 601 25 415 DNA Homo sapiens 25 cccgccagag gccataccca gcccccagaa tcccactctt ggaggggccc atgctgctcc 60 caggagagcc gagcctcccc aataagggga gttgagagag ggaaaggatt aggctggtgg 120 ggtggaagac gggcaccagg gcagtcatgg taacccgaga cccccgcccc gcctgctgtc 180 cacagtgctg gccaagaaca gcaagccrat ccacacgacc atcctgaacc cacacgtgca 240 cgtcattgga gaggatgccg cctgcatcgc ttacatccgg ctcacgcagt acattgacgg 300 gcagggccgg ccccgcacca gccagtctga ggagacccgc gtgtggcacc gccgcgacgg 360 caagtggcag aacgtgcact tccactgctc gggcgcgcct gtggccccgc tgcag 415 26 601 DNA Homo sapiens 26 gcctccccaa taaggggagt tgagagaggg aaaggattag gctggtgggg tggaagacgg 60 gcaccagggc agtcatggta acccgagacc cccgccccgc ctgctgtcca cagtgctggc 120 caagaacagc aagccgatcc acacgaccat cctgaaccca cacgtgcacg tcattggaga 180 ggatgccgcc tgcatcgctt acatccggct cacgcagtac attgacgggc agggccggcc 240 ccgcaccagc cagtctgagg agacccgcgt gtggcaccgc cgcgacggca agtggcagaa 300 ygtgcacttc cactgctcgg gcgcgcctgt ggccccgctg cagtgaaggt gagtgttctg 360 tgctaagtga cagctggggc agaggggtgg cggtggtgtg agtggctgca gcctggggag 420 gcgatgggga gcggtggggc ctgtggcaga gcccatgcct gggaagtccc tgagctttcc 480 tggtgaggcc acaggaatga tgtcaaatta gggaccacgg caggctgggt gtggcaggcc 540 tccccagagg actggggagc tggtgagggc ctgagcagtc cacactggcc agagctgggt 600 g 601 27 601 DNA Homo sapiens 27 tgtggcaaga ggactctgcc tgggctggcc cccctcctgt gtgaggtgtc tgtcccttct 60 ctgctggcca gcagcagatg cactggcagc tcccaaccct gtttccgccc ctcggccctc 120 ccccagcctg ttcggcttct ctgcagcccg caagggggag cagacttttg acaaaggact 180 gcgggcctcg ctcaagtccc tgagccccca gctgaagctg ggaggggagg ccaggctttg 240 tgtctgggca tattcgtctg ctgatggggt ttggggaagc ctggggcttg gggtttggtc 300 rggtggtgca gctagtggca gagcgggatc agaggtggtg gctgcccagc ttctgggctg 360 agacaagggt ctgtgcaggg gtttactgaa gtgggagtgc ctttggaatc tgggccggga 420 gcagaaggga gcaaaagcta cagtgggagc cagcctaggg cacatgggag gcgtgagggc 480 agtgctgccc gtgcagtgtc aggtgtgcca gtgccttggc gggctgcagt gcgtgtgagg 540 gcaccttcta ggtgggccag ggatgcagct atggagataa ggcgggctgg ggacagaaac 600 a 601 28 601 DNA Homo sapiens 28 gcaaactctt aggttggagt aaggagtaac cccctgccaa gtttctcctg tcctcaggct 60 ccacccacca cctatgctgc ctggccccat ggggcacacg ctcaggccca gcctgggaaa 120 gcaactgcac ctgcctgtgc tatgctggcc cttctcagcc tcaatgccct cctccctccc 180 cgacgcaccc tcgtggcccc cgctgggccc cctgatgcac cctcatgtct ccatggcaac 240 ctgctcagag tgtggccctg cccttggctc ccctccacac ctgtgtccca ggcagtgcca 300 yggcactttc ctaaacagaa ggatgggctt caaaacagtc ccagacacta aacacacctg 360 cattttgggt ccaagtaact tctgacaaga cgagtgcccc tacacaccct cagtcctatc 420 cactatgggc aaggagcctg aaggatcccc cagaactggc taaagccctc agtctcctcc 480 tccaccctga gcaccttcac gcggcagagt ggccctggat gtcagcttct tgctccccat 540 ggtctgcacc tggacaggtg ctctcaggtg tgtgggtggg caggtggcag gtcccaagag 600 c 601 29 601 DNA Homo sapiens 29 ccagcctggg aaagcaactg cacctgcctg tgctatgctg gcccttctca gcctcaatgc 60 cctcctccct ccccgacgca ccctcgtggc ccccgctggg ccccctgatg caccctcatg 120 tctccatggc aacctgctca gagtgtggcc ctgcccttgg ctcccctcca cacctgtgtc 180 ccaggcagtg ccacggcact ttcctaaaca gaaggatggg cttcaaaaca gtcccagaca 240 ctaaacacac ctgcattttg ggtccaagta acttctgaca agacgagtgc ccctacacac 300 yctcagtcct atccactatg ggcaaggagc ctgaaggatc ccccagaact ggctaaagcc 360 ctcagtctcc tcctccaccc tgagcacctt cacgcggcag agtggccctg gatgtcagct 420 tcttgctccc catggtctgc acctggacag gtgctctcag gtgtgtgggt gggcaggtgg 480 caggtcccaa gagccaggtg caaagaatct aggccagtgc ccacgagtgc tgcagtgtct 540 gtccccagca tggtatctag ggctccactt gcctatcagc tgtaatcgga ggaggctttc 600 c 601 30 403 DNA Homo sapiens 30 aagaatctag gccagtgccc acgagtgctg cagtgtctgt ccccagcatg gtatctaggg 60 ctccacttgc ctatcagctg taatcggagg aggctttcca ggccaggcct cccccaggaa 120 ggctgcaggc actgcggatc gtgcgccctc acatgcatta ttcctgaggc ccttctgcag 180 atgccatcag ggcagcaact ctgatgaggt attagggcac agcacacagg gctaagccac 240 cctgtactgg gccaagcgct acaggcaaaa aggacaccac cgacgggcat ttcattcatc 300 rcttttattt ttatatattt ttgagaggga gcctcactct gtcgcccagg ctggagtgca 360 gtggcgcgat cttggctcac tgcaacttct ccctcctggg ttc 403

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed