US20020173005A1 - Methods and materials relating to novel CD39-like polypeptides - Google Patents

Methods and materials relating to novel CD39-like polypeptides Download PDF

Info

Publication number
US20020173005A1
US20020173005A1 US10/092,063 US9206302A US2002173005A1 US 20020173005 A1 US20020173005 A1 US 20020173005A1 US 9206302 A US9206302 A US 9206302A US 2002173005 A1 US2002173005 A1 US 2002173005A1
Authority
US
United States
Prior art keywords
polypeptide
polynucleotide
seq
sequence
leu
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/092,063
Inventor
John Ford
Julio Mulero
George Yeung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hyseq Inc
Original Assignee
Hyseq Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/350,836 external-priority patent/US6387645B1/en
Priority claimed from PCT/US1999/016180 external-priority patent/WO2000004041A2/en
Application filed by Hyseq Inc filed Critical Hyseq Inc
Priority to US10/092,063 priority Critical patent/US20020173005A1/en
Publication of US20020173005A1 publication Critical patent/US20020173005A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • polynucleotides encoding the novel nucleic acids are changed via site-directed mutagenesis.
  • This method uses oligonucleotide sequences that encode the polynucleotide sequence of the desired amino acid variant, as well as a sufficient adjacent nucleotide on both sides of the changed amino acid to form a stable duplex on either side of the site being changed.
  • site-directed mutagenesis is well known to those of skill in the art and this technique is exemplified by publications such as, Edelman et al., DNA 2:183 (1983).
  • a versatile and efficient method for producing site-specific changes in a polynucleotide sequence was published by Zoller and Smith, Nucleic Acids Res. 10:6487-6500 (1982).
  • a further technique for generating amino acid variants is the cassette mutagenesis technique described in Wells et al., Gene 34:315 (1985); and other mutagenesis techniques well known in the art, such as, for example, the techniques in Sambrook, et al., supra, and Current Protocols in Molecular Biology, Ausubel, et al.
  • CD39-like gene that result in loss of normal function of the CD39-like gene product underlie CD39-related human disease states.
  • the invention comprehends gene therapy to restore CD39-like activity that would thus be indicated in treating those disease states.
  • Delivery of a functional CD39-like gene to appropriate cells is effected ex vivo, in situ, or in vivo by use of vectors, and more particuarly viral vectors (e.g., adenovirus, adeno-associated virus, or a retrovirus), or ex vivo by use of physical DNA transfer methods (e.g., liposomes or chemical treatments). See, for example, Anderson, Nature, supplement to vol. 392, no 6679, pp. 25-30 (1998).
  • Any animal which is known to produce antibodies can be immunized with a peptide or polypeptide of the invention.
  • Methods for immunization are well known in the art. Such methods include subcutaneous or intraperitoneal injection of the polypeptide.
  • One skilled in the art will recognize that the amount of the protein encoded by the ORF of the present invention used for immunization will vary based on the animal which is immunized, the antigenicity of the peptide and the site of injection.
  • any one of a number of methods well known in the art can be used to identify the hybridoma cell which produces an antibody with the desired characteristics. These include screening the hybridomas with an ELISA assay, western blot analysis, or radioimmunoassay (Lutz, et al., Exp. Cell Research. 175:109-124 (1988)).
  • SEQ ID NO:1 is the nucleotide sequence of an expressed sequence tag corresponding to a polynucleotide isolated from a cDNA library of human fetal liver-spleen.
  • SEQ ID NO:2 is an extended version of SEQ ID NO:1 obtained as described in Example 2, and the encoded polypeptide in SEQ ID NO: 3 is referred to herein as CD39-L4.
  • SEQ ID NO:2 encodes a polypeptide having the amino acid sequence of SEQ ID NO:3 (shown in FIG. 2).
  • the open reading frame corresponding to SEQ ID NO:3 starts at nucleotide 246, as numbered from the 5′ end of SEQ ID NO:2. This open reading frame encodes a polypeptide 428 amino acids in length.
  • the estimated molecular weight of the unglycosylated polypeptide is approximately 47.52 kDa.
  • polypeptides of the invention can be used as molecular weight markers, and as a food supplement.
  • a polypeptide consisting of SEQ ID NO:3, for example, has a molecular mass of approximately 47.52 kD in its unglycosylated form.
  • Protein food supplements are well known and the formulation of suitable food supplements including polypeptides of the invention is within the level of skill in the food preparation art.
  • the compounds may be administered topically, for example, as eye drops.
  • a targeted drug delivery system for example, in a liposome coated with a specific antibody, targeting, for example, arthritic or fibrotic tissue. The liposomes will be targeted to and taken up selectively by the afflicted tissue.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Many of the proteinase inhibiting compounds of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Such pharmaceutically acceptable base addition salts are those salts which retain the biological effectiveness and properties of the free acids and which are obtained by reaction with inorganic or organic bases such as sodium hydroxide, magnesium hydroxide, ammonia, trialkylamine, dialkylamine, monoalkylamine, dibasic amino acids, sodium acetate, potassium benzoate, triethanol amine and the like.
  • the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.01 ⁇ g to about 100 mg (preferably about 0.1 ⁇ g to about 10 mg, more preferably about 0.1 ⁇ g to about 1 mg) of protein of the present invention per kg body weight.
  • the therapeutic method includes administering the composition topically, systematically, or locally as an implant or device.
  • the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form.
  • the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage or tissue damage.
  • compositions may be biodegradable and chemically defined calcium sulfate, tricalciumphosphate, hydroxyapatite, polylactic acid, polyglycolic acid and polyanhydrides.
  • potential materials are biodegradable and biologically well-defined, such as bone or dermal collagen.
  • Further matrices are comprised of pure proteins or extracellular matrix components.
  • Other potential matrices are nonbiodegradable and chemically defined, such as sintered hydroxyapatite, bioglass, aluminates, or other ceramics.
  • a preferred family of sequestering agents is cellulosic materials such as alkylcelluloses (including hydroxyalkylcelluloses), including methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropyl-methylcellulose, and carboxymethylcellulose, the most preferred being cationic salts of carboxymethylcellulose (CMC).
  • CMC carboxymethylcellulose
  • Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer and poly(vinyl alcohol).

Abstract

The invention provides novel polynucleotides isolated from cDNA libraries of human fetal liver-spleen and macrophage as well as polypeptides encoded by these polynucleotides and mutants or variants thereof. The polypeptides correspond to a novel human CD39-like protein. Other aspects of the invention include vectors containing polynucleotides of the invention, and related host cells as well a processes for producing novel CD39-like polypeptides, and antibodies specific for such polypeptides.

Description

    1. RELATED APPLICATIONS
  • This patent application is a continuation-in-part of PCT patent application Serial No. PCT/US99/16180 [Attorney Docket No. 28110/35836] filed Jul. 16, 1999 which is a continuation-in-part of U.S. patent application Ser. No. 09/350836 [Attorney Docket No. 28110/35761] filed Jul. 9, 1999 which is a continuation-in-part of U.S. patent application Ser. No. 09/273,447 filed Mar. 19, 1999 which is a continuation-in-part of U.S. patent application Ser. No. 09/122,449 filed Jul. 24, 1998 and also a continuation-in-part of U.S. patent application Ser. No. 09/244,444 filed Feb. 4, 1999, which in turn is a continuation of U.S. patent application Ser. No. 09/118,205 filed Jul. 16, 1998, the disclosures of all of which are incorporated by reference herein in their entirety.[0001]
  • 2. FIELD OF THE INVENTION
  • This invention relates in general to novel polynucleotides isolated from cDNA libraries of human fetal liver-spleen and macrophages and to polypeptides encoded by these polynucleotides. In particular, the invention relates to a human CD39-like protein with homologies to ATP diphosphohydrolases and variants thereof. [0002]
  • 3. BACKGROUND
  • CD39 (cluster of differentiation 39) is a cell-surface molecule recognized by a “cluster” of monoclonal antibodies that can be used to identify the lineage or stage of differentiation of lymphocytes and thus to distinguish one class of lymphocytes from another. This CD39 molecule was originally defined as a B lymphocyte marker (Rowe, M., et al. Int. J. Cancer 29:373 (1982)). Subsequent studies have shown CD39 to be a marker for a distinct subset of activated lymphocytes within the allosensitized CD8-positive cytotoxic cells (Gouttefangeas C., et al., Eur. J.Immunol. 22:2681 (1992)). Outside of lymphoid tissue, CD39 can be found in quiescent vascular endothelial cells (Kansas, G. S., et al., J. Immunol. 146:2235 (1991)) and throughout rat brain in the neurons of the cerebral cortex, hippocampus, and cerebellum, as well as in glial cells (Wang, T-F. and Guidotti, G., Brain Res. 790:318 (1998)). [0003]
  • CD39 is a 510-amino acid protein with a predicted molecular mass of 57 kDa. However, because of heavy glycosylation at asparagine residues (six potential N-glycosylation sites) the molecule displays a mobility closer to 100 kDa (Maliszewski, C. R., et al., J. Immunol. 153:3574 (1994)). CD39 contains two hydrophobic regions, one near the amino terminus and the other near the carboxyl terminus which are believed to be transmembrane regions. [0004]
  • The role of CD39 in platelet aggregation and ATP/ADP hydrolysis is unclear. Although CD39 was originally reported to be an ectoADPase with a preference for ATP over ADP as a substrate, Wang, et al., [0005] J. Biol. Chem. 271:9898-9901(1996), Marcus, et al., J. Clin. Invest. 99:1351-1360 (1997) reported that CD39 was unique for its high preference for ADP over ATP as a substrate and in 1998, Gayle, et al., J. Clin Invest. 10:1851-1859 (1998), described CD39 as an ectoADPase with no preference for one substrate over the other.
  • Reports that several ATP Diphosphohydrolases (ATPDases) share amino acid sequence homology with CD39 have been substantiated by the showing that CD39 is itself an ATPDase (Wang, T- F., et al., J. Biol. Chem. 271:9898 (1996); Kaczmarek, E., et al., J. Biol. Chem. 271:33116 (1996)). Since CD39 is a plasma membrane-bound enzyme, CD39 has been termed an “ecto-ATPase,” but CD39 is more often referred to as an “ecto-apyrase” because of the reduced rate of hydrolysis of ADP when compared with ecto-ATPases. [0006]
  • This activity has shown to modulate platelet reactivity and aggregation in response to vascular injury. During vascular injury, activated platelets aggregate forming an occlusive thrombus. Excessive platelet accumulation at sites of vascular injury can contribute to vessel occlusion. Endothelial cells respond to the potentially occlusive effects of platelet aggregation by several mechanisms. One of these mechanisms results ecto-apyrase-mediated removal of ADP, which in turn eliminates platelet reactivity and recruitment. It is now known that the endothelial ecto-apyrase responsible for this ADP removal is CD39 (Marcus, A. J., et al., J. Clin. Invest. 99:1351 (1997)). [0007]
  • Recently, CD39 was engineered to produce a soluble form of the molecule. This soluble CD39 was shown to display the same nucleotidase activity as the membrane-bound molecule (Gayle, R. B., et al., J. Clin. Invest. 101:1851 (1998)). Intravenously administered soluble CD39 also remained active in mice for an extensive period of time, indicating that soluble CD39 could be useful as a inhibitor of platelet aggregation in the prophylaxis or treatment of platelet-mediated thrombotic conditions. [0008]
  • Platelet aggregation inhibitors (antithrombotic agents) decrease the formation or the action of chemical signals that promote platelet aggregation. Currently available antithrombotic agents include aspirin, ticlopidine, and dipyridamole. These agents have proven beneficial in the prevention and treatment of occlusive cardiovascular diseases, including myocardial infarction, cerebral ischemia, angina. Antithrombotic therapy has also been used in the maintenance of vascular grafts. [0009]
  • Myocardial infarction is the development of necrosis of the myocardium (the middle muscular layer of the heart wall) due to a critical imbalance between oxygen and myocardial demand. The most common cause of acute myocardium infarction is narrowing of the epicardial blood vessels due to atheromatous plaques. Plaque rupture with subsequent exposure of basement membrane results in platelet aggregation and thrombus formation, which can result in partial or complete occlusion of the vessel and subsequent myocardial ischemia. [0010]
  • In cerebral ischemia, inadequate blood flow results from an occlusion in a blood vessel or hemorrhaging. In the latter case, excessive bleeding in one area of the brain deprives another area of blood. If the damage occurs in a singular small area, “transient” or “focused” cerebral ischemia results. When a major artery is blocked (carotid artery) global or diffused ischemia results. The primary medical strategy for secondary prevention of stroke is antiplatelet therapy. Aspirin is currently employed for reducing the risk of recurrent transient ischemic attacks or stroke in men who have transient ischemia of the brain due to fibrin emboli. [0011]
  • Each year, thousands of patients suffer a decline in blood flow to one or more limbs. Without sufficient blood flow, and, unless blood flow can be restored in time, the limb must be amputated. In some cases, grafts from the patient's veins can be used to form new arteries. However, in cases where the quality of the veins is poor, polymeric vascular grafts are typically used. The polymeric grafts are inherently thrombogenic as the blood constituents passing through the grafts become activated and tend to form clots. Efforts to line the grafts with endothelial cells can reduce blood clotting, but better results are obtained when antithrombotic therapy is employed. [0012]
  • Angina pectoris is a characteristic chest pain caused by inadequate blood flow through the blood vessels of the myocardium. The imbalance between oxygen delivery and utilization may result from a spasm of the vascular smooth muscle or from obstruction of blood vessels caused by atherosclerotic lesions. Three classes of drugs have been shown to be effective in treating angina: nitrates, beta-blockers and calcium channel blockers. Currently, the antithrombotics dipyridamole and aspirin are employed to prophylactically treat angina pectoris. [0013]
  • Ecto-apyrases, such as CD39, offer a number of advantages over several of the standard antithrombotics. For example, aspirin treatment controls the prothrombotic action of thromboxane; however, aspirin also prevents formation of antithrombotic prostacyclin, which limits aspirin's efficacy. Another antithrombotic, endothelium-derived relaxing factor (nitric oxide; “EDRF/NO”), is inhibited in vitro and in vivo by hemoglobin after its rapid diffusion into erythrocytes. In contrast, CD39 is aspirin-insensitive and completely inhibits platelet reactivity even when eicosanoid and EDRF/NO production are blocked. [0014]
  • CD39's ATPDase activity also implicates CD39 in the modulation of neurotransmission. ATP is a major purinergic neurotransmitter that is often co-released into the synaptic cleft with several neurotransmitters. Responses to ATP are mediated by specific plasma membrane receptors, called P2 purinergic receptors (Dubyak, G. R. and El-Motassim,C. Am J. Physiol. 34:C577-C606 (1993)). The distribution of CD39 in the rat brain indicates that CD39 plays a role in terminating P2 purinergic neurotransmission (Wang, T. F. and Guidotti, G., Brain Res. 790:318 (1998)). Furthermore, a decrease in ecto-apyrase activity is believed to lead to an accumulation of the excitatory neurotransmitter, extracellular ATP, as well as a deficiency of the endogenous anticonvulsant extracellular adenosine. [0015]
  • The chomosomal localization of CD39 provides additional support for a role in modulation of neurotransmission. More specifically, CD39 has been mapped to chromosome 10q 23.1-24.1 (Maliszewski, C. R., et al., J. Immunol. 153:3574 (1994)), and this site overlaps with the susceptibility locus for human partial epilepsy with audiogenic symptoms (Ottman, R. et al., Nature Genet. 10:56 (1995)). This co-localization of the CD39 gene and the susceptibility locus has led to the hypothesis that decrease in ecto-apyrase activity in the brain is the primary cause of partial epilepsy (Wang T-F., et al., Mol. Brain Res. 47:295 (1997)). [0016]
  • A screen for human cDNAs that hybridize to cosmids from the human chromosome 9q34 region lead to the identification of a transcript with high homology to a chicken muscle ecto-ATPase (60% identity) and the ecto-apyrase CD39 (41% amino acid identity) (Chadwick, B. P., Mamm. Genome 8.668 (1997)). This gene, designated “CD39-like-1 gene” (CD39L1), has a higher degree of homology to CD39 than does chicken muscle ecto-ATPase. The biological activity of this protein has not been tested but on the basis of the high amino acid homology, CD39L1 is believed to be a new member of the ecto-ATPase family. Recently, a mouse gene with homology to NTPases was cloned and sequenced (Acc. No. AF006482) by Chadwick et al. (Mamm. Gen. 9:162-164 (1998).) [0017]
  • 4. SUMMARY OF THE INVENTION
  • The invention is based on polynucleotides isolated from cDNA libraries prepared from human fetal liver-spleen and macrophages. The compositions of the present invention include novel isolated polypeptides with apyrase and/or NDPase activity, in particular, novel human CD39-like polypeptides, and active variants thereof, isolated polynucleotides encoding such polypeptides, including recombinant DNA molecules, cloned genes or degenerate variants thereof, especially naturally occurring variants such as allelic variants, antisense polynucleotide molecules, and antibodies that specifically recognize one or more epitopes present on such polypeptides, as well as hybridomas producing such antibodies. [0018]
  • The compositions of the invention additionally include vectors, including expression vectors, containing the polynucleotides of the invention, cells genetically engineered to contain such polynucleotides and cells genetically engineered to express such polynucleotides. [0019]
  • The isolated polynucleotides of the invention include naturally occurring or wholly or partially synthetic DNA, e.g., cDNA and genomic DNA and RNA, e.g., mRNA. One polynucleotide according to the invention encodes a novel CD39-like protein having the amino acid sequence shown in FIG. 2 (SEQ ID NO. 3), which has been designated CD39-L4. Another polynucleotide according to the invention encodes a novel CD39-like protein having the amino acid sequence shown in SEQ ID NO: 27, which has been designated CD39-L2. In another embodiment, a polynucleotide according to the invention encodes a novel CD39-like protein having the full length or mature amino acid sequence set forth in SEQ ID NO. 25, which has been designated CD39-L66, and is an isoform of CD39-L4. The isolated polynucleotides of the invention include a polynucleotide comprising the nucleotide sequence of SEQ ID NO. 2, 24 or 26. The polynucleotides of the invention also include polynucleotides that encode polypeptides with a biological activity of the polypeptide of SEQ ID NO. 3 or 27 (including apyrase or NDPase activity) such as (a) the nucleotide sequence of SEQ ID NO. 2, 24, 26 or (b) a nucleotide sequence encoding the full length or mature amino acid sequence of SEQ ID NO. 3, 25, or 27; (c) a polynucleotide which is an allelic variant of any polynucleotide recited above; (d) a polynucleotide that hybridizes under stringent conditions to (a) or (b); (e) or a polynucleotide that encodes a polypeptide comprising at least one CD39-like domain, e.g. catalytic domain. [0020]
  • The polynucleotides of the invention additionally include the complement of any of the polynucleotides recited above. [0021]
  • The invention also provides a polynucleotide including a nucleotide sequence that is substantially equivalent to these polynucleotides. Polynucleotides according to the invention can have at least about 80%, more typically at least about 90%, and even more typically at least about 95%, sequence identity to a polynucleotide of SEQ ID NO. 2, 24 or 26 and specifically include a human polynucleotide which has at least 80% sequence identity to a polynucleotide of SEQ ID NO. 2, 24 or 26; or a polynucleotide which has at least 90% sequence identity to a polynucleotide of SEQ ID NO. 2, 24 or 26 . Similarly, polypeptides of the invention include polypeptides having apyrase or NDPase activity and at least about 80%, 90% or 95% sequence identity to SEQ ID NO. 3, 25 or 27. [0022]
  • A further aspect of the invention is the development of novel CD39-L4 variants which preferably have improved ADPase or NDPase activity compared to wild type CD39-L4 (SEQ ID NO: 5). This aspect of the invention includes polypeptides comprising at least one amino acid substitution selected from the group consisting of: D168-T, S170-Q and L175-F, wherein said substitution(s) result in increased ADPase activity of the polypeptide. One preferred embodiment is the polypeptide having the amino acid sequence set forth in SEQ ID NO: 7 (encoded by the nucleotide sequence of SEQ ID NO. 6), which is a variant CD39-L4 containing all three substitutions that has been designated ACRIII. A plasmid containing this DNA was deposited with the American Type Culture Collection (ATCC), 10801 University Avenue, Manassas, Va., on Jul. 13, 1999 under the terms of the Budapest Treaty (ATCC accession number ______). Alternatively, instead of making the specific D168-T, S170-Q and/or L175-F substitution(s), substitution of amino acids with similar properties is contemplated. Additional conservative substitutions at amino acid positions other than D168, S170 and/or L175 are further contemplated. For example, all of the corresponding amino acids from CD39 could be substituted for amino acids 167-181 of CD39-L66 or CD39-L4. [0023]
  • In addition, development of novel CD39-L2 variants which preferably have improved ADPase or NDPase activity compared to wild type CD39-L2 (SEQ ID NO: 27) is also contemplated. This aspect of the invention includes polypeptides comprising at least one amino acid substitution wherein said substitution(s) result in increased ADPase activity of the polypeptide. [0024]
  • Polynucleotides encoding these polypeptides, vectors and host cells comprising such polynucleotides, methods of using such host cells to produce polypeptides, and other therapeutic products comprising the polypeptides (including fusion proteins in which the CD39-like polypeptide is fused to a heterologous peptide or polypeptide, such as an immunoglobulin constant region, or derivatives in which the CD39-like polypeptide is modified by water soluble polymers to increase its half-life) are also comprehended by the invention, as are methods of treating a subject suffering from a disorder relating to thrombosis, coagulation or platelet aggregation by administering such therapeutic products. [0025]
  • The invention further comprises methods of inhibiting platelet aggregation in a mammalian subject by reducing the ratio of ADP:ATP in a mammalian subject to a less than normal ratio by administering the polypeptides of the invention or by administering polypeptides with ADPase activty and at least about 90% sequence identity to SEQ ID NO: 3, 25 or 27. Preferably the ratio of ADP:ATP is reduced without significantly affecting ATP levels. In one embodiment, the ADP:ATP ratio is reduced systemically in circulation. In another embodiment, the ADP:ATP ratio is reduced locally, for example, in heart, brain, kidney, lungs, limbs or other organs. [0026]
  • Methods of identifying compounds capable of reducing the ratio of ADP:ATP to a less than normal ratio are also contemplated. For example, compounds may be identified by steps including: determining apyrase activity of said compound on ATP; determining apyrase activity of said compounds on ADP; and selecting a compound that has greater activity with respect to ADP compared to ATP. Exemplary compounds to be screened include, but are not limited to, CD39-L4 and CD39-L2 variants. [0027]
  • Gene therapy techniques are also provided to modulate disease states associated with CD39-L4 or CD39-L2 expression and/or biological activity. Delivery of a functional CD39-L4 or CD39-L2 gene to appropriate cells is effected ex vivo, in situ, or in vivo by use of vectors, and more particularly viral vectors (e.g., adenovirus, adeno-associated virus, or a retrovirus), or ex vivo by use of physical DNA transfer methods (e.g., liposomes or chemical treatments). [0028]
  • The invention also relates to methods for producing polypeptides of the invention comprising growing a culture of cells of the invention in a suitable culture medium under conditions permitting expression of the desired polypeptide, and purifying the protein from the cells or the culture medium. Preferred embodiments include those in which the protein produced by such process is a mature form of the protein. [0029]
  • Protein compositions of the present invention, including therapeutic compositions, comprise polypeptides of the invention and optionally an acceptable carrier, such as a hydrophilic (e.g., pharmaceutically acceptable) carrier. [0030]
  • Polynucleotides according to the invention have numerous applications in a variety of techniques known to those skilled in the art of molecular biology. These techniques include use as hybridization probes, use as oligomers for PCR, use for chromosome and gene mapping, use in the recombinant production of protein, and use in generation of anti-sense DNA or RNA, their chemical analogs and the like. For example, because the expression of CD39-L4 and CD39-L2 mRNA is largely restricted to specific tissues (CD39-L4 in macrophages and CD39-L2 in adult heart and fetal brain), polynucleotides of the invention can be used as hybridization probes to detect the presence of specific mRNA in a sample using, e.g., in situ hybridization. [0031]
  • In other exemplary embodiments, the polynucleotides are used in diagnostics as expressed sequence tags for identifying expressed genes or, as well known in the art and exemplified by Vollrath, et al., Science 258:52-59 (1992), as expressed sequence tags for physical mapping of the human genome. [0032]
  • A polynucleotide according to the invention can be joined to any of a variety of other nucleotide sequences by well-established recombinant DNA techniques (see Sambrook, J., et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, NY). Useful nucleotide sequences for joining to polypeptides include an assortment of vectors, e.g., plasmids, cosmids, lambda phage derivatives, phagemids, and the like, that are well known in the art. Accordingly, the invention also provides a vector including a polynucleotide of the invention and a host cell containing the polynucleotide. In general, the vector contains an origin of replication functional in at least one organism, convenient restriction endonuclease sites, and a selectable marker for the host cell. Vectors according to the invention include expression vectors, replication vectors, probe generation vectors, and sequencing vectors. A host cell according to the invention can be a prokaryotic or eukaryotic cell and can be a unicellular organism or part of a multicellular organism. [0033]
  • The polypeptides according to the invention can be used in a variety of conventional procedures and methods that are currently applied to other proteins. For example, a polypeptide of the invention can be used to generate an antibody which specifically binds the polypeptide. The polypeptides of the invention having ATPDase activity are also useful for inhibiting platelet aggregation and can therefore be employed in the prophylaxis or treatment of pathological conditions caused by the inflammatory response. The polypeptides of the invention can also be used as molecular weight markers, and as a food supplement. [0034]
  • Another aspect of the invention is an antibody that specifically binds the polypeptide of the invention. Such antibodies can be either monoclonal or polyclonal antibodies, as well fragments thereof and humanized forms or fully human forms, such as those produced in transgenic animals. The invention further provides a hybridoma that produces an antibody according to the invention and anti-idiotype antibodies. [0035]
  • Antibodies of the invention are useful for detection and/or purification of the polypeptides of the invention. [0036]
  • Methods are also provided for preventing, treating or ameliorating a medical condition, including thrombotic diseases, which comprises administering to a mammalian subject, including but not limited to humans, a therapeutically effective amount of a composition comprising a polypeptide of the invention or a therapeutically effective amount of a composition comprising a binding partner of (e.g., antibody specifically reactive for) CD39-like polypeptides of the invention. The mechanics of the particular condition or pathology will dictate whether the polypeptides of the invention or binding partners (or inhibitors) of these would be beneficial to the individual in need of treatment. [0037]
  • The invention also provides a method of inhibiting platelet function comprising administering a CD39-L4 or CD39-L2 polypeptide of the invention to a medium comprising platelets. According to this method, polypeptides of the invention can be administered to produce an in vitro or in vivo inhibition of platelet function. A polypeptide of the invention can be administered in vivo as antithrombotic agent alone or as an adjunct to other therapies. [0038]
  • Also provided are methods of hydrolyzing nucleotidediphosphates comprising administering CD39-L4 or CD39-L2 polypeptides of the invention to a medium comprising nucleotidediphosphates. According to this method, polypeptides of the invention can be administered to produce an in vitro or in vivo hydrolysis of nucleotidediphosphates. A polypeptide of the invention can be administered in vivo alone or as an adjunct to other therapies. [0039]
  • The invention further provides methods for manufacturing medicaments useful in the above described methods relating to platelet aggregation and thrombosis. [0040]
  • The invention also provides methods for detecting or quantitating the presence of the polynucleotides or polypeptides of the invention in a tissue or fluid sample, and corresponding kits that comprise suitable polynucleotide probes or antibodies, together with an optional quantitative standard. Such methods and kits can be utilized as part of prognostic and diagnostic evaluation of patients and for the identification of subjects exhibiting a predisposition to platelet mediated conditions. [0041]
  • The invention also provides methods for the identification of compounds that modulate (i.e. increase or decrease) the expression or activity of the polynucleotides and/or polypeptides of the invention. Such methods can be utilized, for example, for the identification of compounds and other substances that interact with (e.g., bind to) the polypeptides of the invention, and assays for identifying compounds and other substances that enhance or inhibit the activity of the polypeptides of the invention, such assays comprising the step of measuring activity of such polypeptides in the presence and absence of the test compound.[0042]
  • 5. BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows polynucleotide sequences according to the invention. SEQ ID NO:1 was obtained from the b2HFLS20W cDNA library using standard PCR, sequencing by hybridization signature analysis, and single pass gel sequencing technology. A- adenosine; C-cytosine; G-guanosine; T-thymine. Ambiguous positions are designated as follows: R indicates A or G; M indicates A or C; W indicates A or T; Y indicates C or T; S indicates C or G; K indicates G or T; V indicates A or C or G; H indicates A or C or T; D indicates A or G or T; B indicates C or G or T; and N indicates any of the four bases. [0043]
  • SEQ ID NO:2 is an extended version of SEQ ID NO:1 which was obtained as described in Example 2. [0044]
  • FIG. 2 shows an amino acid sequence corresponding to the polynucleotide sequence of SEQ ID NO:2. This sequence is designated as SEQ ID NO:3. The open reading frame encoding SEQ ID NO:3 begins at nucleotide 246 (numbered from the 5′ end) of SEQ ID NO:2. A- Alanine; R-Arginine; N- Asparagine; D- Aspartic Acid; C- Cysteine; E- Glutamic Acid; Q-Glutamine; G- Glycine; H- Histidine; I- Isoleucine; L- Leucine; K- Lysine; M-Methionine; F- Phenylalanine; P- Proline; S- Serine; T- Threonine; W-Tryptophan; Y- Tyrosine; V- Valine; X - any of the twenty amino acids. [0045]
  • FIG. 3 shows the amino acid sequence alignment of SEQ ID NO:3 (identified as “246 prot”) and human CD39 (“CD39Human.seq”). The amino acid residues are designated as for FIG. 2. The alignment was generated using the Jotun Hein method with the PAM250 residue weight table. Gaps are indicated by dashes; residues that are identical between the two sequences (within 1 distance unit) are boxed. [0046]
  • FIG. 4 shows the amino acid sequence alignment of SEQ ID NO:3 (identified as “264 prot”) and murine NTPase (“mur ntpase”). The amino acid residues are designated as for FIG. 2. The alignment was generated as discussed for FIG. 3 Gaps are indicated by dashes; residues that are identical between the two sequences (within 1 distance unit) are boxed. [0047]
  • FIG. 5 shows the apyrase conserved regions (ACR) in CD39-L4 in bold. ACR I starts at Phe 53, ACR II starts at Pro 124 and ACR III starts at Met 167. The boxed sections highlight the amino acid substitutions that were made in the wild type CD39-L4 amino acid sequence to form mutants designated ACRI, ACRII and ACRIII. [0048]
  • FIG. 6 (SEQ ID NOS: 6 and 7) shows the nucleotide and corresponding amino acid sequences of a preferred ACRIII mutant containing the following substitutions in the wild type CD39-L4 amino acid sequence: D168-T, S170-Q and L175-F. [0049]
  • FIG. 7 shows the ADPase activity of CD39-L4 variants ACRI, ACRII and ACRIII in comparison to wild type CD39-L4: (1) CD39-L4 ACR I mutant; (2) CD39-L4 ACR II mutant; (3) CD39-L4 ACR III mutant; (4) CD39-L4 wild type; (5) sCD39; and (6) pSecTag2 vector (Invitrogen). [0050]
  • FIG. 8 shows the amino acid sequence alignment of SEQ ID NO. 3, SEQ ID NO. 25 (previously identified as SEQ ID NO. 5 in FIG. 5 of U.S. Ser. No. 09/122,449) and human CD39 (“CD39Human.seq”). The alignment was generated using the Jotun Hein method with the PAM250 residue weight table. Gaps are indicated by dashes; residues that are identical between the two sequences (within 1 distance unit) are boxed. [0051]
  • FIG. 9 shows the amino acid sequence alignment of SEQ ID NO. 3, SEQ ID NO. 25 (previously identified as SEQ ID NO. 5 in FIG. 6 of U.S. Ser. No. 09/122,449) and the murine NTPase (“mur ntpase”). The alignment was generated as discussed for FIG. 8. Gaps are indicated by dashes; residues that are identical between the two sequences (within 1 distance unit) are boxed.[0052]
  • 6. DETAILED DESCRIPTION
  • 6.1 Definitions [0053]
  • The term “nucleotide sequence” refers to a heteropolymer of nucleotides or the sequence of these nucleotides. The terms “nucleic acid” and “polynucleotide” are also used interchangeably herein to refer to a heteropolymer of nucleotides. Generally, nucleic acid segments provided by this invention may be assembled from fragments of the genome and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic nucleic acid which is capable of being expressed in a recombinant transcriptional unit comprising regulatory elements derived from a microbial or viral operon. [0054]
  • An “oligonucleotide fragment” or a “polynucleotide fragment”, “portion,” or “segment” is a stretch of polypeptide nucleotide residues which is long enough to use in polymerase chain reaction (PCR) or various hybridization procedures to identify or amplify identical or related parts of mRNA or DNA molecules. [0055]
  • “Oligonucleotides” or “nucleic acid probes” are prepared based on the cDNA sequence provided in the present invention. Oligonucleotides comprise portions of the DNA sequence having at least about 15 nucleotides and usually at least about 20 nucleotides. Nucleic acid probes comprise portions of the sequence having fewer nucleotides than about 6 kb, usually fewer than about 1 kb. After appropriate testing to eliminate false positives, these probes may be used to determine whether mRNAs are present in a cell or tissue or to isolate similar nucleic acid sequences from chromosomal DNA as described by Walsh, P. S., et al (1992 PCR Methods Appl 1:241-250). [0056]
  • The term “probes” includes naturally occurring or recombinant single- or double-stranded nucleic acids or chemically synthesized nucleic acids. They may be labeled by nick translation, Klenow fill-in reaction, PCR or other methods well known in the art. Probes of the present invention, their preparation and/or labeling are elaborated in Sambrook, J., et al (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, NY; or Ausubel, F. M., et al (1989) Current Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., both incorporated herein by reference. [0057]
  • The term “stringent” is used to refer to conditions that are commonly understood in the art as stringent. An exemplary set of conditions include a temperature of 60-70° C., (preferably about 65° C.) and a salt concentration of 0.70 M to 0.80 M (preferably about 0.75M). Further exemplary conditions include, hybridizing conditions that (1) employ low ionic strength and high temperature for washing, for example, 0.015 M NaCl/0.0015 M sodium citrate/0.1% SDS at 50° C.; (2) employ during hybridization a denaturing agent such as formamide, for example, 50% (vol/vol) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM NaCl, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M Sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 g/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC and 0.1% SDS. [0058]
  • The term “recombinant,” as used herein, means that a polypeptide or protein is derived from recombinant (e.g., microbial or mammalian) expression systems. “Microbial” refers to recombinant polypeptides or proteins made in bacterial or fungal (e.g., yeast) expression systems. As a product, “recombinant microbial” defines a polypeptide or protein essentially free of native endogenous substances and unaccompanied by associated native glycosylation. Polypeptides or proteins expressed in most bacterial cultures, e.g., [0059] E. coli, will be free of glycosylation modifications; polypeptides or proteins expressed in yeast will have a glycosylation pattern different from that expressed in mammalian cells.
  • The term “recombinant expression vehicle or vector” refers to a plasmid or phage or virus or vector, for expressing a polypeptide from a DNA (RNA) sequence. The expression vehicle can comprise a transcriptional unit comprising an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription initiation and termination sequences. Structural units intended for use in yeast or eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell. Alternatively, where recombinant protein is expressed without a leader or transport sequence, it may include an N-terminal methionine residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final product. [0060]
  • “Recombinant expression system” means host cells which have stably integrated a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit extrachromosomally. The cells can be prokaryotic or eukaryotic. Recombinant expression systems as defined herein will express heterologous polypeptides or proteins upon induction of the regulatory elements linked to the DNA segment or synthetic gene to be expressed. [0061]
  • The term “open reading frame,” ORF, means a series of triplets coding for amino acids without any termination codons and is a sequence translatable into protein. [0062]
  • The term “expression modulating fragment,” EMF, means a series of nucleotide molecules which modulates the expression of an operably linked ORF or EMF. As used herein, a sequence is said to “modulate the expression of an operably linked sequence” when the expression of the sequence is altered by the presence of the EMF. EMFs include, but are not limited to, promoters, and promoter modulating sequences (inducible elements). One class of EMFs are fragments which induce the expression of an operably linked ORF in response to a specific regulatory factor or physiological event. [0063]
  • As used herein, an “uptake modulating fragment,” UMF, means a series of nucleotide molecules which mediate the uptake of a linked DNA fragment into a cell. UMFs can be readily identified using known UMFs as a target sequence or target motif with the computer-based systems known in the art. [0064]
  • The presence and activity of a UMF can be confirmed by attaching the suspected UMF to a marker sequence. The resulting nucleic acid molecule is then incubated with an appropriate host under appropriate conditions and the uptake of the marker sequence is determined. As described above, a UMF will increase the frequency of uptake of a linked marker sequence. [0065]
  • “Active” refers to those forms of the polypeptide which retain the biologic and/or immunologic activities of any naturally occurring polypeptide. [0066]
  • “Naturally occurring polypeptide” refers to polypeptides produced by cells that have not been genetically engineered and specifically contemplates various polypeptides arising from post-translational modifications of the polypeptide including, but not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. [0067]
  • “Derivative” refers to polypeptides chemically modified by such techniques as ubiquitination, labeling (e.g., with radionuclides or various enzymes), pegylation (derivatization with polyethylene glycol) and insertion or substitution by chemical synthesis of amino acids such as ornithine, which do not normally occur in human proteins. [0068]
  • “Recombinant variant” refers to any polypeptide differing from naturally occurring polypeptides by amino acid insertions, deletions, and substitutions, created using recombinant DNA techniques. Guidance in determining which amino acid residues may be replaced, added or deleted without abolishing activities of interest, such as cellular trafficking, may be found by comparing the sequence of the particular polypeptide with that of homologous peptides and minimizing the number of amino acid sequence changes made in regions of high homology. [0069]
  • Preferably, amino acid “substitutions” are the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine, i.e., conservative amino acid replacements. “Insertions” or “deletions” are typically in the range of about 1 to 5 amino acids. The variation allowed may be experimentally determined by systematically making insertions, deletions, or substitutions of amino acids in a polypeptide molecule using recombinant DNA techniques and assaying the resulting recombinant variants for activity. [0070]
  • As used herein, “substantially equivalent” can refer both to nucleotide and amino acid sequences, for example a mutant sequence, that varies from a reference sequence by one or more substitutions, deletions, or additions, the net effect of which does not result in an adverse functional dissimilarity between the reference and subject sequences. Typically, such a mutant sequence varies from one of those listed herein by no more than about 20% (i.e., the number of substitutions, additions, and/or deletions in a mutant sequence, as compared to the corresponding listed sequence, divided by the total number of residues in the mutant sequence is about 0.2 or less). Such a mutant sequence is said to have 80% sequence identity to the listed sequence. In one embodiment, a mutant sequence of the invention varies from a listed sequence by no more than 10% (90% sequence identity), in a variation of this embodiment, by no more than 5% (95% sequence identity), and in a further variation of this embodiment, by no more than 2% (98% sequence identity). Mutant amino acid sequences according to the invention generally have at least 95% sequence identity with a listed amino acid sequence, whereas mutant nucleotide sequence of the invention can have lower percent sequence identities. For the purposes of the present invention, sequences having substantially equivalent biological activity and substantially equivalent expression characteristics are considered substantially equivalent. For the purposes of determining equivalence, truncation of the mature sequence should be disregarded. [0071]
  • Where desired, an expression vector may be designed to contain a “signal or leader sequence” which will direct the polypeptide through the membrane of a cell. Such a sequence may be naturally present on the polypeptides of the present invention or provided from heterologous protein sources by recombinant DNA techniques. [0072]
  • A polypeptide “fragment,” “portion,” or “segment” is a stretch of amino acid residues of at least about 5 amino acids, often at least about 7 amino acids, typically at least about 9 to 13 amino acids, and, in various embodiments, at least about 17 or more amino acids. To be active, any polypeptide must have sufficient length to display biologic and/or immunologic activity. [0073]
  • Alternatively, recombinant variants encoding these same or similar polypeptides may be synthesized or selected by making use of the “redundancy” in the genetic code. Various codon substitutions, such as the silent changes which produce various restriction sites, may be introduced to optimize cloning into a plasmid or viral vector or expression in a particular prokaryotic or eukaryotic system. Mutations in the polypeptide sequence may be reflected in the polypeptide or domains of other peptides added to the polypeptide to modify the properties of any part of the polypeptide, to change characteristics such as ligand-binding affinities, interchain affinities, or degradation/turnover rate. [0074]
  • “Activated” cells as used in this application are those which are engaged in extracellular or intracellular membrane trafficking, including the export of neurosecretory or enzymatic molecules as part of a normal or disease process. [0075]
  • The term “purified” as used herein denotes that the indicated nucleic acid or polypeptide is present in the substantial absence of other biological macromolecules, e.g., polynucleotides, proteins, and the like. In one embodiment, the polynucleotide or polypeptide is purified such that it constitutes at least 95% by weight, more preferably at least 99.8% by weight, of the indicated biological macromolecules present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 1000 daltons, can be present). [0076]
  • The term “isolated” as used herein refers to a nucleic acid or polypeptide separated from at least one other component (e.g., nucleic acid or polypeptide) present with the nucleic acid or polypeptide in its natural source. In one embodiment, the nucleic acid or polypeptide is found in the presence of (if anything) only a solvent, buffer, ion, or other component normally present in a solution of the same. The terms “isolated” and “purified” do not encompass nucleic acids or polypeptides present in their natural source. [0077]
  • The term “infection” refers to the introduction of nucleic acids into a suitable host cell by use of a virus or viral vector. [0078]
  • The term “transformation” means introducing DNA into a suitable host cell so that the DNA is replicable, either as an extrachromosomal element, or by chromosomal integration. [0079]
  • The term “transfection” refers to the taking up of an expression vector by a suitable host cell, whether or not any coding sequences are in fact expressed. [0080]
  • The term “intermediate fragment” means a nucleic acid between 5 and 1000 bases in length, and preferably between 10 and 40 bp in length. [0081]
  • Each of the above terms is meant to encompasses all that is described for each, unless the context dictates otherwise. [0082]
  • 6.2 Hybridization Conditions [0083]
  • Suitable hybridization conditions may be routinely determined by optimization procedures or pilot studies. Such procedures and studies are routinely conducted by those skilled in the art to establish protocols for use in a laboratory. See e.g., Ausubel, et al., Current Protocols in Molecular Biology, Vol. 1-2, John Wiley & Sons (1989); Sambrook, et al., Molecular Cloning A Laboratory Manual, 2nd Ed., Vols. 1-3, Cold Springs Harbor Press (1989); and Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Cold Spring Harbor, N.Y. (1982), all of which are incorporated by reference herein. For example, conditions such as temperature, concentration of components, hybridization and washing times, buffer components, and their pH and ionic strength may be varied. [0084]
  • 6.3 Nucleic Acids of the Invention [0085]
  • The sequences falling within the scope of the present invention are not limited to the specific sequences herein described, but also include allelic variations thereof. Allelic variations can be routinely determined by comparing the sequence provided in SEQ ID NOs: 1, 2, 24 or 26, a representative fragment thereof, or a nucleotide sequence at least 99.9% identical to SEQ ID NO: 1, 2, 24 or 26 with a sequence from another isolate of the same species. Furthermore, to accommodate codon variability, the invention includes nucleic acid molecules coding for the same amino acid sequences as do the specific ORFs disclosed herein. In other words, in the coding region of an ORF, substitution of one codon for another which encodes the same amino acid is expressly contemplated. [0086]
  • Any specific sequence disclosed herein can be readily screened for errors by resequencing a particular fragment, such as an ORF, in both directions (i.e., sequence both strands). [0087]
  • The present invention further provides recombinant constructs comprising a nucleic acid having the sequence of any one of SEQ ID NO: 1, 2, 24 or 26, the mature protein coding sequence or a fragment thereof. The recombinant constructs of the present invention comprise a vector, such as a plasmid or viral vector, into which a nucleic acid having the sequence of any one of SEQ ID NO: 1, 2, 24 or 26 or a fragment thereof is inserted, in a forward or reverse orientation. In the case of a vector comprising one of the ORFs of the present invention, the vector may further comprise regulatory sequences, including for example, a promoter, operably linked to the ORF. For vectors comprising the EMFs and UMFs of the present invention, the vector may further comprise a marker sequence or heterologous ORF operably linked to the EMF or UMF. Large numbers of suitable vectors and promoters are known to those of skill in the art and are commercially available for generating the recombinant constructs of the present invention. The following vectors are provided by way of example. Bacterial: pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene); pTrc99A, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia). Eukaryotic: pWLneo, pSV2cat, pOG44, PXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). [0088]
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lacI, lacZ, T3, T7, gpt, lambda PR, and trc. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art. [0089]
  • Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of [0090] E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transformation include [0091] E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM 1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. [0092]
  • Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced or derepressed by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period. Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. [0093]
  • Included within the scope of the nucleic acid sequences of the invention are nucleic acid sequences that hybridize under stringent conditions to a fragment of the DNA sequences in FIG. 1, which fragment is greater than about 10 bp, preferably 20-50 bp, and even greater than 100 bp, including 200 bp or greater, 300 bp or greater, 400 bp or greater, and 500 bp or greater. [0094]
  • In accordance with the invention, polynucleotide sequences which encode the novel nucleic acids, or functional equivalents thereof, may be used to generate recombinant DNA molecules that direct the expression of that nucleic acid, or a functional equivalent thereof, in appropriate host cells. [0095]
  • The nucleic acid sequences of the invention are further directed to sequences which encode variants of the described nucleic acids. These amino acid sequence variants may be prepared by methods known in the art by introducing appropriate nucleotide changes into a native or variant polynucleotide. There are two variables in the construction of amino acid sequence variants: the location of the mutation and the nature of the mutation. The amino acid sequence variants of the nucleic acids are preferably constructed by mutating the polynucleotide to give an amino acid sequence that does not occur in nature. These amino acid alterations can be made at sites that differ in the nucleic acids from different species (variable positions) or in highly conserved regions (constant regions). Sites at such locations will typically be modified in series, e.g., by substituting first with conservative choices (e.g., hydrophobic amino acid to a different hydrophobic amino acid) and then with more distant choices (e.g., hydrophobic amino acid to a charged amino acid), and then deletions or insertions may be made at the target site. [0096]
  • Amino acid sequence deletions generally range from about 1 to 30 residues, preferably about 1 to 10 residues, and are typically contiguous. Amino acid insertions include amino- and/or carboxyl-terminal fusions ranging in length from one to one hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Intrasequence insertions may range generally from about 1 to 10 amino residues, preferably from 1 to 5 residues. Examples of terminal insertions include the heterologous signal sequences necessary for secretion or for intracellular targeting in different host cells. [0097]
  • In a preferred method, polynucleotides encoding the novel nucleic acids are changed via site-directed mutagenesis. This method uses oligonucleotide sequences that encode the polynucleotide sequence of the desired amino acid variant, as well as a sufficient adjacent nucleotide on both sides of the changed amino acid to form a stable duplex on either side of the site being changed. In general, the techniques of site-directed mutagenesis are well known to those of skill in the art and this technique is exemplified by publications such as, Edelman et al., DNA 2:183 (1983). A versatile and efficient method for producing site-specific changes in a polynucleotide sequence was published by Zoller and Smith, Nucleic Acids Res. 10:6487-6500 (1982). [0098]
  • PCR may also be used to create amino acid sequence variants of the novel nucleic acids. When small amounts of template DNA are used as starting material, primer(s) that differs slightly in sequence from the corresponding region in the template DNA can generate the desired amino acid variant. PCR amplification results in a population of product DNA fragments that differ from the polynucleotide template encoding the polypeptide at the position specified by the primer. The product DNA fragments replace the corresponding region in the plasmid and this gives the desired amino acid variant. [0099]
  • A further technique for generating amino acid variants is the cassette mutagenesis technique described in Wells et al., Gene 34:315 (1985); and other mutagenesis techniques well known in the art, such as, for example, the techniques in Sambrook, et al., supra, and Current Protocols in Molecular Biology, Ausubel, et al. [0100]
  • Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be used in the practice of the invention for the cloning and expression of these novel nucleic acids. Such DNA sequences include those which are capable of hybridizing to the appropriate novel nucleic acid sequence under stringent conditions. [0101]
  • Furthermore, knowledge of the DNA sequence provided by the present invention allows for the modification of cells to permit, or increase, expression of endogenous CD39-like polypeptides. Cells can be modified (e.g., by homologous recombination) to provide increased CD39-like expression by replacing, in whole or in part, the naturally occurring CD39-like promoter with all or part of a heterologous promoter so that the cells express CD39-like polypeptides at a higher level. The heterologous promoter is inserted in such a manner that it is operatively linked to CD39-like encoding sequences. See, for example, PCT International Publication No. WO94/12650, PCT International Publication No. WO92/20808, and PCT International Publication No. WO91/09955. It is also contemplated that, in addition to heterologous promoter DNA, amplifiable marker DNA (e.g., ada, dhfr, and the multifunctional CAD gene which encodes carbamyl phosphate synthase, aspartate transcarbamylase, and dihydroorotase) and/or intron DNA may be inserted along with the heterologous promoter DNA. If linked to the CD39-like coding sequence, amplification of the marker DNA by standard selection methods results in co-amplification of the CD39-like coding sequences in the cells. [0102]
  • The polynucleotides of the present invention also make possible the development, through, e.g., homologous recombination or knock out strategies, of animals that fail to express functional CD39-like polypeptides or that express a variant of a CD39-like polypeptide. Such animals are useful as models for studying the in vivo activities of CD39-like polypeptides as well as for studying modulators of CD39-like polypeptides. [0103]
  • 6.4 Identification of Polymorphisms [0104]
  • Polymorphisms can be identified in a variety of ways known in the art which all generally involve obtaining a sample from a patient, analyzing DNA from the sample, optionally involving isolation or amplification of the DNA, and identifying the presence of the polymorphism in the DNA. For example, PCR may be used to amplify an appropriate fragment of genomic DNA which may then be sequenced. Alternatively, the DNA may be subjected to allele-specific oligonucleotide hybridization (in which appropriate oligonucleotides are hybridized to the DNA under conditions permitting detection of a single base mismatch) or to a single nucleotide extension assay (in which an oligonucleotide that hybridizes immediately adjacent to the position of the polymorphism is extended with one or more labelled nucleotides). In addition, traditional restriction fragment length polymorphism analysis (using restriction enzymes that provide differential digestion of the genomic DNA depending on the presence or absence of the polymorphism) may be performed. [0105]
  • Alternatively, a polymorphism resulting in a change in the amino acid sequence could also be detected by detecting a corresponding change in amino acid sequence of the protein, e.g., by an antibody specific to the variant sequence. [0106]
  • 6.5 Hosts [0107]
  • The present invention further provides host cells containing SEQ ID NO: 1, 2, 24 or 26 of the present invention, wherein the nucleic acid has been introduced into the host cell using known transformation, transfection or infection methods. The host cell can be a higher eukaryotic host cell, such as a mammalian cell, a lower eukaryotic host cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the recombinant construct into the host cell can be effected by calcium phosphate transfection, DEAE, dextran mediated transfection, or electroporation (Davis, L., et al., Basic Methods in Molecular Biology (1986)). [0108]
  • The host cells containing one of SEQ ID NO: 1, 2, 24 or 26 of the present invention, can be used in conventional manners to produce the gene product encoded by the isolated fragment (in the case of an ORF) or can be used to produce a heterologous protein under the control of the EMF. [0109]
  • Any host/vector system can be used to express one or more of the ORFs of the present invention. These include, but are not limited to, eukaryotic hosts such as HeLa cells, Cv-1 cell, COS cells, and Sf9 cells, as well as prokaryotic host such as [0110] E. coli and B. subtilis. The most preferred cells are those which do not normally express the particular polypeptide or protein or which expresses the polypeptide or protein at low natural level.
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, insect cells or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., in Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y. (1989), the disclosure of which is hereby incorporated by reference. [0111]
  • Various mammalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking nontranscribed sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements. [0112]
  • Recombinant polypeptides and proteins produced in bacterial culture are usually isolated by initial extraction from cell pellets, followed by one or more salting-out, aqueous ion exchange or size exclusion chromatography steps. Protein refolding steps can be used, as necessary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. [0113]
  • 6.6 Peptides [0114]
  • The present invention further provides isolated polypeptides encoded by the nucleic acid fragments of the present invention or by degenerate variants of the nucleic acid fragments of the present invention. Fragments may be fused to carrier molecules such as immunoglobulins for many purposes, including increasing the valency of protein binding sites. For example, fragments of the protein may be fused through “linker” sequences to the Fc portion of an immunoglobulin. For a bivalent form of the protein, such a fusion could be to the Fc portion of an IgG molecule. Other immunoglobulin isotypes may also be used to generate such fusions. For example, a protein-IgM fusion would generate a decavalent form of the protein of the invention. Analogs of the polypeptides of the invention can be fused to another moiety or moieties, e,g., targeting moiety or another therapeutic agent. Such analogs may exhibit improved properties such as activity and/or stability. By “degenerate variant” is intended nucleotide fragments which differ from a nucleic acid fragment of the present invention (e.g., an ORF) by nucleotide sequence but, due to the degeneracy of the genetic code, encode an identical polypeptide sequence. Preferred nucleic acid fragments of the present invention are the ORFs which encode proteins. [0115]
  • The invention also provides both full length and mature forms (for example, without a signal sequence or precursor sequence) of CD39-like polypeptides. The full length form of such proteins is identified in the sequence listing by translation of the nucleotide sequence of each disclosed clone. The mature form of such protein may be obtained by expression of the full-length polynucleotide in a suitable mammalian cell or other host cell. The sequence of the mature form of the protein is also determinable from the amino acid sequence of the full length form. [0116]
  • A variety of methodologies known in the art can be utilized to obtain any one of the isolated polypeptides or proteins of the present invention. At the simplest level, the amino acid sequence can be synthesized using commercially available peptide synthesizers. This is particularly useful in producing small peptides and fragments of larger polypeptides. Fragments are useful, for example, in generating antibodies against the native polypeptide. In an alternative method, the polypeptide or protein is purified from bacterial cells which naturally produce the polypeptide or protein. One skilled in the art can readily follow known methods for isolating polypeptides and proteins in order to obtain one of the isolated polypeptides or proteins of the present invention. These include, but are not limited to, immunochromatography, HPLC, size-exclusion chromatography, ion-exchange chromatography, and immuno-affinity chromatography. See, e.g., Scopes, Protein Purification: Principles and Practice, Springer-Verlag (1994); Sambrook, et al., in Molecular Cloning: A Laboratory Manual; Ausubel, et al., Current Protocols in Molecular Biology. [0117]
  • The polypeptides and proteins of the present invention can alternatively be purified from cells which have been altered to express the desired polypeptide or protein. As used herein, a cell is said to be altered to express a desired polypeptide or protein when the cell, through genetic manipulation, is made to produce a polypeptide or protein which it normally does not produce or which the cell normally produces at a lower level. One skilled in the art can readily adapt procedures for introducing and expressing either recombinant or synthetic sequences into eukaryotic or prokaryotic cells in order to generate a cell which produces one of the polypeptides or proteins of the present invention. [0118]
  • The purified polypeptides are used in in vitro binding assays which are well known in the art to identify molecules which bind to the polypeptides. These molecules include but are not limited to, for example, small molecules, molecules from combinatorial libraries, antibodies or other proteins. The molecules identified in the binding assay are then tested for antagonist or agonist activity in in vivo tissue culture or animal models that are well known in the art. In brief, the molecules are titrated into a plurality of cell cultures or animals and then tested for either cell/animal death or prolonged survival of the animal/cells. [0119]
  • In addition, the binding molecules may be complexed with toxins, e.g., ricin or cholera, or with other compounds that are toxic to cells. The toxin-binding molecule complex is then targeted to the tumor or other cell by the specificity of the binding molecule for SEQ ID NOs:3-4. [0120]
  • 6.7 Gene Therapy [0121]
  • Mutations in the CD39-like gene that result in loss of normal function of the CD39-like gene product underlie CD39-related human disease states. The invention comprehends gene therapy to restore CD39-like activity that would thus be indicated in treating those disease states. Delivery of a functional CD39-like gene to appropriate cells is effected ex vivo, in situ, or in vivo by use of vectors, and more particuarly viral vectors (e.g., adenovirus, adeno-associated virus, or a retrovirus), or ex vivo by use of physical DNA transfer methods (e.g., liposomes or chemical treatments). See, for example, Anderson, Nature, supplement to vol. 392, no 6679, pp. 25-30 (1998). For additional reviews of gene therapy technology, see Friedmann, Science, 244: 1275-1281 (1989); Verma, Scientific American: 68-84 (1990); and Miller, Nature, 357: 455-460 (1992). Alternatively, it is contemplated that in other human disease states, preventing the expression of or inhibiting the activity of CD39-like polypeptides will be useful in treating the disease states. It is contemplated that antisense therapy or gene therapy could be applied to negatively regulate the expression of CD39-like polypeptides. [0122]
  • 6.8 Deposit of clone [0123]
  • A plasmid containing DNA encoding the ACR III mutant was deposited with the American Type Culture Collection (ATCC), 10801 University Avenue, Manassas, Va., on Jul. 13, 1999 under the terms of the Budapest Treaty (ATCC accession no. ______). [0124]
  • 6.9 Antibodies [0125]
  • In general, techniques for preparing polyclonal and monoclonal antibodies as well as hybridomas capable of producing the desired antibody are well known in the art (Campbell, A. M., Monoclonal Antibodies Technology: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1984); St. Groth, et al., J. Immunol. 35:1-21 (1990); Kohler and Milstein, Nature 256:495-497 (1975)), the trioma technique, the human B-cell hybridoma technique (Kozbor, et al., Immunology Today 4:72 (1983); Cole, et al., in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. (1985), pp. 77-96). In addition, techniques for preparing chimeric,and humanized antibodies (including polypeptides containing CDR and/or antigen-binding sequences of antibodies) are well known in the art. [0126]
  • Any animal (mouse, rabbit, etc.) which is known to produce antibodies can be immunized with a peptide or polypeptide of the invention. Methods for immunization are well known in the art. Such methods include subcutaneous or intraperitoneal injection of the polypeptide. One skilled in the art will recognize that the amount of the protein encoded by the ORF of the present invention used for immunization will vary based on the animal which is immunized, the antigenicity of the peptide and the site of injection. [0127]
  • The protein which is used as an immunogen may be modified or administered in an adjuvant in order to increase the protein's antigenicity. Methods of increasing the antigenicity of a protein are well known in the art and include, but are not limited to, coupling the antigen with a heterologous protein (such as globulin or -galactosidase) or through the inclusion of an adjuvant during immunization. [0128]
  • For monoclonal antibodies, spleen cells from the immunized animals are removed, fused with myeloma cells, such as SP2/0-Ag14 myeloma cells, and allowed to become monoclonal antibody producing hybridoma cells. [0129]
  • Any one of a number of methods well known in the art can be used to identify the hybridoma cell which produces an antibody with the desired characteristics. These include screening the hybridomas with an ELISA assay, western blot analysis, or radioimmunoassay (Lutz, et al., Exp. Cell Research. 175:109-124 (1988)). [0130]
  • Hybridomas secreting the desired antibodies are cloned and the class and subclass is determined using procedures known in the art (Campbell, A. M., Monoclonal Antibody Technology: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1984)). [0131]
  • Techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce single chain antibodies to proteins of the present invention. [0132]
  • For polyclonal antibodies, antibody containing antiserum is isolated from the immunized animal and is screened for the presence of antibodies with the desired specificity using one of the above-described procedures. [0133]
  • The present invention further provides the above-described antibodies in detectably labeled form. Antibodies can be detectably labeled through the use of radioisotopes, affinity labels (such as biotin, avidin, etc.), enzymatic labels (such as horseradish peroxidase, alkaline phosphatase, etc.) fluorescent labels (such as FITC or rhodamine, etc.), paramagnetic atoms, etc. Procedures for accomplishing such labeling are well-known in the art, for example, see (Sternberger, L. A. et al., J. Histochem. Cytochem. 18:315 (1970); Bayer, E. A. et al., Meth. Enzym. 62:308 (1979); Engval, E. et al., Immunol. 109:129 (1972); Goding, J. W. J. Immunol. Meth. 13:215 (1976)). [0134]
  • The labeled antibodies of the present invention can be used for in vitro, in vivo, and in situ assays to identify cells or tissues in which a fragment of the polypeptide of interest is expressed. The antibodies may also be used directly in therapies or other diagnostics. [0135]
  • The present invention further provides the above-described antibodies immobilized on a solid support. Examples of such solid supports include plastics such as polycarbonate, complex carbohydrates such as agarose and sepharose, acrylic resins and polyacrylamide and latex beads. Techniques for coupling antibodies to such solid supports are well known in the art (Weir, D. M. et al., “Handbook of Experimental Immunology” 4th Ed., Blackwell Scientific Publications, Oxford, England, Chapter 10 (1986); Jacoby, W. D. et al., Meth. Enzym. 34 Academic Press, N.Y. (1974)). The immobilized antibodies of the present invention can be used for in vitro, in vivo, and in situ assays as well as for immuno-affinity purification of the proteins of the present invention. [0136]
  • 6.10 Computer Readable Sequences [0137]
  • In one application of this embodiment, a nucleotide sequence of the present invention can be recorded on computer readable media. As used herein, “computer readable media” refers to any medium which can be read and accessed directly by a computer. Such media include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage medium, and magnetic tape; optical storage media such as CD-ROM; electrical storage media such as RAM and ROM; and hybrids of these categories such as magnetic/optical storage media. A skilled artisan can readily appreciate how any of the presently known computer readable mediums can be used to create a manufacture comprising computer readable medium having recorded thereon a nucleotide sequence of the present invention. [0138]
  • As used herein, “recorded” refers to a process for storing information on computer readable medium. A skilled artisan can readily adopt any of the presently known methods for recording information on computer readable medium to generate manufactures comprising the nucleotide sequence information of the present invention. A variety of data storage structures are available to a skilled artisan for creating a computer readable medium having recorded thereon a nucleotide sequence of the present invention. The choice of the data storage structure will generally be based on the means chosen to access the stored information. In addition, a variety of data processor programs and formats can be used to store the nucleotide sequence information of the present invention on computer readable medium. The sequence information can be represented in a word processing text file, formatted in commercially-available software such as WordPerfect and Microsoft Word, or represented in the form of an ASCII file, stored in a database application, such as DB2, Sybase, Oracle, or the like. A skilled artisan can readily adapt any number of dataprocessor structuring formats (e.g. text file or database) in order to obtain computer readable medium having recorded thereon the nucleotide sequence information of the present invention. [0139]
  • By providing the nucleotide sequence of SEQ ID NO: 1, 2, 24 or 26, a representative fragment thereof, or a nucleotide sequence at least 99.9% identical to SEQ ID NO: 1, 2, 24 or 26 in computer readable form, a skilled artisan can routinely access the sequence information for a variety of purposes. Computer software is publicly available which allows a skilled artisan to access sequence information provided in a computer readable medium. Software which implements the BLAST (Altschul, et al., J. Mol. Biol. 215:403-410 (1990)) and BLAZE (Brutlag, et al., Comp. Chem. 17:203-207 (1993)) search algorithms on a Sybase system may be used to identify open reading frames (ORFs) within a nucleic acid sequence. Such ORFs may be protein encoding fragments and may be useful in producing commercially important proteins such as enzymes used in fermentation reactions and in the production of commercially useful metabolites. [0140]
  • As used herein, “a computer-based system” refers to the hardware means, software means, and data storage means used to analyze the nucleotide sequence information of the present invention. The minimum hardware means of the computer-based systems of the present invention comprises a central processing unit (CPU), input means, output means, and data storage means. A skilled artisan can readily appreciate that any one of the currently available computer-based systems is suitable for use in the present invention. [0141]
  • As stated above, the computer-based systems of the present invention comprise a data storage means having stored therein a nucleotide sequence of the present invention and the necessary hardware means and software means for supporting and implementing a search means. As used herein, “data storage means” refers to memory which can store nucleotide sequence information of the present invention, or a memory access means which can access manufactures having recorded thereon the nucleotide sequence information of the present invention. [0142]
  • As used herein, “search means” refers to one or more programs which are implemented on the computer-based system to compare a target sequence or target structural motif with the sequence information stored within the data storage means. Search means are used to identify fragments or regions of a known sequence which match a particular target sequence or target motif. A variety of known algorithms are disclosed publicly and a variety of commercially available software for conducting search means are and can be used in the computer-based systems of the present invention. Examples of such software includes, but is not limited to, MacPattern (EMBL), BLASTN and BLASTA (NPOLYPEPTIDEIA). A skilled artisan can readily recognize that any one of the available algorithms or implementing software packages for conducting homology searches can be adapted for use in the present computer-based systems. [0143]
  • As used herein, a “target sequence” can be any nucleic acid or amino acid sequence of six or more nucleotides or two or more amino acids. A skilled artisan can readily recognize that the longer a target sequence is, the less likely a target sequence will be present as a random occurrence in the database. The most preferred sequence length of a target sequence is from about 10 to 100 amino acids or from about 30 to 300 nucleotide residues. However, it is well recognized that searches for commercially important fragments, such as sequence fragments involved in gene expression and protein processing, may be of shorter length. [0144]
  • As used herein, “a target structural motif,” or “target motif,” refers to any rationally selected sequence or combination of sequences in which the sequence(s) are chosen based on a three-dimensional configuration which is formed upon the folding of the target motif. There are a variety of target motifs known in the art. Protein target motifs include, but are not limited to, enzyme active sites and signal sequences. Nucleic acid target motifs include, but are not limited to, promoter sequences, hairpin structures and inducible expression elements (protein binding sequences). [0145]
  • 6.11 Expression Modulating Sequences [0146]
  • EMF sequences can be identified within a genome by their proximity to the ORFs. An intergenic segment, or a fragment of the intergenic segment, from about 10 to 200 nucleotides in length, taken 5′ from any ORF will modulate the expression of an operably linked 3′ ORF in a fashion similar to that found with the naturally linked ORF sequence. As used herein, an “intergenic segment” refers to the fragments of a genome which are between two ORF(S) herein described. Alternatively, EMFs can be identified using known EMFs as a target sequence or target motif in the computer-based systems of the present invention. [0147]
  • The presence and activity of an EMF can be confirmed using an EMF trap vector. An EMF trap vector contains a [0148] cloning site 5′ to a marker sequence. A marker sequence encodes an identifiable phenotype, such as antibiotic resistance or a complementing nutrition auxotrophic factor, which can be identified or assayed when the EMF trap vector is placed within an appropriate host under appropriate conditions, As described above, an EMF will modulate the expression of an operably linked marker sequence. A more detailed discussion of various marker sequences is provided below. A sequence which is suspected of being an EMF is cloned in all three reading frames in one or more restriction sites upstream from the marker sequence in the EMF trap vector. The vector is then transformed into an appropriate host using known procedures and the phenotype of the transformed host is examined under appropriate conditions. As described above, an EMF will modulate the expression of an operably linked marker sequence.
  • 6.12 Triplex Helix Formation [0149]
  • In addition, the fragments of the present invention, as broadly described, can be used to control gene expression through triple helix formation or antisense DNA or RNA, both of which methods are based on the binding of a polynucleotide sequence to DNA or RNA. Polynucleotides suitable for use in these methods are usually 20 to 40 bases in length and are designed to be complementary to a region of the gene involved in transcription (triple helix—see Lee, et al., Nucl. Acids Res. 6:3073 (1979); Cooney, et al., Science 15241:456 (1988); and Dervan, et al., Science 251:1360 (1991)) or to the mRNA itself (antisense—Olmno, J. Neurochem. 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)). [0150]
  • Triple helix- formation optimally results in a shut-off of RNA transcription from DNA, while antisense RNA hybridization blocks translation of an mRNA molecule into polypeptide. Both techniques have been demonstrated to be effective in model systems. Information contained in the sequences of the present invention is necessary for the design of an antisense or triple helix oligonucleotide. [0151]
  • 6.13 Diagnostic Assays and Kits [0152]
  • The present invention further provides methods to identify the expression of one of the ORFs of the present invention, or homolog thereof, in a test sample, using a nucleic acid probe or antibodies of the present invention. [0153]
  • In detail, such methods comprise incubating a test sample with one or more of the antibodies or one or more of nucleic acid probes of the present invention and assaying for binding of the nucleic acid probes or antibodies to components within the test sample. [0154]
  • Conditions for incubating a nucleic acid probe or antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the nucleic acid probe or antibody used in the assay. One skilled in the art will recognize that any one of the commonly available hybridization, amplification or immunological assay formats can readily be adapted to employ the nucleic acid probes or antibodies of the present invention. Examples of such assays can be found in Chard, T., An Introduction to Radioimmunoassay and Related Techniques, Elsevier Science Publishers, Amsterdam, The Netherlands (1986); Bullock, G. R. et al., Techniques in Immunocytochemistry, Academic Press, Orlando, Fla. Vol. 1 (1982), Vol. 2 (1983), Vol. 3 (1985); Tijssen, P., Practice and Theory of immunoassays: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1985). [0155]
  • The test samples of the present invention include cells, protein or membrane extracts of cells, or biological fluids such as sputum, blood, serum, plasma, or urine. The test sample used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed. Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can be readily be adapted in order to obtain a sample which is compatible with the system utilized. [0156]
  • In another embodiment of the present invention, kits are provided which contain the necessary reagents to carry out the assays of the present invention. [0157]
  • Specifically, the invention provides a compartment kit to receive, in close confinement, one or more containers which comprises: (a) a first container comprising one of the probes or antibodies of the present invention; and (b) one or more other containers comprising one or more of the following: wash reagents, reagents capable of detecting presence of a bound probe or antibody. [0158]
  • In detail, a compartment kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers or strips of plastic or paper. Such containers allow one to efficiently transfer reagents from one compartment to another compartment such that the samples and reagents are not cross-contaminated, and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another. Such containers will include a container which will accept the test sample, a container which contains the antibodies used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents used to detect the bound antibody or probe. [0159]
  • Types of detection reagents include labeled nucleic acid probes, labeled secondary antibodies, or in the alternative, if the primary antibody is labeled, the enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody. One skilled in the art will readily recognize that the disclosed probes and antibodies of the present invention can be readily incorporated into one of the established kit formats which are well known in the art. [0160]
  • 6.14 Screening Assays [0161]
  • Using the isolated proteins of the present invention, the present invention further provides methods of obtaining and identifying agents which bind to a protein encoded by one of the ORFs from a nucleic acid with a sequence of one of SEQ ID NO: 1, 2, 24 or 26, or to a nucleic acid with a sequence of one of SEQ ID NO: 1, 2, 24 or 26. [0162]
  • In detail, said method comprises the steps of: (a) contacting an agent with an isolated protein encoded by one of the ORFs of the present invention, or nucleic acid of the invention; and (b) determining whether the agent binds to said protein or said nucleic acid. [0163]
  • The agents screened in the above assay can be, but are not limited to, peptides, carbohydrates, vitamin derivatives, or other pharmaceutical agents. The agents can be selected and screened at random or rationally selected or designed using protein modeling techniques. [0164]
  • For random screening, agents such as peptides, carbohydrates, pharmaceutical agents and the like are selected at random and are assayed for their ability to bind to the protein encoded by the ORF of the present invention. [0165]
  • Alternatively, agents may be rationally selected or designed. As used herein, an agent is said to be “rationally selected or designed” when the agent is chosen based on the configuration of the particular protein. For example, one skilled in the art can readily adapt currently available procedures to generate peptides, pharmaceutical agents and the like capable of binding to a specific peptide sequence in order to generate rationally designed antipeptide peptides, for example see Hurby, et al., Application of Synthetic Peptides: Antisense Peptides,” In Synthetic Peptides, A User's Guide, W. H. Freeman, N.Y. (1992), pp. 289-307, and Kaspczak, et al., Biochemistry 28:9230-8 (1989), or pharmaceutical agents, or the like. [0166]
  • In addition to the foregoing, one class of agents of the present invention, as broadly described, can be used to control gene expression through binding to one of the ORFs or EMFs of the present invention. As described above, such agents can be randomly screened or rationally designed/selected. Targeting the ORF or EMF allows a skilled artisan to design sequence specific or element specific agents, modulating the expression of either a single ORF or multiple ORFs which rely on the same EMF fore expression control. [0167]
  • One class of DNA binding agents are agents which contain base residues which hybridize or form a triple helix formation by binding to DNA or RNA. Such agents can be based on the classic phosphodiester, ribonucleic acid backbone, or can be a variety of sulfhydryl or polymeric derivatives which have base attachment capacity. [0168]
  • Agents suitable for use in these methods usually contain 20 to 40 bases and are designed to be complementary to a region of the gene involved in transcription (triple helix—see Lee, et al., Nucl. Acids Res. 6:3073 (1979); Cooney, et al., Science 241:456 (1988); and Dervan, et al., Science 251:1360 (1991)) or to the mRNA itself (antisense—Okano, J. Neurochem. 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)). Triple helix- formation optimally results in a shut-off of RNA transcription from DNA, while antisense RNA hybridization blocks translation of an mRNA molecule into polypeptide. Both techniques have been demonstrated to be effective in model systems. Information contained in the sequences of the present invention is necessary for the design of an antisense or triple helix oligonucleotide and other DNA binding agents. [0169]
  • Agents which bind to a protein encoded by one of the ORFs of the present invention can be used as a diagnostic agent, in the control of bacterial infection by modulating the activity of the protein encoded by the ORF. Agents which bind to a protein encoded by one of the ORFs of the present invention can be formulated using known techniques to generate a pharmaceutical composition. [0170]
  • 6.15 Use of Nucleic Acids as Probes [0171]
  • Another aspect of the subject invention is to provide for polypeptide-specific nucleic acid hybridization probes capable of hybridizing with naturally occurring nucleotide sequences. The hybridization probes of the subject invention may be derived from the nucleotide sequence of the SEQ ID NO: 1, 2, 24 or 26. Because the corresponding gene is expressed in only one out of 18 tissues tested, namely macrophages, a hybridization probe derived from SEQ ID NO: 1, 2, 24 or 26 can be used as an indicator of the presence of macrophage RNA in a sample. Any suitable hybridization technique can be employed, such as, for example, in situ hybridization. [0172]
  • PCR as described U.S. Pat. Nos. 4,683,195 and 4,965,188 provides additional uses for oligonucleotides based upon the nucleotide sequences. Such probes used in PCR may be of recombinant origin, may be chemically synthesized, or a mixture of both. The probe will comprise a discrete nucleotide sequence for the detection of identical sequences or a degenerate pool of possible sequences for identification of closely related genomic sequences. [0173]
  • Other means for producing specific hybridization probes for nucleic acids include the cloning of nucleic acid sequences into vectors for the production of mRNA probes. Such vectors are known in the art and are commercially available and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerase as T7 or SP6 RNA polymerase and the appropriate radioactively labeled nucleotides. [0174]
  • The nucleotide sequences may be used to construct hybridization probes for mapping their respective genomic sequences. The nucleotide sequence provided herein may be mapped to a chromosome or specific regions of a chromosome using well known genetic and/or chromosomal mapping techniques. These techniques include in situ hybridization, linkage analysis against known chromosomal markers, hybridization screening with libraries or flow-sorted chromosomal preparations specific to known chromosomes, and the like. The technique of fluorescent in situ hybridization of chromosome spreads has been described, among other places, in Verma, et al (1988) Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York N.Y. Fluorescent in situ hybridization of chromosomal preparations and other physical chromosome mapping techniques may be correlated with additional genetic map data. Examples of genetic map data can be found in the 1994 Genome Issue of Science (265:1981f). Correlation between the location of a nucleic acid on a physical chromosomal map and a specific disease (or predisposition to a specific disease) may help delimit the region of DNA associated with that genetic disease. The nucleotide sequences of the subject invention may be used to detect differences in gene sequences between normal, carrier or affected individuals. [0175]
  • The nucleotide sequence may be used to produce purified polypeptides using well known methods of recombinant DNA technology. Among the many publications that teach methods for the expression of genes after they have been isolated is Goeddel, (1990) Gene Expression Technology, Methods and Enzymology, Vol 185, Academic Press, San Diego. Polypeptides may be expressed in a variety of host cells, either prokaryotic or eukaryotic. Host cells may be from the same species from which a particular polypeptide nucleotide sequence was isolated or from a different species. Advantages of producing polypeptides by recombinant DNA technology include obtaining adequate amounts of the protein for purification and the availability of simplified purification procedures. [0176]
  • Each sequence so obtained was compared to sequences in GenBank using a search algorithm developed by Applied Biosystems and incorporated into the INHERIT™ 670 Sequence Analysis System. In this algorithm, Pattern Specification Language (developed by TRW Inc., Los Angeles, Calif.) was used to determine regions of homology. The three parameters that determine how the sequence comparisons run were window size, window offset, and error tolerance. Using a combination of these three parameters, the DNA database was searched for sequences containing regions of homology to the query sequence, and the appropriate sequences were scored with an initial value. Subsequently, these homologous regions were examined using dot matrix homology plots to distinguish regions of homology from chance matches. Smith-Waterman alignments were used to display the results of the homology search. [0177]
  • Peptide and protein sequence homologies were ascertained using the INHERIT™ 670 Sequence Analysis System in a way similar to that used in DNA sequence homologies. Pattern Specification Language and parameter windows were used to search protein databases for sequences containing regions of homology which were scored with an initial value. Dot-matrix homology plots were examined to distinguish regions of significant homology from chance matches. [0178]
  • Alternatively, BLAST, which stands for Basic Local Alignment Search Tool, is used to search for local sequence alignments (Altschul, S. F., (1993) J Mol Evol 36:290-300; Altschul, S. F., et al (1990) J Mol Biol 215:403-10). BLAST produces alignments of both nucleotide and amino acid sequences to determine sequence similarity. Because of the local nature of the alignments, BLAST is especially useful in determining exact matches or in identifying homologs. Whereas it is ideal for matches which do not contain gaps, it is inappropriate for performing motif-style searching. The fundamental unit of BLAST algorithm output is the High-scoring Segment Pair (HSP). [0179]
  • An HSP consists of two sequence fragments of arbitrary but equal lengths whose alignment is locally maximal and for which the alignment score meets or exceeds a threshold or cutoff score set by the user. The BLAST approach is to look for HSPs between a query sequence and a database sequence, to evaluate the statistical significance of any matches found, and to report only those matches which satisfy the user-selected threshold of significance. The parameter E establishes the statistically significant threshold for reporting database sequence matches. E is interpreted as the upper bound of the expected frequency of chance occurrence of an HSP (or set of HSPs) within the context of the entire database search. Any database sequence whose match satisfies E is reported in the program output. [0180]
  • In addition, BLAST analysis was used to search for related molecules within the libraries of the LIFESEQ™ database. This process, an “electronic northern” analysis is analogous to northern blot analysis in that it uses one cellubrevin sequence at a time to search for identical or homologous molecules at a set stringency. The stringency of the electronic northern is based on “product score”. The product score is defined as (% nucleotide or amino acid [between the query and reference sequences] in Blast multiplied by the % maximum possible BLAST score [based on the lengths of query and reference sequences]) divided by 100. At a product score of 40, the match will be exact within a 1-2% error; and at 70, the match will be exact. Homologous or related molecules can be identified by selecting those which show product scores between approximately 15 and 30. [0181]
  • 6.16 SEQ ID NOs:1-8, 23 - 24 and 26 - 27 [0182]
  • Referring to FIG. 1, SEQ ID NO:1 is the nucleotide sequence of an expressed sequence tag corresponding to a polynucleotide isolated from a cDNA library of human fetal liver-spleen. SEQ ID NO:2 is an extended version of SEQ ID NO:1 obtained as described in Example 2, and the encoded polypeptide in SEQ ID NO: 3 is referred to herein as CD39-L4. SEQ ID NO:2 encodes a polypeptide having the amino acid sequence of SEQ ID NO:3 (shown in FIG. 2). The open reading frame corresponding to SEQ ID NO:3 starts at [0183] nucleotide 246, as numbered from the 5′ end of SEQ ID NO:2. This open reading frame encodes a polypeptide 428 amino acids in length. The estimated molecular weight of the unglycosylated polypeptide is approximately 47.52 kDa.
  • Protein database searches with the BLAST algorithm indicate that SEQ ID NO:3 is homologous to the CD39 family. FIG. 3 shows the amino acid sequence alignment between SEQ ID NO:3 (identified as “246 prot”) and human CD39 (“CD39Human.seq”), indicating that the two sequences share 30% amino acid sequence identity. Moreover, a higher degree of homology between the apyrase conserved regions (Kaczmarek et al., J. Biol. Chem. 271:33116-33122 (1996) is observed. In particular, an almost perfect match to a putative ATP-binding region was found from amino acids 54-58, DAGST (DAGSS in CD39). In addition, the DLGGASTQ motif (DLGGASTQ in CD39), which is very well conserved among ATPDases, is found from amino acids 199-206 in SEQ ID NO:3. Other regions conserved in apyrases were found from amino acids 129-134, ATAGLR (ATAGMR in CD39) and from amino acids 169-173, GSDEG (GQEEG in CD39). [0184]
  • SEQ ID NO:3 differs from CD39 in that SEQ ID NO:3 contains a hydrophobic stretch of 22 amino acids at its amino terminus, which is indicative of a leader peptide. SEQ ID NO:3 also lacks the transmembrane domain found at the carboxyl terminus of CD39. These features indicate that SEQ ID NO:3 is a soluble ATPDase. [0185]
  • SEQ ID NO:3 shares an even higher degree of homology (86% identity) with a murine NTPase, as shown in the amino acid sequence alignment presented in FIG. 4 (SEQ ID NO:3 is identified as “246 prot,” and mouse CD39 as “mur ntpase”). [0186]
  • The message encoding SEQ ID NO:3 is tightly regulated in a tissue-specific manner. An expression study using a semiquantitative PCR/Southern blot approach revealed a significant level of expression in macrophage. In contrast, human CD39 is expressed in tissues such as placenta, lung, skeletal muscle, kidney, and heart. [0187]
  • SEQ ID NO: 4 is the polynucleotide sequence for CD39-L4. SEQ ID NO: 5 is the corresponding amino acid sequence. [0188]
  • SEQ ID NO: 6 is the polynucleotide sequence for a CD39-L4 variant designated ACRIII, wherein the following amino acid substitutions have been made: D168-T, S170-Q and L175-F; SEQ ID NO: 7 is the corresponding amino acid sequence. [0189]
  • SEQ ID NO: 8 is the genomic sequence for the human CD39-L4 gene; exons appear at nucleotides 1-288 (exon 1), 1281-1580 (exon 2), 1820-1855 (exon 3) 2467-2555 (exon 4), 2863-2942 (exon 5), 3889-3950 (exon 6), 4894-4995 (exon 7), 5847-5987 (exon 8), 6966-7138 (exon 9) and 8556-9365 (exon 10). [0190]
  • SEQ ID NO: 24 is the polynucleotide sequence for a CD39-L4 splice variant that creates an isoform designated CD39-L66. SEQ ID NO: 25 is the corresponding amino acid sequence. [0191]
  • SEQ ID NO: 26 is the polynucleotide sequence for CD39-L2. SEQ ID NO: 27 is the corresponding amino acid sequence. [0192]
  • 6.17 Uses of Novel CD39-Like Polypeptides and Antibodies [0193]
  • Polypeptides of the invention having ATPDase, including NDPase, activity are useful for inhibiting platelet function and can therefore be employed in the prophylaxis or treatment of pathological conditions caused by or involving thrombosis or excessive coagulation or excessive platelet aggregation, such as myocardial infarction, cerebral ischemia, angina, and the like. Polypeptides of the invention can also be used in the maintenance of vascular grafts. Platelet function can be measured by any of a number of standard assays, such as, for example, the platelet aggregation assay described in Example 5. [0194]
  • Such pathological conditions include conditions caused by or involving arterial thrombosis, such as coronary artery thrombosis and resulting myocardial infarction, cerebral artery thrombosis or intracardiac thrombosis (due to, e.g., atrial fibrillation) and resulting stroke, and other peripheral arterial thrombosis and occlusion; conditions associated with venous thrombosis, such as deep venous thrombosis and pulmonary embolism; conditions associated with exposure of the patient's blood to a foreign or injured tissue surface, including diseased heart valves, mechanical heart valves, vascular grafts, and other extracorporeal devices such as intravascular cannulas, vascular access shunts in hemodialysis patients, hemodialysis machines and cardiopulmonary bypass machines; and conditions associated with coagulapathies, such as hypercoagulability and disseminated intravascular coagulopathy. Co-administration of other agents suitable for treating the pathological condition, e.g., other anti-coagulation agents, is also contemplated. [0195]
  • In particular, variants like the ACRIII mutant described herein are expected to be superior therapeutics for treating such pathological conditions because (1) ACRIII exhibits six-fold greater activity compared to wild type CD39-L4, and (2) ACRIII, like CD39-L4, is uniquely specific for ADP and does not hydrolyze ATP. Thus, adverse side effects from hydrolysis of circulating ATP are avoided. [0196]
  • For instance, ATP is known to act as an extracellular signal in many tissues. In the heart, extracellular ATP modulates ionic processes and contractile function (for review see Burnstock, G., Neuropharmacology 36:1127). Recently, it has been shown that extracellular ATP markedly inhibits glucose transport in rat cardiomyocytes (Fisher, Y. et al., J. Biol. Chem. 274:755-761. Another source of extracellular ATP is that released from parenchymal cells under hypoxic or ischemic conditions (Skobel, E., and Kammermeier, H. Biochim. Biophys. Acta 1362:128-134). ATP is also involved in the modulation of anti-IgE-induced release of histamine from human lung mast cells (Schulman, E. S., et al., Am. J. Respir. Cell Mol. Biol. 20:520-537). [0197]
  • Furthermore, the ability of CD39-L4 to hydrolyze NDPs other than ADP has implications outside the circulatory system. For instance, it has been reported that UDP is the most potent agonist for the human P2Y[0198] 6 receptor. Communi, et al., Bioch Bioph Res Com 222:303-308 (1996). This receptor is expressed in several tissues including infiltrating T cells present in inflammatory bowel disease. Somers, et al., Lab Invest 78:1375-1383 (1998). In this microenvironment, a molecule with the enzymatic properties of CD39-L4 could influence T cell responses by modifying the extracellular half-life of UDP. Another role for CD39-L4 has been suggested by the report that mouse CD39-L4 maps closely to a locus associated with audigenic brain seizures in mice. See Chadwick, et al., Genomics 50:357-367 (1998); Seyfried, et al., Genetics 99:117-126 (1981). This locus, known as Asp-1, is thought to be linked or to correspond to a factor that influences Ca2+-ATPase activity. Neumann, et al., Behav. Genetics 20:307-323 (1990).
  • Additionally, the polypeptides of the invention can be used as molecular weight markers, and as a food supplement. A polypeptide consisting of SEQ ID NO:3, for example, has a molecular mass of approximately 47.52 kD in its unglycosylated form. Protein food supplements are well known and the formulation of suitable food supplements including polypeptides of the invention is within the level of skill in the food preparation art. [0199]
  • The polypeptides of the invention are also useful for making antibody substances that are specifically immunoreactive with CD39-like proteins. Antibodies and portions thereof (e.g., Fab fragments) which bind to the polypeptides of the invention can be used to identify the presence of such polypeptides in a sample. For example, the level of the native protein corresponding to SEQ ID NO:3 in a blood sample can be determined as an indication of vascular condition. Such determinations are carried out using any suitable immunoassay format, and any polypeptide of the invention that is specifically bound by the antibody can be employed as a positive control. [0200]
  • Additionally, the polypeptides of the invention are useful for modulating the ratios of levels of adenosine molecules in vivo to regulate homeostasis. Adenosine diphosphate (ADP) is an agonist of platelet activation and aggregation. It has been demonstrated that the P2Y receptor (and others including P2T and P2Y1 and potentially others) transduces this signal. Adenosine triphosphate (ATP) also binds to this receptor, but acts as an antagonist. Therefore, the ratios of levels of ATP/ADP can significantly influence in vivo platelet activation and aggregation. Agents that specifically decrease levels of ADP not only decrease the amount of agonist available to signal, but also increase the relative antagonistic effects of ATP, because of less competition for the common receptor. [0201]
  • The polypeptides of the invention are administered by any route that delivers an effective dosage to the desired site of action. The determination of a suitable route of administration and an effective dosage for a particular indication is within the level of skill in the art. For treatment of vascular disease, polypeptides according to the invention are generally administered intravenously. In vivo murine studies with soluble human CD39 have shown that mice injected intravenously with 50 mg recombinant soluble human CD39 in 100 ml sterile saline had biologically active CD39 in their sera for an extended period of time, with an elimination half-life of almost 2 days (Gayle, R. B., et al., J. Clinical Invest. 101:1851-1859(1998)). Suitable dosage ranges for the polypeptides of the invention can be extrapolated from these dosages or from similar studies in appropriate animal models. Dosages can then be adjusted as necessary by the clinician to provide maximal therapeutic benefit. [0202]
  • 6.18 PHARMACEUTICAL FORMULATIONS AND ROUTES OF ADMINISTRATION [0203]
  • A protein of the present invention (from whatever source derived, including without limitation from recombinant and non-recombinant sources) may be administered to a patient in need, by itself, or in pharmaceutical compositions where it is mixed with suitable carriers or excipient(s) at doses to treat or ameliorate a variety of disorders. Such a composition may also contain (in addition to protein and a carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration. The pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL4, IL-5, IL6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IFN, TNF0, TNF1, TNF2, G-CSF, Meg-CSF, thrombopoietin, stem cell factor, and erythropoietin. [0204]
  • The pharmaceutical composition may further contain other agents which either enhance the activity of the protein or compliment its activity or use in treatment. For example, CD39-L2 or CD39-L4 may be co-administered with platelet ADP receptor antagonists, e.g. ATP derivatives, ADP derivatives. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect with protein of the invention, or to minimize side effects. Conversely, protein of the present invention may be included in formulations of the particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to minimize side effects of the cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent. A protein of the present invention may be active in multimers (e.g., heterodimers or homodimers) or complexes with itself or other proteins. As a result, pharmaceutical compositions of the invention may comprise a protein of the invention in such multimeric or complexed form. [0205]
  • Techniques for formulation and administration of the compounds of the instant application may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition. A therapeutically effective dose further refers to that amount of the compound sufficient to result in amelioration of symptoms, e.g., treatment, healing, prevention or amelioration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions. When applied to an individual active ingredient, administered alone, a therapeutically effective dose refers to that ingredient alone. When applied to a combination, a therapeutically effective dose refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously. [0206]
  • In practicing the method of treatment or use of the present invention, a therapeutically effective amount of protein of the present invention is administered to a mammal having a condition to be treated. Protein of the present invention may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors. When co-administered with one or more cytokines, lymphokines or other hematopoietic factors, protein of the present invention may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering protein of the present invention in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors. [0207]
  • 6.18.1. ROUTES OF ADMINISTRATION [0208]
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.Administration of protein of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, topical application or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Intravenous administration to the patient is preferred. [0209]
  • Alternately, one may administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into a arthritic joints or in fibrotic tissue, often in a depot or sustained release formulation. In order to prevent the scarring process frequently occurring as complication of glaucoma surgery, the compounds may be administered topically, for example, as eye drops.Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with a specific antibody, targeting, for example, arthritic or fibrotic tissue. The liposomes will be targeted to and taken up selectively by the afflicted tissue. [0210]
  • 6.18.2. COMPOSITIONS/FORMULATIONS [0211]
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. These pharmaceutical compositions may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.Proper formulation is dependent upon the route of administration chosen. When a therapeutically effective amount of protein of the present invention is administered orally, protein of the present invention will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder contain from about 5 to 95% protein of the present invention, and preferably from about 25 to 90% protein of the present invention. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of protein of the present invention, and preferably from about 1 to 50% protein of the present invention. [0212]
  • When a therapeutically effective amount of protein of the present invention is administered by intravenous, cutaneous or subcutaneous injection, protein of the present invention will be in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable protein solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to protein of the present invention, an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0213]
  • For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [0214]
  • Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. [0215]
  • For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0216]
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. [0217]
  • The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. [0218]
  • A pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent system may be the VPD co-solvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the [0219] nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPDD:5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various of sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
  • The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Many of the proteinase inhibiting compounds of the invention may be provided as salts with pharmaceutically compatible counterions. Such pharmaceutically acceptable base addition salts are those salts which retain the biological effectiveness and properties of the free acids and which are obtained by reaction with inorganic or organic bases such as sodium hydroxide, magnesium hydroxide, ammonia, trialkylamine, dialkylamine, monoalkylamine, dibasic amino acids, sodium acetate, potassium benzoate, triethanol amine and the like. [0220]
  • The pharmaceutical composition of the invention may be in the form of a complex of the protein(s) of present invention along with protein or peptide antigens. The protein and/or peptide antigen will deliver a stimulatory signal to both B and T lymphocytes. B lymphocytes will respond to antigen through their surface immunoglobulin receptor. T lymphocytes will respond to antigen through the T cell receptor (TCR) following presentation of the antigen by MHC proteins. MHC and structurally related proteins including those encoded by class I and class II MHC genes on host cells will serve to present the peptide antigen(s) to T lymphocytes. The antigen components could also be supplied as purified MHC-peptide complexes alone or with co-stimulatory molecules that can directly signal T cells. Alternatively antibodies able to bind surface immunoglobulin and other molecules on B cells as well as antibodies able to bind the TCR and other molecules on T cells can be combined with the pharmaceutical composition of the invention. The pharmaceutical composition of the invention may be in the form of a liposome in which protein of the present invention is combined, in addition to other pharmaceutically acceptable carriers, with amphipathic agents such as lipids which exist in aggregated form as micelles, insoluble monolayers, liquid crystals, or lamellar layers in aqueous solution. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. Nos. 4,235,871; 4,501,728; 4,837,028; and 4,737,323, all of which are incorporated herein by reference. [0221]
  • The amount of protein of the present invention in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of protein of the present invention with which to treat each individual patient. Initially, the attending physician will administer low doses of protein of the present invention and observe the patient's response. Larger doses of protein of the present invention may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further. It is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.01 μg to about 100 mg (preferably about 0.1 μg to about 10 mg, more preferably about 0.1 μg to about 1 mg) of protein of the present invention per kg body weight. For compositions of the present invention which are useful for bone, cartilage, tendon or ligament regeneration, the therapeutic method includes administering the composition topically, systematically, or locally as an implant or device. When administered, the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form. Further, the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage or tissue damage. Topical administration may be suitable for wound healing and tissue repair. Therapeutically useful agents other than a protein of the invention which may also optionally be included in the composition as described above, may alternatively or additionally, be administered simultaneously or sequentially with the composition in the methods of the invention. Preferably for bone and/or cartilage formation, the composition would include a matrix capable of delivering the protein-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and optimally capable of being resorbed into the body. Such matrices may be formed of materials presently in use for other implanted medical applications. [0222]
  • The choice of matrix material is based on biocompatibility, biodegradability, mechanical properties, cosmetic appearance and interface properties. The particular application of the compositions will define the appropriate formulation. Potential matrices for the compositions may be biodegradable and chemically defined calcium sulfate, tricalciumphosphate, hydroxyapatite, polylactic acid, polyglycolic acid and polyanhydrides. Other potential materials are biodegradable and biologically well-defined, such as bone or dermal collagen. Further matrices are comprised of pure proteins or extracellular matrix components. Other potential matrices are nonbiodegradable and chemically defined, such as sintered hydroxyapatite, bioglass, aluminates, or other ceramics. Matrices may be comprised of combinations of any of the above mentioned types of material, such as polylactic acid and hydroxyapatite or collagen and tricalciumphosphate. The bioceramics may be altered in composition, such as in calcium-aluminate-phosphate and processing to alter pore size, particle size, particle shape, and biodegradability. Presently preferred is a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns. In some applications, it will be useful to utilize a sequestering agent, such as carboxymethyl cellulose or autologous blood clot, to prevent the protein compositions from disassociating from the matrix. [0223]
  • A preferred family of sequestering agents is cellulosic materials such as alkylcelluloses (including hydroxyalkylcelluloses), including methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropyl-methylcellulose, and carboxymethylcellulose, the most preferred being cationic salts of carboxymethylcellulose (CMC). Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer and poly(vinyl alcohol). The amount of sequestering agent useful herein is 0.5-20 wt %, preferably 1-10 wt % based on total formulation weight, which represents the amount necessary to prevent desorbtion of the protein from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the protein the opportunity to assist the osteogenic activity of the progenitor cells. In further compositions, proteins of the invention may be combined with other agents beneficial to the treatment of the bone and/or cartilage defect, wound, or tissue in question. These agents include various growth factors such as epidermal growth factor (EGF), platelet derived growth factor (PDGF), transforming growth factors (TGF-.alpha. and TGF-.beta.), and insulin-like growth factor (IGF). [0224]
  • The therapeutic compositions are also presently valuable for veterinary applications. Particularly domestic animals and thoroughbred horses, in addition to humans, are desired patients for such treatment with proteins of the present invention. The dosage regimen of a protein-containing pharmaceutical composition to be used in tissue regeneration will be determined by the attending physician considering various factors which modify the action of the proteins, e.g., amount of tissue weight desired to be formed, the site of damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue (e.g., bone), the patient's age, sex, and diet, the severity of any infection, time of administration and other clinical factors. The dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition. For example, the addition of other known growth factors, such as IGF I (insulin like growth factor I), to the final composition, may also effect the dosage. Progress can be monitored by periodic assessment of tissue/bone growth and/or repair, for example, X-rays, histomorphometric determinations and tetracycline labeling. [0225]
  • Polynucleotides of the present invention can also be used for gene therapy. Such polynucleotides can be introduced either in vivo or ex vivo into cells for expression in a mammalian subject. Polynucleotides of the invention may also be administered by other known methods for introduction of nucleic acid into a cell or organism (including, without limitation, in the form of viral vectors or naked DNA). Cells may also be cultured ex vivo in the presence of proteins of the present invention in order to proliferate or to produce a desired effect on or activity in such cells. Treated cells can then be introduced in vivo for therapeutic purposes. [0226]
  • 6.18.3. EFFECTIVE DOSAGE [0227]
  • Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC[0228] 50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the C-proteinase activity). Such information can be used to more accurately determine useful doses in humans.
  • A therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms or a prolongation of survival in a patient. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD[0229] 50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. See, e.g., Fingl et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p.1.Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the C-proteinase inhibiting effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; for example, the concentration necessary to achieve 50-90% inhibition of the C-proteinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. [0230]
  • The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. [0231]
  • 6.18.4. PACKAGING [0232]
  • The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labelled for treatment of an indicated condition. [0233]
  • The present invention is illustrated in the following examples. Upon consideration of the present disclosure, one of skill in the art will appreciate that many other embodiments and variations may be made in the scope of the present invention. Accordingly, it is intended that the broader aspects of the present invention not be limited to the disclosure of the following examples. [0234]
  • EXAMPLE 1 Isolation of SEQ ID NO:1 From a cDNA Library of Human Fetal Liver-Spleen
  • A plurality of novel nucleic acids were obtained from a b2HFLS20W cDNA library prepared from human fetal liver-spleen, as described in Bonaldo et al., Genome Res. 6:791-806 (1996), using standard PCR, Sequencing by hybridization sequence signature analysis, and Sanger sequencing techniques. The inserts of the library were amplified with PCR using primers specific for vector sequences flanking the inserts. These samples were spotted onto nylon membranes and interrogated with oligonucleotide probes to give sequence signatures. The clones were clustered into groups of similar or identical sequences, and single representative clones were selected from each group for gel sequencing. The 5′ sequence of the amplified inserts was then deduced using the reverse M13 sequencing primer in a typical Sanger sequencing protocol. PCR products were purified and subjected to fluorescent dye terminator cycle sequencing. Single-pass gel sequencing was done using a 377 Applied Biosystems (ABI) sequencer. One of these inserts was identified as a novel sequence not previously obtained from this library and not previously reported in public databases. This sequence is shown in FIG. 1 as SEQ ID NO:1. [0235]
  • EXAMPLE 2 Isolation of SEQ ID NO:2 and Determination of a Nucleotide Sequence Encoding a 428-Amino Acid Protein with Sequence Homology to CD39
  • The nucleotide sequence shown in FIG. 1, and labeled SEQ ID NO:2, encodes the translated amino acid sequence SEQ ID NO:3, which is shown in FIG. 2. The extended nucleotide sequence was obtained by isolating colonies generated from pools of clones from a human macrophage cDNA library (Invitrogen, Cat. # A550-25). Briefly, the macrophage cDNA library was plated on LB/Amp plates (containing 100 mg/ml ampicillin) at a density of about 40,000 colonies/plate. The colonies were lifted onto nitrocellulose filters and hybridized with a radiolabeled probe generated from the original clone (i.e., SEQ ID NO:1). [0236]
  • That the identified clones corresponded to SEQ ID NOs:1 and 2 was confirmed by using gene-specific primers (5′-GCTACCTCACTTCCTTTGAG-3′ [SEQ ID NO: 9] and 5′-CTGGCTGGTGAAGTTTTCCTC-3′ [SEQ ID NO: 10]) in a PCR-based assay. Then PCR using vector- and gene-specific primers was employed to amplify the 5′ portion of the cDNA. Nested primers were used to generate sequence from the amplified product(s). Laser gene™ software was used to edit and “contig” the partial sequences into a full-length sequence. As discussed above, the amino acid sequence has striking homology to CD39, which is involved in modulating platelet reactivity during vascular inflammation. Based in part on the observed sequence similarity to CD39, the polypeptide encoded by SEQ ID NO: 2 was designated CD39-L4. [0237]
  • EXAMPLE 3 A. Expression of SEQ ID NOS. 3 and 5 in COS-7 Cells
  • COS-7 cells were grown in DMEM (ATCC) and 10% fetal bovine serum (FBS) (Gibco) to 70% confluence. Prior to transfection the media was changed to DMEM and 0.5% FCS. Cells were transfected with cDNAs for SEQ ID NOs. 3 and 5 or with pBGaI vector by the FuGENE-6 transfection reagent (Boehringer). In summary, 4 μl of FuGENE6 was diluted in 100 μl of DMEM and incubated for 5 minutes. Then, this was added to 1 μg of DNA and incubated for 15 minutes before adding it to a 35 mm dish of COS-7 cells. The COS-7 cells were incubated at 37° C. with 5% CO[0238] 2. After 24 hours, media and cell lysates were collected, centrifuged and dialyzed against assay buffer (15 mM Tris pH 7.6, 134 mM NaCl, 5 mM glucose, 3 mM CaCl2 and MgCl2. More robust expression can be achieved using the protocol described in Example 6 below.
  • B. Expression Study Using SEQ ID NO:2
  • The expression of SEQ.ID NO. 2 in various tissues was analyzed using a semi-quantitative polymerase chain reaction-based technique. Human cDNA libraries were used as sources of expressed genes from tissues of interest (adult brain, adult heart, adult kidney, adult lymph node, adult liver, adult lung, adult ovary, adult placenta, adult spleen, adult testis, bone marrow, fetal kidney, fetal liver, fetal liver-spleen, fetal skin, fetal brain, fetal leukocyte and macrophage). Gene-specific primers (5′-GCTACCTCACTTCCTTTGAG-3′ [SEQ ID NO: 9] and 5′-GCAGGTCTCCAAGGAAGTACG-3′ [SEQ ID NO: 11]) were used to amplify portions of the SEQ ID NO:2 sequence from the samples. Amplified products were separated on an agarose gel, transferred and chemically linked to a nylon filter. The filter was then hybridized with a radioactively labeled (α[0239] 33P-dCTP) double-stranded probe generated from the full-length SEQ ID NO:2 sequence using a Klenow polymerase, random-prime method. The filters were washed (high stringency) and used to expose a phosphorimaging screen for several hours. Bands indicated the presence of cDNA including SEQ ID NO:2 sequences in a specific library, and thus mRNA expression in the corresponding cell type or tissue.
  • Of the 18 human tissues tested, macrophage was the only sample that provided a signal, indicating that expression of SEQ ID NO:2 is tightly regulated. In contrast, the CD39 molecule has been found in tissues such as placenta, lung, skeletal muscle, kidney and heart. [0240]
  • EXAMPLE 4 Chromosomal Localization of the Gene Corresponding to SEQ ID NOs:1 and 2
  • Chromosome mapping technologies allow investigators to link genes to specific regions of chromosomes. Assignment to [0241] chromosome 14 was performed with the Coriell cell repository monochromosomal panel #2 (NIGMS cell repository). This human rodent somatic cell hybrid panel consists of DNA isolated from 24 hybrid cell cultures retaining 1 human chromosome each. The panel was screened with gene-specific primers (5′-GCTACCTCACTTCCTTTGAG-3′ [SEQ ID NO: 9] and 5′-CTGGCTGGTGAAGTTTTCCTC-3′ [SEQ ID NO: 10]) that generated a sequence tag site (STS). The Genebridge 4 radiation hybrid panel was also screened (Research Genetics), and the results of the PCR screening were submitted to the Whitehead/MIT Radiation Hybrid mapping email server at http://www-genome.wi.mit.edu.
  • EXAMPLE 5 Platelet Aggregation Assay
  • Blood is anticoagulated with 0.1 volume 3.2% sodium citrate. Platelet-rich plasma (PRP) is prepared with an initial whole blood centrifugation (200×g, 15 min., 25° C.) and a second centrifugation of the PRP (90×g, 10 min.) to eliminate residual erythrocytes and leukocytes. The stock suspension of PRP is maintained at room temperature under 5% CO[0242] 2-air. The platelet aggregation assay uses a two-sample, four-channel Whole Blood Lumi-Aggregometor, model 560 (Chronolog Corp., Havertown, Pa.). PRP containing 1.22×108 platelets is preincubated with the sample to be tested for inhibition of aggregation for 10 min. at 37° C. in a siliconized glass cuvette containing a stirring bar, followed by stimulation with either ADP (5 mm), collagen (5 mg/ml), or thrombin (0.1 unit/ml). Platelet aggregation is recorded for at least 10 min. Data are expressed as the percentage of light transmission with platelet-poor plasma equal to 100%.
  • EXAMPLE 6 CD39-L4 is a Soluble Apyrase
  • The mammalian ectoapyrase CD39 is an integral membrane protein with two transmembrane domains (one at each end of the protein) (Maliszewski, C. R. et al., J. Immunol. 153:3574-3583). The hydrophobicity profiles for the deduced amino acid sequence of other family members, such as CD39L1 and CD39L3, are very similar to CD39 (Chadwick, B. P. and Frischauf A. M., Genomics 50:357-367), suggesting that these proteins also have two membrane spanning domains. However, CD39-L4 does not appear to have a second transmembrane domain at its C-terminus, suggesting that the N-terminus hydrophobic region could code for a secretory signal. To test this hypothesis, CD39-L4 was subcloned into the mammalian expression vector pCDNA3.1 and a 6-Histidine tag was inserted into the coding sequence. [0243]
  • The CD39-L4 cDNA sequence was initially isolated from a macrophage cDNA library (Invitrogen). The sense primer (5′-TTAAAGCTTGGGAAAAGAATGGCCACTTC-3′, SEQ ID NO. 20) with a HindIII site and the antisense primer (5′-AGACTCGAGGTGGCTCAATGGGAGATGCC-3′, SEQ ID NO. 21) with a XhoI site were used to subclone the coding sequences into the mammalian expression vector pcDNA3.1 (Invitrogen). The nucleotide sequence of the insert is set forth in SEQ ID NO. 4. In order to immunologically detect the protein, the coding region was further modified so that it would include a Gly-Ser6His epitope tag immediately following Arg[0244] 24. Briefly, two partially overlapping complementary oligonucleotides (5′-GCGCTGTCTCCCACAGAGGATCGCATCACCATCACCATCACAACCAGCA GACTTGGTT-3′ (SEQ ID. NO. 22) and 5′-AACCAAGTCTGCTGGTTGTGATGGTGATGGTGATGCGATCCTCTGTGGG AGACAGCGC-3′ (SEQ ID NO. 23)) were used on the CD39-L4 pcDNA3.1 template. The primers were extended in opposite directions around the plasmid using a 12 cycle PCR program (95° C., 1 minute; 60° C., 1 minute; 72° C., 15 minutes) (Stratagene). The reaction was treated with DpnI to digest the methylated parental DNA and then transformed into E. coli. Colonies were screened for the insert.
  • To ascertain whether CD39-L4-6His is secreted, the coding region of the CD39-L4-6His protein was inserted into the pcDNA3.1 expression vector and transiently transfected into COS-7 cells. Cos-7 cells obtained from the American Tissue Type Culture Collection were grown in DMEM supplemented with 10% FBS and 100 units/ml penicillin G and 100 μg/ml streptomycin sulfate at 37° C. in 10% CO[0245] 2. Transfections were performed at 75% confluency in 10 cm plates with Fugene-6 according to the manufacturers instructions. The cells in 7 mls of medium were incubated with 16 μl of Fugene-6 and 8 μg of DNA for 14-18 hours. At the end of the transfection the medium was replaced with DMEM medium containing low serum (1% FBS). The cells were then incubated for 24-48 hours prior to harvesting.
  • The CD39-L4-6His was concentrated by treating the cell lysates and medium with Nickel-NTA agarose (Qiagen) followed by SDS/PAGE and immunoblot analysis with an antibody against the Arg-Gly-Ser-6His epitope. Cells were washed twice with PBS containing 0.5 μg/ml leupeptin, 0.7 μg/ml pepstatin and 0.2 μg/ml aprotinin. After a brief sonication and centrifugation step to clear the lysate, the samples were then incubated with a Nickel-NTA resin at 4° C. for 2-3 hours. The histidine-tagged protein complexed to the resin was washed three times with PBS before loading onto a 10% SDS/PAGE gel for Western blot analysis. CD39-L4 was detected in both the cell lysate and the medium from cells transfected with the CD39-L4-6His expression vector, but not from control cells. While the predicted molecular weight of CD39-L4-6His is 46 kDa, the immunoreactive protein exhibited a mobility by SDS/PAGE corresponding to a molecular mass of approximately 51 kDa in the media and approximately 48 kDa in the cell lysate. The difference in apparent molecular weight may be due to posttranslational medications of three potential N-glycosylation sites in the CD39-L4 predicted amino acid sequence. [0246]
  • Secretion of CD39-L4 was also examined by treatment of the transfected cells with brefeldin A, an inhibitor of translocation of secretory proteins from the endoplasmic reticulum to the Golgi apparatus. Chadwick, et al., Genomics 50:357-367 (1998). Brefeldin A was dissolved in ethanol and added to the transfected cells 48 hours after transfection. Both control and brefeldin A treated cells were washed once with PBS and incubated for 8 hours in medium with none or varying dosages of brefeldin A. Increasing dosages of brefeldin A blocked secretion of CD39-L4-6His and led to massive intracellular accumulation. [0247]
  • EXAMPLE 7 Assay For ATPase Activity
  • Apyrase activity was determined by measuring the amount of [[0248] 33P]Pi released from [γ33P]ATP. In summary, 50 μl of samples were incubated in the presence of 10 μCi of [γ33P]ATP for one hour at 37° C. The [33P]Pi released and the [γ33P]ATP were separated by thin layer chromatography (TLC) plates (EM Science). The solvent system consisted of 1 M KH2PO4. The separated compounds were scanned for radioactivity with a Phosphoimager (Molecular Dynamics, Sunnyvale, Calif.) and quantitated by ImageQuant software. COS-7 cells transfected with SEQ ID NOs. 3 and 25 had at least a four fold increase in activity over cells transfected with the vector alone. Although ATPase activity was present, Example 13 demonstrates that CD39-L4 has significantly more NDPase activity.
  • EXAMPLE 8 Site-directed Mutagenesis of CD39L4
  • Site directed mutagenesis was employed to increase the enzymatic activity of CD39L4. Amino acid sequence comparisons between CD39 family members reveal four highly homologous regions in all five human members (Chadwick and Frischauf, Genomics 50:357-367, 1998). These regions, termed apyrase-conserved regions (ACRs), are present not only in the CD39 family members but other apyrases from species as distant as yeast and plants. Examination of similarities and differences in the CD39 ACRs led to the design of three CD39L4 mutants (see FIG. 5). In these mutants, codons encoding CD39 ACR specific residues were used to replace codons from the CD39L4 wild type ACR sequence. Only residues with significantly different structural or chemical properties were replaced. A PCR based approach was used to produce these mutations. [0249]
  • Briefly, the expression vector pCDNA3.1 (Invitrogen) containing the full coding sequence of the CD39L4 gene (with a 6 Histidine tag inserted after Arg 24 in the coding sequence to allow purification of the secreted mature form of the protein) was subjected to a PCR-based site-directed mutagenesis approach using overlapping oligonucleotides [CD39-L4 ACR I mutant (nt 177-148 and 160-204): 5′-GTG AGT GCT CCC TGC ATC TAA CAT AAT TCC-3′ (SEQ ID NO: 12) and 5′-GAT GCA GGG AGC ACT CAC ACT AGT ATT CAT GTT TAC ACC TTT GTG-3′ (SEQ ID NO: 13); CD39-L4 ACR II mutant (nt 402-359 and 385-415): 5′-GCG TAG TCC TGC TGT TGC CCC TAG GTA CAC TGG GGT CTT TTA CC-3′ (SEQ ID NO: 14) and 5′-GCA ACA GCA GGA CTA CGC TTA CTG CCA GAA C-3′ (SEQ ID NO: 15); and CD39-L4 ACR III mutant (nt 532-485 and 513-540): 5′-CCC AAG CGA ATA TGC CTT CGT CTT GTC CAG TCA TGA TGC TAA CAC TGC-3′ (SEQ ID NO: 16) and 5′-CGA AGG CAT ATT CGC TTG GGT TAC TGT G-3′ (SEQ ID NO:17)]. After amplification of the whole plasmid with Pfu DNA polymerase (Stratagene) (95° C./1 min; 60° C./1 min; 72° C./15 min for 12 cycles), the methylated parental DNA was digested with the restriction enzyme DpnI, leaving only the unmethylated PCR amplified products. The resulting annealed double-stranded nicked products were then transformed into bacteria and the resulting colonies were screened for the desired mutations by sequencing. The subsequent constructs were fully sequenced to verify that the mutations were in fact introduced and that no extraneous mutations were generated. [0250]
  • EXAMPLE 9 ACR III Mutant Increases ADPase Activity
  • Plasmids containing the mutated and wild type forms of the CD39L4 gene were transfected into COS-7 cells. After two days, protein was purified from the culture medium using a Nickel-NTA resin approach to concentrate the tagged proteins. These proteins were then assayed for ATPase and ADPase activity by measuring the inorganic phosphate released (Wang, T. F., et al., J. Biol. Chem. 273:24814-24821, 1998). The proteins were incubated in apyrase buffer (15 mM Tris pH 7.4, 135 mM NaCl, 2 mM EGTA and 10 mM glucose) for 1 hour at 37° C. with or without 2 mM CaCl[0251] 2 or 2 mM MgCl2. Phosphatase reactions were initiated by the addition of ADP or ATP to a final concentration of 1 mM. The reaction of inorganic phosphorus with ammonium molybdate in the presence of sulfuric acid, produces an unreduced phosphomolybdate complex. The absorbance of this complex at 340 nm is directly proportional to the inorganic phosphorus concentration (Daly, J. A., and Ertingshausen G., Clin. Chem. 18:263 (1972) (Sigma Diagnostics)).
  • As seen in FIG. 7, mutations in ACR I and II eliminate activity, whereas the mutations in ACR III increase activity six-fold over wild type. This increased activity therefore offers a greater therapeutic potential, as less protein could be administered to offer the same pharmacological effect. The replacement of three amino acids in the III region (amino acids 167 to 181 in CD39-L4) and the resulting increase in ADPase activity predicts that replacement of additional amino acids within this region by amino acids from the equivalent region of CD39 may also enhance the activity of the protein over wild type CD39L4. The increase in ADPase activity over wild type may also be due to the replacement of only one or two of the three amino acids; this can be confirmed by replacing one or two amino acids at a time. [0252]
  • The polynucleotide and amino acid sequences of a CD39-L4 variant termed ACRIII and having the amino acid substitutions D168-T, S170-Q and L175-F compared to wild type CD39-L4 (SEQ ID NO: 5) are set forth in SEQ ID NOs: 6 and 7, respectively, and in FIG. 6. [0253]
  • EXAMPLE 10 ACR III Mutant and Wild Type Forms Are Specific For ADP and Not ATP
  • Both the CD39L4 wild type and the CD39L4 variant with mutations in the ACRIII region hydrolyze ADP. However, when ATP was tested as a substrate, neither the CD39L4 nor the CD39L4 mutant, ACR III, catalyzed hydrolysis. In contrast, CD39 as a membrane bound molecule (Marcus, et al., The Journal of Clinical Investigation, 99: 1351-1360) or as a genetically engineered soluble form (Gayle, et al., The Journal of Clinical Investigation, 101:1851-1858, 1998) is able to hydrolyze both ATP and ADP substrates efficiently. The specificity that both CD39L4 wild type and the CD39L4 ACR III mutant have for ADP is an advantageous feature that makes these CD39L4-type molecules better antiplatelet therapeutic candidates than CD39, as ADP is the agonist that causes platelet aggregation. Therapeutics that have both ADPase and ATPase activities potentially could create adverse side effects by interfering with levels of ATP in the circulation. [0254]
  • EXAMPLE 11 Organization of the Human CD39-L4 Gene
  • A human CITB BAC genomic library (Research Genetics) was screened with gene specific primers [246-16 (nt 5522-5543), 5′-CTTCCTTCACTGGGAATTCAGG-3′ (SEQ ID NO: 18) and 246-K4 (nt 4922-4945), 5′-CTGTTTACCGAGATGGTTGGAAGC-3′ (SEQ ID NO: 19)] using a PCR based assay. [0255]
  • Briefly, gene specific primers were used to screen pools of BAC DNAs. BAC pools that produced an amplified DNA fragment of the predicted size were pursued until an individual BAC was identified. BAC63-I18 was isolated and sequenced with gene specific primers for the CD39-L4 cDNA, as well as intron specific primers. The CD39-L4 coding sequence was found to be distributed over 10 exons spanning 9.3 kb of genomic DNA as set out in SEQ ID NO: 8. [0256]
  • EXAMPLE 12 CD39-L4 and CD39-L2 Are Stimulated By Divalent Cations
  • The high degree of conservation in the apyrase conserved regions of CD39-L4 and CD39-L2 suggests similar function to other apyrases. To test this hypothesis, COS-7 cells were transfected with the CD39-L4-6His and CD39-L2myc-His construct as described herein. The medium from transfected cells was incubated with Nickel-NTA resin (Qiagen) in order to capture the 6His tagged protein, the resin was washed with assay buffer (buffer A, 15 mM Tris pH 7.5, 134 mM NaCl and 5 mM glucose) and the protein still tethered to the resin in a suspension was assayed for ADPase activity. Nucleotidase activity was determined by measuring the amount of inorganic phosphate released from nucleotide substrates using the technique of Dlay and Ertingshausen, Clin. Chem. 18:263-265 (1972). In this reaction the complex of inorganic phosphorus with phosphor reagent (ammonium molybdate in the presence of sulfuric acid) produces an unreduced phosphomolybdate compound. The absorbance of this complex at 340 nm is directly proportional to the inorganic phosphorus concentration. The protein still tethered to the resin as a 30% (50% for CD39-L2) suspension in buffer A was assayed by the addition of the nucleotide to a final concentration of 1 mM and incubated at 37° C. for 30 minutes. The reaction was stopped by adding 100 volumes of phosphor reagent. The amount of phosphate released from the reaction was quantified using a calcium/phosphorus combined standard (Sigma). The amount of protein used in the assays was estimated by comparing the intensity of the bands in Western blots with that of a series of standards of known quantity. CD39-L4 protein from transfected cells displayed a 2.3 fold increase in activity over the cells transfected with the vector alone. When Ca[0257] 2+ and Mg2+ were added, the activity increased 3.6 fold and 6 fold, respectively. CD39-L2 protein from transfected cells displayed an 8.7 fold increase in activity over the cells transfected with the vector alone. When Ca2+ and Mg2+ were added, the activity of the CD39-L2 cells increased another 2-3 fold.
  • EXAMPLE 13 Characterization of CD39-L4 Activity
  • CD39-L4 protein was assayed for ADPase activity in the presence of different kinds of inhibitors of ADPases. Control ecto-apyrase activity was determined with protein tethered to the Nickel-NTA resin. Both assays were performed as described above except the protein was in buffer A containing 2 mM CaCl[0258] 2 and 2 mM MgCl2. As shown by Table 1 below, inhibitors of phosphatases (F) and adenylate kinase (Ap5A) did not inhibit activity. The inhibitors of vacuolar ATPases (NEM), mitochondrial ATPases (N3) and Na+, K+, ATPase (ouabain) did not significantly inhibit the Ca2+ and Mg2+ stimulated activity. However, metal chelators (EDTA and EGTA) significantly inhibited activity. These results show that the overwhelming majority of the activity in the assays originates from a protein bound to the resin with characteristics of an E-type apyrase.
    TABLE 1
    Inhibition of CD39-L4 activity
    INHIBITORS % OF CONTROL
    Control
    100 ± 7
    Ouabain (1 mM)  96 ± 6
    NEM (10 mM) 106 ± 5
    N3 (1 mM)  100 ± 12
    F (10 mM) 113 ± 5
    Ap5A (10 μM) 121 ± 9
    EGTA (2 mM)  35 ± 3
    EDTA (2 mM)  52 ± 3
  • As shown in Table 2 below, the nucleotide specificity of CD39-L4 was also assayed as described above. The CD39-L4 activity was determined with protein tethered to the Ni-NTA resin. The protein was in buffer A containing 1 mM EGTA, as well as 2 mM CaCl[0259] 2 and MgCl2. The assay was started by adding the nucleotides to a final concentration of 1 mM. The values below are expressed relative to ADP. The relative activity of the nucleotide triphosphates varies almost seven-fold with ATP being the poorest substrate. No phosphate release was detected with AMP and ADP was hydrolyzed at a rate approximately twenty-fold higher than ATP. The other nucleotide diphosphates (GDP and UDP) were also very efficiently hydrolyzed by CD39-L4. These results indicate that CD39-L4 defines a new class of E-type apyrase in humans with a specificity for NDPs as enzymatic substrates.
    TABLE 2
    Substrate specificity of CD39-L4
    NUCLEOTIDE % OF CONTROL
    ADP
    100 ± 15 
    ATP 5 ± 1
    AMP 0
    CTP 26 ± 2 
    GTP 34 ± 1 
    UTP 12 ± 4 
    CDP 268 ± 11 
    GDP 334 ± 38 
    UDP 408 ± 14 
  • EXAMPLE 14 Glycosylation Is Not Essential For CD39-L4 Activity
  • Posttranslational modifications such as N-linked glycosylation are common in secreted and membrane-bound mammalian proteins. These modifications may be important for correct protein folding or enzymatic activity and are not easily reproduced when the proteins are expressed in other organisms such as bacteria. In order to test whether CD39-L4 is glycosylated, COS-7 cells, transfected as described in Example 6, were treated with tunicamycin (Sigma), which blocks the formation of N-glycosidic linkages. [0260]
  • COS-7 cells were grown to 75% confluency and transfected with the CD39-L4-6His construct. After 24 hours, a fraction of the COS-7 cells were treated with Tunicamycin at a concentration of 5 μg/ml. The media was replaced again after 24 hours with fresh tunicamycin and harvested after 48 hours. The CD39-L4-6His protein was concentrated by treating the media with Nickel-NTA agarose (Qiagen). The resin was washed with assay buffer and the protein still tethered to the resin in a suspension was assayed for a shift in electrophoretic mobility as well as its ADPase activity. [0261]
  • Western blot analysis using an antibody against the 6-His epitope revealed that the glycosylated CD39-L4 protein isolated from the control cells had an approximate size of 51 kDa. However, tunicamycin treated cells had a molecular weight of approximately 46 kDa indicating that the protein was deglycosylated. [0262]
  • ADPase activity of the tunicamycin treated cells was assayed as described in Example 12 above. The deglycosylated CD39-L4 protein had ADPase activity comparable to an equal amount of the glycosylated protein isolated from control cells. This demonstrates that glycosylation of the protein is not important for ADPase activity. [0263]
  • EXAMPLE 15 Cloning and Expression of CD39-L2
  • The CD39-L2 coding sequence (SEQ ID NO: 26) was subcloned into pcDNA3.1/myc-His(+)A (Invitrogen) via the EcoRI and XbaI sites. Briefly, a human adult heart cDNA library (Gibco BRL) was subjected to polymerase chain reaction (PCR) using gene-specific primers L2-5′B (5′-CGTATCCCGCGGGTGGAGGCCGGGGTG-3′, SEQ ID NO: 28) and L2-3′B (5′-CTTCTGCAAGTCCCAGAGCCAGTGTGC-3′, SEQ ID NO: 29). The resulting products were diluted 100-fold and subjected to a second round of PCR with primers L2-5′A (5′-GGAGCCCAAAAGACCGGCTGC-3′, SEQ ID NO: 30) and L2-3′A (5′-TGAAGTCACGTCCAGGACAGG-3′, SEQ ID NO: 31). The product represented a single band by agarose gel and was purified and sequenced to confirm its identity. Primers corresponding to the translational start region and the carboxy terminal region, excluding the stop codon, of the CD39-L2 coding sequence, L2EcoMet (5′-CGGAATTCAACATGAAAAAAGGTAATCCGTTATGAA-3′, SEQ ID NO: 32) and L2Xba3′ (5′-TGTCTAGATGAGGCTGGACTCTTCTG-3′, SEQ ID NO: 33) were used on the purified DNA to produce a DNA fragment corresponding to the entire coding region of the CD39-L2 gene, flanked by EcoRI and XbaI sites. This PCR product was digested to generate overhang ends that were ligated into the EcoRI and XbaI sites of pcDNA3.1/myc-His(+)A. The resulting plasmid allowed expression of the CD39-L2 coding sequence fused in frame with the myc-6His epitope at the carboxy terminus. [0264]
  • Transfection of COS-7 cells was performed as described below. COS-7 cells obtained from the American Tissue Type Culture Collection were grown in MDEM supplemented with 10% FBS and 100 units/ml penicillin G and 100 μg/ml streptomycin sulfact at 37° C. in 10% CO[0265] 2. Transfections were performed at 75% confluency in 10 cm plates with Fugene-6 according to the manufacturer's instructions. The cells in 10 ml of medium were incubated with 16 μl of Fugene-6 and 8 μg of DNA for 48 hours. The medium was then replaced by DMEM containing low serum (1% FBS), and incubated for 48 hours before harvesting.
  • EXAMPLE 16 Cellular Localization of CD39-L2
  • Western blot analysis was performed on COS cells transfected with the plasmid described in Example 15 above to determine the cellular localization of CD39-L2. To detect myc epitope tagged recombinant proteins, the blot was incubated with a 2000-fold dilution of the anti-c-myc monoclonal antibody (Invitrogen) at room termperature for two hours. The secondary antibody (anti-mouse Ig AP conjugate) was diluted 1000-fold and incubated for 1-2 hours at room temperature. Bound antibody was detected by using Sigma Fast™ 5-bromo4-chloro-3indolyl phosphate/nitro blue tetrazolium (BCIP/NTP) as the alkaline phosphatase substrate according to instructions of the manufacturer. [0266]
  • The recombinant protein was detected in the media and the membrane fractions of the CD39-L2 transfected cells, but not in the cytosolic fraction or control transfections. The relative band intensities suggest that the majority of the recombinant CD39-L2 protein is secreted into the media and a fraction resides in the membrane. The predicted molecular weight of unprocessed CD39-L2 is 53 kD. However, the membrane and secreted fractions displayed slower mobility by SDS/PAGE than that predicted by its amino acid content, suggesting post translational modification. [0267]
  • To confirm that recombinant CD39-L2 is secreted, the cellular localization was performed using increasing amounts of brefeldin A, an inhibitor of translocation of secretory proteins from the endoplasmic reticulum to the Golgi apparatus. Recombinant CD39-L2 in the media decreased in a brefeldin A dose dependent manner. Recombinant CD39-L2 also accumulated in the cytosol in a dose dependent manner. Therefore, recombinant CD39-L2 secretion follows the conventional cellular secretory pathway. [0268]
  • Flow cytometric analysis was used to determine if recombinant CD39-L2 is expressed on cell surfaces. COS-7 cells were transfected as described above with either pcDNA3.1/myc-His(+)A or pCD39-L2myc-HIS. After 72 hours of transfection, the cells were washed twice with PBS, and dislodged with 10 mM EDTA in PBS. Cells were pelleted by centrifugation at 300 g for five minutes, washed with PBS and resuspended in binding buffer (PBS containing 3% FBS and 0.02% sodium azide) at a concentration of 1×10[0269] 6 cells per 100 μl. The cells were first stained with 20 μg/ml of monoclonal anti-myc antibody for 30 minutes at 4° C. The cells were then washed with binding buffer and stained with 20 μg/ml of R-phycoerythrin conjugated goat anti-mouse IgG antibody (Molecular Probes, Eugene, Oreg.). After washing with binding buffer, the cells were resuspended in 1 ml of binding buffer and analyzed on the FACScalibur flow cytometer (Becton Dickinson Immunocytometry Systems, San Jose, Calif.).
  • Expression of cell surface recombinant CD39-L2 was found only on cells transfected with pCD39-L2myc-His, while cells from the control transfection showed no antibody binding. These results confirm that CD39-L2 is a secreted apyrase. [0270]
  • EXAMPLE 17 Characterization of CD39-L2 Activity
  • CD39-L2 protein was assayed for ADPase activity in the presence of different kinds of inhibitors of ADPases. Control ecto-apyrase activity was determined with protein tethered to the Nickel-NTA resin. Both assays were performed as described in Example 12 above except the protein was in buffer A containing 1 mM EGTA and 3 mM CaCl[0271] 2. The assay was started by adding ADP to 1 mM followed by a 30 minute incubation at 37° C. As shown by Table 3 below, CD39-L2 is not inhibited by inhibitors of vacuolar adenosine triphospatase (ATPases) (NEM), mitochondrial ATPase (N3 ) and Na+, K+ ATPase (oubain). An inhibitor of adenylate kinase (Ap5A) did not inhibit activity, while an inhibitor of phosphatases (F) partially inhibited activity. Metal chelators (EDTA and EGTA) inhibited CD39-L2 activity thereby demonstrating that CD39-L2 activity is dependent on divalent cations.
    TABLE 3
    Inhibition of CD39-L2 activity
    INHIBITORS % OF CONTROL
    Control
    100 ± 3 
    Ouabain (1 mM) 101 ± 9 
    NEM (10 mM) 88.4 ± 13  
    N3 (1 mM) 90 ± 13
    F (10 mM) 63 ± 9 
    Ap5A (10 μM) 87 ± 11
    EGTA (2 mM) 34 ± 10
    EDTA (2 mM) 18.4 ± 9  
  • As shown in Table 4 below, the nucleotide specificity of CD39-L2 was also assayed as described in Example 12. The CD39-L2 activity was determined with protein tethered to the Ni-NTA resin. The protein was in assay buffer A containing 1 mM EGTA, 3 mM CaCl[0272] 2 and 3 mM MgCl2. The assay was started by adding the nucleotides to a final concentration of 1 mM. The values are expressed relative to ADP. The samples were assayed at 37° C. for 30 minutes.
    TABLE 4
    Substrate Specificity of CD39-L2
    NUCLEOTIDE % OF CONTROL
    ADP
    100 ± 8 
    ATP 16 ± 2
    AMP 0.6 ± 1 
    CTP 44 ± 4
    GTP 39 ± 1
    UTP 13 ± 1
    CDP 282 ± 18
    GDP 338 ± 52
    UDP 303 ± 5 
  • These results confirm that CD39-L2 along with CD39-L4 define a new class of E-type apyrase in humans with a specificity for NDPs as enzymatic substrates. [0273]
  • EXAMPLE 18 CD39-L4 and CD39-L2 Expression Using In Situ Hybridization
  • A. In situ hybridization of CD39-L4 in kidney [0274]
  • A 298 nt fragment of the [0275] CD39L4 cDNA 3′-untranslated region was amplified by PCR with oligonucleotide primers 246D13 and 246D4 (5′-ATCCTGGACTTGAGCCTAGAG-3′, SEQ ID NO: 34 and 5′-CTGATATTGATGGGTCTTGGG-3′, SEQ ID NO: 35). The fragment was subcloned into the pCR™ II-TOPO plasmid (Invitrogen) and sense and antisense RNA were synthesized. The probe was labeled using the digoxigenin labeling kit supplied by Boehringer-Mannheim as described in the manufacturers protocol. Automated in situ hybridization was performed by QualTek Molecular Labs (Santa Barbara, Calif.) using a modified version of a previously published procedure (Myers, J. A., et al., (1995) J. Surg. Path. 1, 191-203). The Ventana Medical Systems, Inc. (Tucson, Ariz.) TechMate™ Automated Staining System was used for this procedure. All tissues were fixed in 10% neutral buffered formalin, paraffin-embedded and cut into 4 μm thick sections. Sections were placed onto Ventana's ChemMate™ Capillary Gap Slides (POP075).
  • Staining of kidney sections revealed that specific cell types hybridized with the antisense probe but not the sense probe in a highly specific manner. The staining of the glomerulus revealed that the epithelia of the Bowman's capsule, podocyte epithelia and mesangial cells were specifically stained. The expression of the CD39L4 protein in this region could be necessary to prevent platelet aggregation in the Bowman's capsule because platelets become highly concentrated in this particular region as water and ions are filtered from the blood. The bloody region within the kidney showed staining of white blood cells, presumably macrophages. This staining is consistent with previous studies where a macrophage cDNA library showed expression of the CD39L4 cDNA. CD39L4 staining was also found in some tubule epithelial cells in the kidney. [0276]
  • B. In situ hybridization of CD39-L2 in heart [0277]
  • A 186 nt fragment of the CD39L2 cDNA was amplified by PCR with oligonucleotide primers L2RNA3 and L2RNA2 (5′-GGATGGAAAGGAGTTGGTCAG-3′, SEQ ID NO: 36 and 5′-GTCCACATGCTTCACTTCCTC-3′ SEQ ID NO: 37). The fragment was subcloned into the pCR™ II-TOPO plasmid (Invitrogen) and sense and antisense RNA were synthesized and labeled as described above. Automated in situ hybridization was performed as described above. [0278]
  • Staining of heart sections revealed that specific cell types hybridized with the antisense probe but not the sense probe in a highly specific manner. The cardiac muscle cells as well as capillary endothelial cells and white blood cells within a blood vessel showed specific staining. This staining is consistent with previous studies where a heart cDNA library showed robust expression of the CD39L2 cDNA. [0279]
  • This in situ hybridization data is consistent with a physiological role for CD39-L4 and CD39-L2 in regulating platelet aggregation and hemostasis. Further in situ hybridization may be carried out to confirm this activity. [0280]
  • The present invention is not to be limited in scope by the exemplified embodiments which are intended as illustrations of single aspects of the invention, and compositions and methods which are functionally equivalent are within the scope of the invention. Indeed, numerous modifications and variations in the practice of the invention are expected to occur to those skilled in the art upon consideration of the present preferred embodiments. Consequently, the only limitations which should be placed upon the scope of the invention are those which appear in the appended claims. All references cited within the body of the instant specification are hereby incorporated by reference in their entirety. [0281]
  • 0
    SEQUENCE LISTING
    <160> NUMBER OF SEQ ID NOS: 39
    <210> SEQ ID NO 1
    <211> LENGTH: 300
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 1
    ggcatattag cttgggttac tgtgaatttt ctgacaggtc agctgcatgg ccacagacag 60
    gagactgtgg ggaccttgga cctaggggga gcctccaccc aaatcacgtt cctgccccag 120
    tttgagaaaa ctctggaaca aactcctagg ggctacctca cttcctttga gatgtttaac 180
    agcacttata agctctatac acatagttac ctgggatttg gattgaaagc tgcaagacta 240
    gcaaccctgg gagccctgga gacagaaggg actgatgggc acactttccg gagtgcctgt 300
    <210> SEQ ID NO 2
    <211> LENGTH: 1799
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: CDS
    <222> LOCATION: (246)..(1529)
    <221> NAME/KEY: misc_feature
    <222> LOCATION: (1718)
    <223> OTHER INFORMATION: n = adenine or guanine or cytosine or thymine
    <400> SEQUENCE: 2
    gcgggctgcc gcgcaagggt ggcgcgcgcg cgttttcctt gttcctggtc aacaaagaaa 60
    tgtggagtgt cttggctgaa tcctcataca gacaagatca ttatggtgct gttaggttga 120
    aaaagtgata taataaagga accaaggaga aaattcagaa ggaaagaaaa aattgcctct 180
    gcaggtgtgc gagcaggatt gcttctgcaa caaaagcctc cacccagcca catcttggga 240
    aaaga atg gcc act tct tgg ggc aca gtc ttt ttc atg ctg gtg gta tcc 290
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser
    1 5 10 15
    tgt gtt tgc agc gct gtc tcc cac agg aac cag cag act tgg ttt gag 338
    Cys Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu
    20 25 30
    ggt atc ttc ctg tct tcc atg tgc ccc atc aat gtc agc gcc agc acc 386
    Gly Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr
    35 40 45
    ttg tat gga att atg ttt gat gca ggg agc act gga act cga att cat 434
    Leu Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His
    50 55 60
    gtt tac acc ttt gtg cag aaa atg cca gga cag ctt cca att cta gaa 482
    Val Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu
    65 70 75
    ggg gaa gtt ttt gat tct gtg aag cca gga ctt tct gct ttt gta gat 530
    Gly Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp
    80 85 90 95
    caa cct aag cag ggt gct gag acc gtt caa ggg ctc tta gag gtg gcc 578
    Gln Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala
    100 105 110
    aaa gac tca atc ccc cga agt cac tgg aaa aag acc cca gtg gtc cta 626
    Lys Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu
    115 120 125
    aag gca aca gca gga cta cgc tta ctg cca gaa cac aaa gcc aag gct 674
    Lys Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala
    130 135 140
    ctg ctc ttt gag gta aag gag atc ttc agg aag tca cct ttc ctg gta 722
    Leu Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val
    145 150 155
    cca aag ggc agt gtt agc atc atg gat gga tcc gac gaa ggc ata tta 770
    Pro Lys Gly Ser Val Ser Ile Met Asp Gly Ser Asp Glu Gly Ile Leu
    160 165 170 175
    gct tgg gtt act gtg aat ttt ctg aca ggt cag ctg cat ggc cac aga 818
    Ala Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg
    180 185 190
    cag gag act gtg ggg acc ttg gac cta ggg gga gcc tcc acc caa atc 866
    Gln Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile
    195 200 205
    acg ttc ctg ccc cag ttt gag aaa act ctg gaa caa act cct agg ggc 914
    Thr Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly
    210 215 220
    tac ctc act tcc ttt gag atg ttt aac agc act tat aag ctc tat aca 962
    Tyr Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr
    225 230 235
    cat agt tac ctg gga ttt gga ttg aaa gct gca aga cta gca acc ctg 1010
    His Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu
    240 245 250 255
    gga gcc ctg gag aca gaa ggg act gat ggg cac act ttc cgg agt gcc 1058
    Gly Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala
    260 265 270
    tgt tta ccg aga tgg ttg gaa gca gag tgg atc ttt ggg ggt gtg aaa 1106
    Cys Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys
    275 280 285
    tac cag tat ggt ggc aac caa gaa ggg gag gtg ggc ttt gag ccc tgc 1154
    Tyr Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys
    290 295 300
    tat gcc gaa gtg ctg agg gtg gta cga gga aaa ctt cac cag cca gag 1202
    Tyr Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu
    305 310 315
    gag gtc cag aga ggt tcc ttc tat gct ttc tct tac tat tat gac cga 1250
    Glu Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg
    320 325 330 335
    gct gtt gac aca gac atg att gat tat gaa aag ggg ggt att tta aaa 1298
    Ala Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys
    340 345 350
    gtt gaa gat ttt gaa aga aaa gcc agg gaa gtg tgt gat aac ttg gaa 1346
    Val Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu
    355 360 365
    aac ttc acc tca ggc agt cct ttc ctg tgc atg gat ctc agc tac atc 1394
    Asn Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile
    370 375 380
    aca gcc ctg tta aag gat ggc ttt ggc ttt gca gac agc aca gtc tta 1442
    Thr Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu
    385 390 395
    cag ctc aca aag aaa gtg aac aac ata gag acg ggc tgg gcc ttg ggg 1490
    Gln Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly
    400 405 410 415
    gcc acc ttt cac ctg ttg cag tct ctg ggc atc tcc cat tgaggccacg 1539
    Ala Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    tacttccttg gagacctgca tttgccaaca cctttttaag gggaggagag agcacttagt 1599
    ttctgaacta gtctggggac atcctggact tgagcctaga gattwrgtta attaascggc 1659
    cgagcttatc cttwatragg taatttactt gcmtggccgc gtttacacgt cgtgatggna 1719
    aacctgcgtc ccaactaacg cttgasamat ccccttcgca gctgcgatac caaaagccga 1779
    cgacgccttc cacagtgcca 1799
    <210> SEQ ID NO 3
    <211> LENGTH: 428
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 3
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    Lys Gly Ser Val Ser Ile Met Asp Gly Ser Asp Glu Gly Ile Leu Ala
    165 170 175
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly Ala
    405 410 415
    Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    <210> SEQ ID NO 4
    <211> LENGTH: 1287
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: CDS
    <222> LOCATION: (1)..(1284)
    <400> SEQUENCE: 4
    atg gcc act tct tgg ggc aca gtc ttt ttc atg ctg gtg gta tcc tgt 48
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    gtt tgc agc gct gtc tcc cac agg aac cag cag act tgg ttt gag ggt 96
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    atc ttc ctg tct tcc atg tgc ccc atc aat gtc agc gcc agc acc ttg 144
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    tat gga att atg ttt gat gca ggg agc act gga act cga att cat gtt 192
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    tac acc ttt gtg cag aaa atg cca gga cag ctt cca att cta gaa ggg 240
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    gaa gtt ttt gat tct gtg aag cca gga ctt tct gct ttt gta gat caa 288
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    cct aag cag ggt gct gag acc gtt caa ggg ctc tta gag gtg gcc aaa 336
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    gac tca atc ccc cga agt cac tgg aaa aag acc cca gtg gtc cta aag 384
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    gca aca gca gga cta cgc tta ctg cca gaa cac aaa gcc aag gct ctg 432
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    ctc ttt gag gta aag gag atc ttc agg aag tca cct ttc ctg gta cca 480
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    aag ggc agt gtt agc atc atg gat gga tcc gac gaa ggc ata tta gct 528
    Lys Gly Ser Val Ser Ile Met Asp Gly Ser Asp Glu Gly Ile Leu Ala
    165 170 175
    tgg gtt act gtg aat ttt ctg aca ggt cag ctg cat ggc cac aga cag 576
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    gag act gtg ggg acc ttg gac cta ggg gga gcc tcc acc caa atc acg 624
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    ttc ctg ccc cag ttt gag aaa act ctg gaa caa act cct agg ggc tac 672
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    ctc act tcc ttt gag atg ttt aac agc act tat aag ctc tat aca cat 720
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    agt tac ctg gga ttt gga ttg aaa gct gca aga cta gca acc ctg gga 768
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    gcc ctg gag aca gaa ggg act gat ggg cac act ttc cgg agt gcc tgt 816
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    tta ccg aga tgg ttg gaa gca gag tgg atc ttt ggg ggt gtg aaa tac 864
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    cag tat ggt ggc aac caa gaa ggg gag gtg ggc ttt gag ccc tgc tat 912
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    gcc gaa gtg ctg agg gtg gta cga gga aaa ctt cac cag cca gag gag 960
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    gtc cag aga ggt tcc ttc tat gct ttc tct tac tat tat gac cga gct 1008
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    gtt gac aca gac atg att gat tat gaa aag ggg ggt att tta aaa gtt 1056
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    gaa gat ttt gaa aga aaa gcc agg gaa gtg tgt gat aac ttg gaa aac 1104
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    ttc acc tca ggc agt cct ttc ctg tgc atg gat ctc agc tac atc aca 1152
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    gcc ctg tta aag gat ggc ttt ggc ttt gca gac agc aca gtc tta cag 1200
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    ctc aca aag aaa gtg aac aac ata gag acg ggc tgg gcc ttg ggg gcc 1248
    Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly Ala
    405 410 415
    acc ttt cac ctg ttg cag tct ctg ggc atc tcc cat tga 1287
    Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    <210> SEQ ID NO 5
    <211> LENGTH: 428
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 5
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    Lys Gly Ser Val Ser Ile Met Asp Gly Ser Asp Glu Gly Ile Leu Ala
    165 170 175
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly Ala
    405 410 415
    Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    <210> SEQ ID NO 6
    <211> LENGTH: 1287
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: CDS
    <222> LOCATION: (1)..(1284)
    <400> SEQUENCE: 6
    atg gcc act tct tgg ggc aca gtc ttt ttc atg ctg gtg gta tcc tgt 48
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    gtt tgc agc gct gtc tcc cac agg aac cag cag act tgg ttt gag ggt 96
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    atc ttc ctg tct tcc atg tgc ccc atc aat gtc agc gcc agc acc ttg 144
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    tat gga att atg ttt gat gca ggg agc act gga act cga att cat gtt 192
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    tac acc ttt gtg cag aaa atg cca gga cag ctt cca att cta gaa ggg 240
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    gaa gtt ttt gat tct gtg aag cca gga ctt tct gct ttt gta gat caa 288
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    cct aag cag ggt gct gag acc gtt caa ggg ctc tta gag gtg gcc aaa 336
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    gac tca atc ccc cga agt cac tgg aaa aag acc cca gtg gtc cta aag 384
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    gca aca gca gga cta cgc tta ctg cca gaa cac aaa gcc aag gct ctg 432
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    ctc ttt gag gta aag gag atc ttc agg aag tca cct ttc ctg gta cca 480
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    aag ggc agt gtt agc atc atg act gga caa gac gaa ggc ata ttc gct 528
    Lys Gly Ser Val Ser Ile Met Thr Gly Gln Asp Glu Gly Ile Phe Ala
    165 170 175
    tgg gtt act gtg aat ttt ctg aca ggt cag ctg cat ggc cac aga cag 576
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    gag act gtg ggg acc ttg gac cta ggg gga gcc tcc acc caa atc acg 624
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    ttc ctg ccc cag ttt gag aaa act ctg gaa caa act cct agg ggc tac 672
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    ctc act tcc ttt gag atg ttt aac agc act tat aag ctc tat aca cat 720
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    agt tac ctg gga ttt gga ttg aaa gct gca aga cta gca acc ctg gga 768
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    gcc ctg gag aca gaa ggg act gat ggg cac act ttc cgg agt gcc tgt 816
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    tta ccg aga tgg ttg gaa gca gag tgg atc ttt ggg ggt gtg aaa tac 864
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    cag tat ggt ggc aac caa gaa ggg gag gtg ggc ttt gag ccc tgc tat 912
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    gcc gaa gtg ctg agg gtg gta cga gga aaa ctt cac cag cca gag gag 960
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    gtc cag aga ggt tcc ttc tat gct ttc tct tac tat tat gac cga gct 1008
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    gtt gac aca gac atg att gat tat gaa aag ggg ggt att tta aaa gtt 1056
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    gaa gat ttt gaa aga aaa gcc agg gaa gtg tgt gat aac ttg gaa aac 1104
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    ttc acc tca ggc agt cct ttc ctg tgc atg gat ctc agc tac atc aca 1152
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    gcc ctg tta aag gat ggc ttt ggc ttt gca gac agc aca gtc tta cag 1200
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    ctc aca aag aaa gtg aac aac ata gag acg ggc tgg gcc ttg ggg gcc 1248
    Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly Ala
    405 410 415
    acc ttt cac ctg ttg cag tct ctg ggc atc tcc cat tga 1287
    Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    <210> SEQ ID NO 7
    <211> LENGTH: 428
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 7
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    Lys Gly Ser Val Ser Ile Met Thr Gly Gln Asp Glu Gly Ile Phe Ala
    165 170 175
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    Leu Thr Lys Lys Val Asn Asn Ile Glu Thr Gly Trp Ala Leu Gly Ala
    405 410 415
    Thr Phe His Leu Leu Gln Ser Leu Gly Ile Ser His
    420 425
    <210> SEQ ID NO 8
    <211> LENGTH: 9365
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: exon
    <222> LOCATION: (1)..(288)
    <221> NAME/KEY: exon
    <222> LOCATION: (1281)..(1580)
    <221> NAME/KEY: exon
    <222> LOCATION: (1820)..(1855)
    <221> NAME/KEY: exon
    <222> LOCATION: (2467)..(2555)
    <221> NAME/KEY: exon
    <222> LOCATION: (2863)..(2942)
    <221> NAME/KEY: exon
    <222> LOCATION: (3889)..(3950)
    <221> NAME/KEY: exon
    <222> LOCATION: (4894)..(4995)
    <221> NAME/KEY: exon
    <222> LOCATION: (5847)..(5987)
    <221> NAME/KEY: exon
    <222> LOCATION: (6966)..(7138)
    <221> NAME/KEY: exon
    <222> LOCATION: (8556)..(9365)
    <221> NAME/KEY: misc_feature
    <222> LOCATION: (3409)
    <223> OTHER INFORMATION: n = a or g or t or c
    <221> NAME/KEY: misc_feature
    <222> LOCATION: (9214)
    <223> OTHER INFORMATION: n = a or g or t or c
    <221> NAME/KEY: misc_feature
    <222> LOCATION: (9303)
    <223> OTHER INFORMATION: n = a or g or t or c
    <221> NAME/KEY: misc_feature
    <222> LOCATION: (9311)
    <223> OTHER INFORMATION: n = a or g or t or c
    <400> SEQUENCE: 8
    gcctctgcag gtgtgcgagc aggattgctt ctgcaacaaa agcctccacc cagccacatc 60
    ttgggaaaag aatggccact tcttggggca cagtcttttt catgctggtg gtatcctgtg 120
    tttgcagcgc tgtctcccac aggaaccagc agacttggtt tgagggtatc ttcctgtctt 180
    ccatgtgccc catcaatgtc agcgccagca ccttgtatgg aattatgttt gatgcaggga 240
    gcactggaac tcgaattcat gtttacacct ttgtgcagaa aatgccaggt aagtgcaact 300
    gggrccctta gtagagtctg taaatccaca ctttagcatc tcctcccaga aacaaatatg 360
    ctgagagttt attatgtgaa ttacagaatc tcacacctag tggatgtctt tcttcagaga 420
    actttggact acaattgaac atgtgggtta tttatttatt tttatttatt tgttttgttt 480
    ttatttttta actttttttt tgagacaagg tcttgctttg ttgcccggtc tgtagtgcag 540
    tggcatgatg acacatcact gcaaccttga cctcctgggc tcaagcagtc cttccacctc 600
    agccccctga gttgttgaga ctacaggctt gtgccaccat gcccagctca tttttaaatt 660
    tttttataga gacctgctca gactggcctc aaactcctag gctcaattga tcctcccacc 720
    tcagcctccc aaagtactgg gattataggt gtaagtcacc atgcttggcc agaacacatg 780
    gcttaattca atgtgaaatt agaagagagc tgggctgtct gtagtctgaa acccatgtgt 840
    tcaaaaagaa tagttataat ttgttcttcc tctttaaaca tgggatactc cagggatcca 900
    taatattcag aatatgggga gtggttttgg gagaaggatc acatgagaat ttcactgcca 960
    tccttggaca tgaggctagg aatccctgaa gattaacttt ttctgaattt gtcagtgttt 1020
    tttcctcagg tcacttatgg agcctgggga aaggtggagg agttaggtgt ccaccagaga 1080
    aatggtagca gaaatggacc ctcagaggtt gctctagtcc ttctttccag tactcctgca 1140
    agacattcct cacaactagg atcattgggg taacttcagg gaagtcatag gaaaacttac 1200
    agagacagag cccagcatct gaagcagcct aacttttggt aaccagctct ctcttctgtt 1260
    ttgttccatg racaaaatag gacagcttcc aattctagaa ggggaagttt ttgattctgt 1320
    gaagccagga ctttctgctt ttgtagatca acctaagcag ggtgctgaga ccgttcaagg 1380
    gctcttagag gtggccaaag actcaatccc ccgaagtcac tggaaaaaga ccccagtggt 1440
    cctaaaggca acagcaggac tacgcttact gccagaacac aaagccaagg ctctgctctt 1500
    tgaggtaaag gagatcttca ggaagtcacc tttcctggta ccaaagggca gtgttagcat 1560
    catggatgga tccgacgaag gtgggagagg tgttgatatg cgttccaggg ggagaggggc 1620
    aggatcagtg aaagatctaa ctaaaggaac tggggccagg aataaacaga aggaatgaga 1680
    tagcaggaaa tagaagacag ggagaaggga acatgtgctc tagacatgga atttagagag 1740
    gaaaaaaaaa aaacaaggtt ggggccagga aagagaaaaa atgctctggg atctaatcct 1800
    tgtctttctt tctttttagg catattagct tgggttactg tgaattttct gacaggtaat 1860
    acatcctcaa gtttatcttt agagcttaac tagcttttac atgcatagtc agaggagtaa 1920
    aagcctcttc tttcattctg tattgtttct tcttctttaa aaaaggaaaa gaggctgggt 1980
    gtggcagttc atgcctgtta attccagcgc tttgggaggc tgagttgggc agatcacttg 2040
    aggccaggag ttcaagacca gcctggccaa catggcgaaa ctccgtctct accaaaaata 2100
    caaaaatagc tgggcatggt ggtgtgtacc tgtagtccca gctactcagg aggctggaga 2160
    atcacttgaa cccaggaggc agaggttgca gtgagctgag agccgagatt gcgccactgc 2220
    actccaggct ggatgataga gcaagactct gtctccaaaa aggccttcca aaaaaaaaaa 2280
    aaacacctgc cttgaaggcc tctgctgcaa caagagtcct tccgagttga cattcacctg 2340
    cagccttggg gctggggagc agtggagtat atatggaata ccttcagtgt atgataagag 2400
    caagagagac aagtgttggg ctgcccagga tgtcgaggct atttagagct ggctctcatt 2460
    tgacaggtca gctgcatggc cacagacagg agactgtggg gaccttggac ctagggggag 2520
    cctccaccca aatcacgttc ctgccccagt ttgaggtgag tcatttaatg aagatctggt 2580
    tagaagtgca cttggcaggc gtatcatggt gccaagaaag aggcgcccca ttttcagcca 2640
    gcagctctac cacgcttagg cagagtcaag tcaattaata actaggtgaa tgttcccttg 2700
    ccatctcact gttcagaatc ccttcgtttc ctcaagccta gtgagattag ccccttaatc 2760
    tgtcttcatc tctgattttt tgctgggagg gacgggtggt ggtgtgaaca tcttcaggta 2820
    attacagatc ctgaatagct ttttgctttt tctgatttgc agaaaactct ggaacaaamt 2880
    cyatrgggct acctcacttc ctttgagatg tttaacagca cttataagct ctatacacat 2940
    aggtgaggac ggggacaggg aagaagaata tttmwtkttg tatgatksty ytamctktss 3000
    maagcwtkct caaatctstk aytkyatctg attmgcaaaa acaaagdctg tgccaattcc 3060
    ctaaggccta tcaactgaaa cccggwccac ttacaaagcc ggaggagcct aagaggcttc 3120
    tccattcttg gcctcaaaag cattaatata tgacttaaga gtcaaaagtt ttggstgggg 3180
    cagtggcttc atgcctgtaa tccctgcact ttgggaggcc gaggtgggtg ggtcacctga 3240
    ggtcaggcgt ttragaccag cctggcaaac atggtgaaac cccgtctyta ctaaaataca 3300
    aaaattagct ggatatgaca gcgcacacct gtaatcctag ctattcagga ggctgaggca 3360
    ggagaatcat ttgaaccctg gaggcggaga ttgcagtgag ccgagatcnc accmctgcac 3420
    ttcagccgga gcgacagagc aagactcagt ctcaaaaaaa aaaaaaaaaa gaatcaaaag 3480
    ctttctgtag ggagaggaca cttcaagaag gctcaggcaa agctccttgc cagctccttt 3540
    gagctggcct tcagaggttc agaatccagc ctggaatgtg atcccagttg gggctaggag 3600
    ctaagctaaa gagagctttt ctgggaatgg ttcctagwgt gggaccctag gaattgtcac 3660
    tgtctctggc ctttgaatga taactgtggg gaattcttac tgcatagcct tgatccaaac 3720
    tgtgcagaaa ttaccccttg ttgaccacag gagatgaata tgtcacagac agaacaaggt 3780
    tttcatcttt ccagagggac acaggaacaa tgttactttt gaaagaggta gctttaggct 3840
    agagaacttc aggaccagca tgaaattagt caatcctgta ttttacagtt acctgggatt 3900
    tggattgaaa gctgcaagac tagcaaccct gggagccctg gagacagaag gtttgtctgg 3960
    gtacctgtgc tgggggggga tggtgagggt gacacagata ctccgcttgc ttcttccctt 4020
    ccttgatagc cattctatgg aggaaaagat tatgttgaat tgggaggcaa atgttgtata 4080
    atggacctaa taatggcaaa ctccttttct agtttataag ttcagaagtt ttgatgtata 4140
    ttattagcca tttttagaat gaggtctact tgttcagggg taacagccta tgtctaggca 4200
    gctgaagtgt ctgcagaaat cccaggcttt acgaatacat tcagcaggag cttgctcaag 4260
    ccctgagctt tacattggag gcacaggaag cagagtctgt tctacatgca ggtggaacaa 4320
    cagagtaact ccattgatct cttcacaggt caggcagaac tgggttcagt cccagtgttg 4380
    tgatatgagg cragtaacct atctgtgccc ctttcctcac attaaatgag aatttgcatt 4440
    taaggcactt tgtacagtaa tctgttattg ggatgacatc tattttgcat ttcagagtat 4500
    acaaaacatc ttcaagtata tttaattgaa gcctctcagc aaccagtgag gaaggtagca 4560
    tagcatttct ttcctgtttt tataaagggg aaagttgctg takgaaggtt ykrgatctct 4620
    twragatgtg atraaagcca tggacccctc tgacaaaagc acatatgcat gaaaatttgc 4680
    ttctggtttc agggggttca ccaaccccac aaagcctatc tttgaaccct gagttaagga 4740
    ttcctgtcac aggatgttgt catggaatta atttcatagg attttaaggc ccagccccca 4800
    tggtgaytct tttccacctc actggcttct tgcttgcctt cctccctctc tctcacttac 4860
    ttacctctta ccttgtgccc tggattcttt cagggactga tgggcacact ttccggagtg 4920
    cctgtttacc gagatggttg gaagcagagt ggatctttgg gggtgtgaaa taccagtatg 4980
    gtggcaacca agaaggcaag tgatgttttt tcactggtta aagttacgtt tacaatggaa 5040
    gctctggaaa agtcccatgg gaaacttttt ccagaactca agagaagctt atcttgttgc 5100
    agggasttat tccaaagatc ttggcatgcc tccaaggact aatgtgaagt gacagtgaac 5160
    aaagcagctg tcattctgca tcagccaagt gtcatggacc cattagatac ctgcccttag 5220
    ccaagtgctg tggtgcacat ctattgtcct agctactcca aaggttgagg caagaggatc 5280
    acttgagccc atgagttcaa ggctatagtg cgcaatgcca ctgcactcca gcctgggcaa 5340
    cagggagacc ctacctctta caaattaatt aagaagcata ttctaagcct aggtctaatg 5400
    cagcagtgtg aaagcctgtt tagttaatgg ttagctattt aaattatagt aaaacttaaa 5460
    accaagacaa gaatgattca tcttcttata aaaggtatat acctgaatat caaggaatga 5520
    acctgaattc ccagtgaagg aagcaggcga gccctttagc tacttgctta caaatgctat 5580
    ggaatgtaat gctaggcagc agcacaaggt tggccatgat ctggtgaata cagattaggc 5640
    aggagagcgg ccatggagaa acagactggt gaggctgcag acgtttgctc atctttgttt 5700
    tgacgcctct tgtcccaagc ctcagccttc tcctgctttc ttgaccttcc tgctgttccc 5760
    tcattgtctc cagcagcctg cctcagagag tgtccccttc ccccagcgtc gttctcacct 5820
    tacccctgtg cacctttgcc tggcagggga ggtgggcttt gagccctgct atgccgaagt 5880
    gctgagggtg gtacgaggaa aacttcacca gccagaggag gtccagagag gttccttcta 5940
    tgctttctct tactattatg accgagctgt tgacacagac atgattggtg agttcacccc 6000
    aggtgtcagt ccagagagga aggtggatag ggctgtggtg gggaaggtca aggagaaaga 6060
    gcacttgagg tgctttgtcg gggtgattac ccacctcttt tctagtcact cgaacaaaag 6120
    ggtggaaatg acttagagtc ttttggaggt gagagatgac caaaacaact atatgaggtc 6180
    tttttttttt taacatgttt attgaggtat aattggcata caataagtgc cacatttaaa 6240
    gtatacaatt taagttttgt catgtataca cccatgaatc catccagcac attgaagata 6300
    ataaacatat ttcaccacaa aaagtttcct cctgtctctt tataactttt cttcttatca 6360
    caaaagcagt gtttttgcct aactgtgaaa gtatatgtac ctgatctgtc atggcctgag 6420
    agagatgaat taatttccta ttattgtggg ggttttgttg ttgttgttgt tttggttttt 6480
    tgtttgtttg tttgtttttt gagacagagt ctcactctgt tacccaggct ggagtgcaat 6540
    ggcatgatct aggctcactg caacctctgc ctcccgggtt caaccgattc tcctgcccca 6600
    gtctcctgag tagctgggat tacaggtgcc tgccaccaca cccggctaat ttttttttta 6660
    atagagacga ggtttcacca tgttggtcag gctggtcttg aactcctgac ctcgttatct 6720
    gccttcctcg gcctcccaaa gtgctgggat tacaggcatg agccaccaca cccggcctat 6780
    tgtgttttat gggtctgttt tttccattgt ggttaaatat acataacatg gaatagattg 6840
    taaataagta aattaggttg catagattac attatgtaca tgtgtatata atgaatgaat 6900
    gaatgaattt ccttatgctt ccttgaaggc gttttgatat cagataatct tctgttttat 6960
    ttcagattat gaaaaggggg gtattttaaa agttgaagat tttgaaagaa aagccaggga 7020
    agtgtgtgat aacttggaaa acttcacctc aggcagtcct ttcctgtgca tggatctcag 7080
    ctacatcaca gccctgttaa aggatggctt tggctttgca gacagcacag tcttacaggt 7140
    aagagacagg acaccagagt ctcataacag ccctcttttg tgggggttga gaaggagtaa 7200
    gagcttgttc agtaatcaga gtagctagaa gtgaaattat gaggtatttt tgtttgggct 7260
    atggacaagg tactgtgctg ggcaccatga atgtgggaaa ttatctcaat gcaatggtag 7320
    cctccgagtg tattaccagg caagctatcg cacaggtcac agaacagaaa gactagcagc 7380
    ccaaattaag atgccaagtc acatggttta tttatttatt tatttattta ttattatttt 7440
    tttgagacgg agtctygctc ttgttkccyr ggctggagtg cartggcryg atcwcrgctc 7500
    actgcarcct ycrcctcctg ggttcaagcg attctyctgc ctcagcctcc cragtagctg 7560
    ggattacagg crygcgccac cacgccyggc taattttttt gtatttttag tagagacggg 7620
    gtttcaccat gttggccagg ctrktctyra actyctgayc tcaggtgatc cacccrcctc 7680
    rgcctcccaa agtgctrgra ttayaggyrt gagccaccac kccyrgcctt ttttgktcgk 7740
    ttcttttttt ttchtttttt tttttttttt gagacagggt cttgctctgt cacccatgct 7800
    ggagtgcagt ggcatgatct cagttcactg caacctctgc ctcccgggtt caagtgaccc 7860
    tcccacctca gccctctgag tagctgggat tacaggtgtg tgccaccact cttgtctaat 7920
    ttttttgtag agacggggtt ttgccatgtt gcccaggctg gtcttgaact cctggcctca 7980
    agcaatccac ctgccttggc ctcccaaagt gccaggagta caggcatgag ccactgcgcc 8040
    tggccccatg tttggttatt attagtgctt aggaagaggc acttgcttac atagtaggag 8100
    ttgagaagct tggtttgttc tttcctaccc ctagatctat tctcacctcc tgaccatgct 8160
    ctttctgcca catctattat cattacaagt tgccttatct gaaattagtg aatcagaaaa 8220
    taaagcaggg gatactttgt gtagtttcaa cgttagggaa agttcagaat actgtctgtc 8280
    taaactatct ctctagaagg cctgatgggc cacaacctgg gccagaagca ttcagttcag 8340
    atatgagaat ggtgggtgta ggggcaatgg ccaatgggcc atggccggaa ggaaattgtt 8400
    acagagtagt gggaagcctg caaagactgg cttctgtccg ttttgccttg gtttgcccat 8460
    gtggatattc tttgccaata ttttctgccc aagagctgtg cttgctagag ttggaaactg 8520
    gatgaaaagg tgaagacttt ttttcttctc aacagctcac aaagaaagtg aacaacatag 8580
    agacgggctg ggccttgggg gccacctttc acctgttgca gtctctgggc atctcccatt 8640
    gaggccacgt acttccttgg agacctgcat ttgccaacac ctttttaagg ggaggagaga 8700
    gcacttagtt tctgaactag tctggggaca tcctggactt gagcctagag atttaggttt 8760
    aattaatttt acacatctaa tagtgaactg ctgcctaacc actcaagagt acacagctgg 8820
    caccagagca tcacagagag ccctgtgagc caaaaagtat agttttggaa cttaaccttg 8880
    gagtgagagc ccagggacag gtccctggaa accaaagaaa aatcgcattt caaccctttg 8940
    agtgcctcat tccactgaat atttaaattt tcctcttaaa tgggaaactg acttattgca 9000
    atcccaagac ccatcaatat cagtattttt ttcctcccta tacagggccc tgcccaccct 9060
    tatctgcacc cacctcccct gaaaaagaga gaaaaaaaaa aamccbggtt ttgctttccw 9120
    tgtwtaatyc amcgacmcaa aakgggacca tgtcaaaatc tgtwtgatcc tattytgggt 9180
    tascyccaat cagccagctg aragccttcc taanttttaw taggatgara gagtaccycc 9240
    taactgtgca taaattcagc cttaaaaaaa aaggcacccg ggctttgggg acatgtttgg 9300
    ganggggggg ntgcctcata tacccacctt tggtttaata acattttatc agcactttgg 9360
    gataa 9365
    <210> SEQ ID NO 9
    <211> LENGTH: 20
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 9
    gctacctcac ttcctttgag 20
    <210> SEQ ID NO 10
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 10
    ctggctggtg aagttttcct c 21
    <210> SEQ ID NO 11
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 11
    gcaggtctcc aaggaagtac g 21
    <210> SEQ ID NO 12
    <211> LENGTH: 30
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 12
    gtgagtgctc cctgcatcta acataattcc 30
    <210> SEQ ID NO 13
    <211> LENGTH: 45
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 13
    gatgcaggga gcactcacac tagtattcat gtttacacct ttgtg 45
    <210> SEQ ID NO 14
    <211> LENGTH: 44
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 14
    gcgtagtcct gctgttgccc ctaggtacac tggggtcttt ttcc 44
    <210> SEQ ID NO 15
    <211> LENGTH: 31
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 15
    gcaacagcag gactacgctt actgccagaa c 31
    <210> SEQ ID NO 16
    <211> LENGTH: 48
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 16
    cccaagcgaa tatgccttcg tcttgtccag tcatgatgct aacactgc 48
    <210> SEQ ID NO 17
    <211> LENGTH: 28
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 17
    cgaaggcata ttcgcttggg ttactgtg 28
    <210> SEQ ID NO 18
    <211> LENGTH: 22
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 18
    cttccttcac tgggaattca gg 22
    <210> SEQ ID NO 19
    <211> LENGTH: 24
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 19
    ctgtttaccg agatggttgg aagc 24
    <210> SEQ ID NO 20
    <211> LENGTH: 29
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 20
    ttaaagcttg ggaaaagaat ggccacttc 29
    <210> SEQ ID NO 21
    <211> LENGTH: 29
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 21
    agactcgagg tggctcaatg ggagatgcc 29
    <210> SEQ ID NO 22
    <211> LENGTH: 58
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 22
    gcgctgtctc ccacagagga tcgcatcacc atcaccatca caaccagcag acttggtt 58
    <210> SEQ ID NO 23
    <211> LENGTH: 58
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 23
    aaccaagtct gctggttgtg atggtgatgg tgatgcgatc ctctgtggga gacagcgc 58
    <210> SEQ ID NO 24
    <211> LENGTH: 1601
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 24
    gcgggctgcc gcgcaagggt ggcgcgcgcg cgttttcctt gttcctggtc aacaaagaaa 60
    tgtggagtgt cttggctgaa tcctcataca gacaagatca ttatggtgct gttaggttga 120
    aaaagtgata taataaagga accaaggaga aaattcagaa ggaaagaaaa aattgcctct 180
    gcaggtgtgc gagcaggatt gcttctgcaa caaaagcctc cacccagcca catcttggga 240
    aaagaatggc cacttcttgg ggcacagtct ttttcatgct ggtggtatcc tgtgtttgca 300
    gcgctgtctc ccacaggaac cagcagactt ggtttgaggg tatcttcctg tcttccatgt 360
    gccccatcaa tgtcagcgcc agcaccttgt atggaattat gtttgatgca gggagcactg 420
    gaactcgaat tcatgtttac acctttgtgc agaaaatgcc aggacagctt ccaattctag 480
    aaggggaagt ttttgattct gtgaagccag gactttctgc ttttgtagat caacctaagc 540
    agggtgctga gaccgttcaa gggctcttag aggtggccaa agactcaatc ccccgaagtc 600
    actggaaaaa gaccccagtg gtcctaaagg caacagcagg actacgctta ctgccagaac 660
    acaaagccaa ggctctgctc tttgaggtaa aggagatctt caggaagtca cctttcctgg 720
    taccaaaggg cagtgttagc atcatggatg gatccgacga aggcatatta gcttgggtta 780
    ctgtgaattt tctgacaggt cagctgcatg gccacagaca ggagactgtg gggaccttgg 840
    acctaggggg agcctccacc caaatcacgt tcctgcccca gtttgagaaa actctggaac 900
    aaactcctag gggctacctc acttcctttg agatgtttaa cagcacttat aagctctata 960
    cacatagtta cctgggattt ggattgaaag ctgcaagact agcaaccctg ggagccctgg 1020
    agacagaagg gactgatggg cacactttcc ggagtgcctg tttaccgaga tggttggaag 1080
    cagagtggat ctttgggggt gtgaaatacc agtatggtgg caaccaagaa ggggaggtgg 1140
    gctttgagcc ctgctatgcc gaagtgctga gggtggtacg aggaaaactt caccagccag 1200
    aggaggtcca gagaggttcc ttctatgctt tctcttacta ttatgaccga gctgttgaca 1260
    cagacatgat tgattatgaa aaggggggta ttttaaaagt tgaagatttt gaaagaaaag 1320
    ccagggaagt gtgtgataac ttggaaaact tcacctcagg cagtcctttc ctgtgcatgg 1380
    atctcagcta catcacagcc ctgttaaagg atggctttgg ctttgcagac agcacagtct 1440
    tacaggctgc cgtactgagg tgatgggcca agctggagat atccccaaag cccatgttga 1500
    caccctgtcc tgcaagcgga tggactctgt gggctgcatc cctaagaata aagcagagtt 1560
    caggtgtgac ctctggcagc aaaaaaaaaa aaaaaaaaaa a 1601
    <210> SEQ ID NO 25
    <211> LENGTH: 405
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 25
    Met Ala Thr Ser Trp Gly Thr Val Phe Phe Met Leu Val Val Ser Cys
    1 5 10 15
    Val Cys Ser Ala Val Ser His Arg Asn Gln Gln Thr Trp Phe Glu Gly
    20 25 30
    Ile Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Ser Thr Leu
    35 40 45
    Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val
    50 55 60
    Tyr Thr Phe Val Gln Lys Met Pro Gly Gln Leu Pro Ile Leu Glu Gly
    65 70 75 80
    Glu Val Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln
    85 90 95
    Pro Lys Gln Gly Ala Glu Thr Val Gln Gly Leu Leu Glu Val Ala Lys
    100 105 110
    Asp Ser Ile Pro Arg Ser His Trp Lys Lys Thr Pro Val Val Leu Lys
    115 120 125
    Ala Thr Ala Gly Leu Arg Leu Leu Pro Glu His Lys Ala Lys Ala Leu
    130 135 140
    Leu Phe Glu Val Lys Glu Ile Phe Arg Lys Ser Pro Phe Leu Val Pro
    145 150 155 160
    Lys Gly Ser Val Ser Ile Met Asp Gly Ser Asp Glu Gly Ile Leu Ala
    165 170 175
    Trp Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly His Arg Gln
    180 185 190
    Glu Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr
    195 200 205
    Phe Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr
    210 215 220
    Leu Thr Ser Phe Glu Met Phe Asn Ser Thr Tyr Lys Leu Tyr Thr His
    225 230 235 240
    Ser Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly
    245 250 255
    Ala Leu Glu Thr Glu Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys
    260 265 270
    Leu Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr
    275 280 285
    Gln Tyr Gly Gly Asn Gln Glu Gly Glu Val Gly Phe Glu Pro Cys Tyr
    290 295 300
    Ala Glu Val Leu Arg Val Val Arg Gly Lys Leu His Gln Pro Glu Glu
    305 310 315 320
    Val Gln Arg Gly Ser Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala
    325 330 335
    Val Asp Thr Asp Met Ile Asp Tyr Glu Lys Gly Gly Ile Leu Lys Val
    340 345 350
    Glu Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Glu Asn
    355 360 365
    Phe Thr Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Ser Tyr Ile Thr
    370 375 380
    Ala Leu Leu Lys Asp Gly Phe Gly Phe Ala Asp Ser Thr Val Leu Gln
    385 390 395 400
    Ala Ala Val Leu Arg
    405
    <210> SEQ ID NO 26
    <211> LENGTH: 2762
    <212> TYPE: DNA
    <213> ORGANISM: Homo sapiens
    <220> FEATURE:
    <221> NAME/KEY: CDS
    <222> LOCATION: (148)..(1599)
    <400> SEQUENCE: 26
    gtggggtcgt atcccgcggg tggaggccgg ggtggcgccg gccggggcgg gggagcccaa 60
    aagaccggct gccgcctgct ccccggaaaa gggcactcgt ctccgtgggt gtggcggagc 120
    gcgcggtgca tggaatgggc tatgtga atg aaa aaa ggt atc cgt tat gaa act 174
    Met Lys Lys Gly Ile Arg Tyr Glu Thr
    1 5
    tcc aga aaa acg agc tac att ttt cag cag ccg cag cac ggt cct tgg 222
    Ser Arg Lys Thr Ser Tyr Ile Phe Gln Gln Pro Gln His Gly Pro Trp
    10 15 20 25
    caa aca agg atg aga aaa ata tcc aac cac ggg agc ctg cgg gtg gcg 270
    Gln Thr Arg Met Arg Lys Ile Ser Asn His Gly Ser Leu Arg Val Ala
    30 35 40
    aag gtg gca tac ccc ctg ggg ctg tgt gtg ggc gtg ttc atc tat gtt 318
    Lys Val Ala Tyr Pro Leu Gly Leu Cys Val Gly Val Phe Ile Tyr Val
    45 50 55
    gcc tac atc aag tgg cac cgg gcc acc gcc acc cag gcc ttc ttc agc 366
    Ala Tyr Ile Lys Trp His Arg Ala Thr Ala Thr Gln Ala Phe Phe Ser
    60 65 70
    atc acc agg gca gcc ccg ggg gcc cgg tgg ggt cag cag gcc cac agc 414
    Ile Thr Arg Ala Ala Pro Gly Ala Arg Trp Gly Gln Gln Ala His Ser
    75 80 85
    ccc ctg ggg aca gct gca gac ggg cac gag gtc ttc tac ggg atc atg 462
    Pro Leu Gly Thr Ala Ala Asp Gly His Glu Val Phe Tyr Gly Ile Met
    90 95 100 105
    ttt gat gca gga agc act ggc acc cga gta cac gtc ttc cag ttc acc 510
    Phe Asp Ala Gly Ser Thr Gly Thr Arg Val His Val Phe Gln Phe Thr
    110 115 120
    cgg ccc ccc aga gaa act ccc acg tta acc cac gaa acc ttc aaa gca 558
    Arg Pro Pro Arg Glu Thr Pro Thr Leu Thr His Glu Thr Phe Lys Ala
    125 130 135
    gtg aag cca ggt ctt tct gcc tat gct gat gat gtt gaa aag agc gct 606
    Val Lys Pro Gly Leu Ser Ala Tyr Ala Asp Asp Val Glu Lys Ser Ala
    140 145 150
    cag gga atc cgg gaa cta ctg gat gtt gct aaa cag gac att ccg ttc 654
    Gln Gly Ile Arg Glu Leu Leu Asp Val Ala Lys Gln Asp Ile Pro Phe
    155 160 165
    gac ttc tgg aag gcc acc cct ctg gtc ctc aag gcc aca gct ggc tta 702
    Asp Phe Trp Lys Ala Thr Pro Leu Val Leu Lys Ala Thr Ala Gly Leu
    170 175 180 185
    cgc ctg tta cct gga gaa aag gcc cag aag tta ctg cag aag gtg aaa 750
    Arg Leu Leu Pro Gly Glu Lys Ala Gln Lys Leu Leu Gln Lys Val Lys
    190 195 200
    gaa gta ttt aaa gca tcg cct ttc ctt gta ggg gat gac tgt gtt tcc 798
    Glu Val Phe Lys Ala Ser Pro Phe Leu Val Gly Asp Asp Cys Val Ser
    205 210 215
    atc atg aac gga aca gat gaa ggc gtt tcg gcg tgg atc acc atc aac 846
    Ile Met Asn Gly Thr Asp Glu Gly Val Ser Ala Trp Ile Thr Ile Asn
    220 225 230
    ttc ctg aca ggc agc ttg aaa act cca gga ggg agc agc gtg ggc atg 894
    Phe Leu Thr Gly Ser Leu Lys Thr Pro Gly Gly Ser Ser Val Gly Met
    235 240 245
    ctg gac ttg ggc gga gga tcc act cag atc gcc ttc ctg cca cgc gtg 942
    Leu Asp Leu Gly Gly Gly Ser Thr Gln Ile Ala Phe Leu Pro Arg Val
    250 255 260 265
    gag ggc acc ctg cag gcc tcc cca ccc ggc tac ctg acg gca ctg cgg 990
    Glu Gly Thr Leu Gln Ala Ser Pro Pro Gly Tyr Leu Thr Ala Leu Arg
    270 275 280
    atg ttt aac agg acc tac aag ctc tat tcc tac agc tac ctc ggg ctc 1038
    Met Phe Asn Arg Thr Tyr Lys Leu Tyr Ser Tyr Ser Tyr Leu Gly Leu
    285 290 295
    ggg ctg atg tcg gca cgc ctg gcg atc ctg ggc ggc gtg gag ggg cag 1086
    Gly Leu Met Ser Ala Arg Leu Ala Ile Leu Gly Gly Val Glu Gly Gln
    300 305 310
    cct gct aag gat gga aag gag ttg gtc agc cct tgc ttg tct ccc agt 1134
    Pro Ala Lys Asp Gly Lys Glu Leu Val Ser Pro Cys Leu Ser Pro Ser
    315 320 325
    ttc aaa gga gag tgg gaa cac gca gaa gtc acg tac agg gtt tca ggg 1182
    Phe Lys Gly Glu Trp Glu His Ala Glu Val Thr Tyr Arg Val Ser Gly
    330 335 340 345
    cag aaa gca gcg gca agc ctg cac gag ctg tgt gct gcc aga gtg tca 1230
    Gln Lys Ala Ala Ala Ser Leu His Glu Leu Cys Ala Ala Arg Val Ser
    350 355 360
    gag gtc ctt caa aac aga gtg cac agg acg gag gaa gtg aag cat gtg 1278
    Glu Val Leu Gln Asn Arg Val His Arg Thr Glu Glu Val Lys His Val
    365 370 375
    gac ttc tat gct ttc tcc tac tat tac gac ctt gca gct ggt gtg ggc 1326
    Asp Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Leu Ala Ala Gly Val Gly
    380 385 390
    ctc ata gat gcg gag aag gga ggc agc ctg gtg gtg ggg gac ttc gag 1374
    Leu Ile Asp Ala Glu Lys Gly Gly Ser Leu Val Val Gly Asp Phe Glu
    395 400 405
    atc gca gcc aag tac gtg tgt cgg acc ctg gag aca cag ccg cag agc 1422
    Ile Ala Ala Lys Tyr Val Cys Arg Thr Leu Glu Thr Gln Pro Gln Ser
    410 415 420 425
    agc ccc ttc tca tgc atg gac ctc acc tac gtc agc ctg cta ctc cag 1470
    Ser Pro Phe Ser Cys Met Asp Leu Thr Tyr Val Ser Leu Leu Leu Gln
    430 435 440
    gag ttc ggc ttt ccc agg agc aaa gtg ctg aag ctc act cgg aaa att 1518
    Glu Phe Gly Phe Pro Arg Ser Lys Val Leu Lys Leu Thr Arg Lys Ile
    445 450 455
    gac aat gtt gag acc agc tgg gct ctg ggg gcc att ttt cat tac atc 1566
    Asp Asn Val Glu Thr Ser Trp Ala Leu Gly Ala Ile Phe His Tyr Ile
    460 465 470
    gac tcc ctg aac aga cag aag agt cca gcc tca tagtggccga gccatccctg 1619
    Asp Ser Leu Asn Arg Gln Lys Ser Pro Ala Ser
    475 480
    tccccgtcag cagtgtctgt gtgtctgcat aaaccctcct gtcctggacg tgacttcatc 1679
    ctgaggagcc acagcacagg ccgtgctggc actttctgca cactggctct gggacttgca 1739
    gaaggcctgg tgctgccctg gcatcagcct cttccagtca catctggcca gagggctgtc 1799
    tggacctggg ccctgctcaa tgccacctgt ctgcctgggc tccaagtggg caggaccagg 1859
    acagaaccac aggcacacac tgagggggca gtgtggctcc ctgcctgtcc catccccatg 1919
    ccccgtccgc ggggctgtgg ctgctgctgt gcatgtccct gcgatgggag tcttgtctcc 1979
    cagcctgtca gtttcctccc cagggcagag ctccccttcc tgcaagagtc tgggaggcgg 2039
    tgcaggctgt cctggctgct ctggggaagc cgagggacag ccataacacc cccgggacag 2099
    taggtctggg cggcaccact gggaactctg gacttgagtg tgtttgctct tccttgggta 2159
    tgaatgtgtg agttcaccca gaggcctgct ctcctcacac attgtgtggt ttggggttaa 2219
    tgatggaggg agacacctct tcatagacgg caggtgccca cctttcaggg agtctcccag 2279
    catgggcgga tgccgggcat gagctgctgt aaactatttg tggctgtgct gcttgagtga 2339
    cgtctctgtc gtgtgggtgc caagtgcttg tgtagaaact gtgttctgag cccccttttc 2399
    tggacaccaa ctgtgtcctg tgaatgtatc gctactgtga gctgttcccg cctagccagg 2459
    gccatgtctt aggtgcagct gtgccacggg tcagctgagc cacagtccca gaaccaagct 2519
    ctcggtgtct cgggccacca tccgcccacc tcgggctgac cccacctcct ccatggacag 2579
    tgtgagcccc gggccgtgca tcctgctcag tgtggcgtca gtgtcggggc tgagcccctt 2639
    gagctgcttc agtgaatgta cagtgcccgg cacgagctga acctcatgtg ttccactccc 2699
    aataaaaggt tgacaggggc ttctccttca aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 2759
    aaa 2762
    <210> SEQ ID NO 27
    <211> LENGTH: 484
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 27
    Met Lys Lys Gly Ile Arg Tyr Glu Thr Ser Arg Lys Thr Ser Tyr Ile
    1 5 10 15
    Phe Gln Gln Pro Gln His Gly Pro Trp Gln Thr Arg Met Arg Lys Ile
    20 25 30
    Ser Asn His Gly Ser Leu Arg Val Ala Lys Val Ala Tyr Pro Leu Gly
    35 40 45
    Leu Cys Val Gly Val Phe Ile Tyr Val Ala Tyr Ile Lys Trp His Arg
    50 55 60
    Ala Thr Ala Thr Gln Ala Phe Phe Ser Ile Thr Arg Ala Ala Pro Gly
    65 70 75 80
    Ala Arg Trp Gly Gln Gln Ala His Ser Pro Leu Gly Thr Ala Ala Asp
    85 90 95
    Gly His Glu Val Phe Tyr Gly Ile Met Phe Asp Ala Gly Ser Thr Gly
    100 105 110
    Thr Arg Val His Val Phe Gln Phe Thr Arg Pro Pro Arg Glu Thr Pro
    115 120 125
    Thr Leu Thr His Glu Thr Phe Lys Ala Val Lys Pro Gly Leu Ser Ala
    130 135 140
    Tyr Ala Asp Asp Val Glu Lys Ser Ala Gln Gly Ile Arg Glu Leu Leu
    145 150 155 160
    Asp Val Ala Lys Gln Asp Ile Pro Phe Asp Phe Trp Lys Ala Thr Pro
    165 170 175
    Leu Val Leu Lys Ala Thr Ala Gly Leu Arg Leu Leu Pro Gly Glu Lys
    180 185 190
    Ala Gln Lys Leu Leu Gln Lys Val Lys Glu Val Phe Lys Ala Ser Pro
    195 200 205
    Phe Leu Val Gly Asp Asp Cys Val Ser Ile Met Asn Gly Thr Asp Glu
    210 215 220
    Gly Val Ser Ala Trp Ile Thr Ile Asn Phe Leu Thr Gly Ser Leu Lys
    225 230 235 240
    Thr Pro Gly Gly Ser Ser Val Gly Met Leu Asp Leu Gly Gly Gly Ser
    245 250 255
    Thr Gln Ile Ala Phe Leu Pro Arg Val Glu Gly Thr Leu Gln Ala Ser
    260 265 270
    Pro Pro Gly Tyr Leu Thr Ala Leu Arg Met Phe Asn Arg Thr Tyr Lys
    275 280 285
    Leu Tyr Ser Tyr Ser Tyr Leu Gly Leu Gly Leu Met Ser Ala Arg Leu
    290 295 300
    Ala Ile Leu Gly Gly Val Glu Gly Gln Pro Ala Lys Asp Gly Lys Glu
    305 310 315 320
    Leu Val Ser Pro Cys Leu Ser Pro Ser Phe Lys Gly Glu Trp Glu His
    325 330 335
    Ala Glu Val Thr Tyr Arg Val Ser Gly Gln Lys Ala Ala Ala Ser Leu
    340 345 350
    His Glu Leu Cys Ala Ala Arg Val Ser Glu Val Leu Gln Asn Arg Val
    355 360 365
    His Arg Thr Glu Glu Val Lys His Val Asp Phe Tyr Ala Phe Ser Tyr
    370 375 380
    Tyr Tyr Asp Leu Ala Ala Gly Val Gly Leu Ile Asp Ala Glu Lys Gly
    385 390 395 400
    Gly Ser Leu Val Val Gly Asp Phe Glu Ile Ala Ala Lys Tyr Val Cys
    405 410 415
    Arg Thr Leu Glu Thr Gln Pro Gln Ser Ser Pro Phe Ser Cys Met Asp
    420 425 430
    Leu Thr Tyr Val Ser Leu Leu Leu Gln Glu Phe Gly Phe Pro Arg Ser
    435 440 445
    Lys Val Leu Lys Leu Thr Arg Lys Ile Asp Asn Val Glu Thr Ser Trp
    450 455 460
    Ala Leu Gly Ala Ile Phe His Tyr Ile Asp Ser Leu Asn Arg Gln Lys
    465 470 475 480
    Ser Pro Ala Ser
    <210> SEQ ID NO 28
    <211> LENGTH: 27
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 28
    cgtatcccgc gggtggaggc cggggtg 27
    <210> SEQ ID NO 29
    <211> LENGTH: 27
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 29
    cttctgcaag tcccagagcc agtgtgc 27
    <210> SEQ ID NO 30
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 30
    ggagcccaaa agaccggctg c 21
    <210> SEQ ID NO 31
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 31
    tgaagtcacg tccaggacag g 21
    <210> SEQ ID NO 32
    <211> LENGTH: 36
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 32
    cggaattcaa catgaaaaaa ggtaatccgt tatgaa 36
    <210> SEQ ID NO 33
    <211> LENGTH: 26
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence: primer
    <400> SEQUENCE: 33
    tgtctagatg aggctggact cttctg 26
    <210> SEQ ID NO 34
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence:
    oligonucleotide primer
    <400> SEQUENCE: 34
    atcctggact tgagcctaga g 21
    <210> SEQ ID NO 35
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence:
    oligonucleotide primer
    <400> SEQUENCE: 35
    ctgatattga tgggtcttgg g 21
    <210> SEQ ID NO 36
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence:
    oligonucleotide primer
    <400> SEQUENCE: 36
    ggatggaaag gagttggtca g 21
    <210> SEQ ID NO 37
    <211> LENGTH: 21
    <212> TYPE: DNA
    <213> ORGANISM: Artificial Sequence
    <220> FEATURE:
    <223> OTHER INFORMATION: Description of Artificial Sequence:
    oligonucleotide primer
    <400> SEQUENCE: 37
    gtccacatgc ttcacttcct c 21
    <210> SEQ ID NO 38
    <211> LENGTH: 502
    <212> TYPE: PRT
    <213> ORGANISM: Homo sapiens
    <400> SEQUENCE: 38
    Met Glu Asp Thr Lys Glu Ser Asn Val Lys Thr Phe Cys Ser Lys Asn
    1 5 10 15
    Ile Leu Ala Ile Leu Gly Phe Ser Ser Ile Ile Ala Val Ile Ala Leu
    20 25 30
    Leu Ala Val Gly Leu Thr Gln Asn Lys Ala Leu Pro Glu Asn Val Lys
    35 40 45
    Tyr Gly Ile Val Leu Asp Ala Gly Ser Ser His Thr Ser Leu Tyr Ile
    50 55 60
    Tyr Lys Trp Pro Ala Glu Lys Glu Asn Asp Thr Gly Val Val His Gln
    65 70 75 80
    Val Glu Glu Cys Arg Val Lys Gly Pro Gly Ile Ser Lys Phe Val Gln
    85 90 95
    Lys Val Asn Glu Ile Gly Ile Tyr Leu Thr Asp Cys Met Glu Arg Ala
    100 105 110
    Arg Glu Val Ile Pro Arg Ser Gln His Gln Glu Thr Pro Val Tyr Leu
    115 120 125
    Gly Ala Thr Ala Gly Met Arg Leu Leu Arg Met Glu Ser Glu Glu Leu
    130 135 140
    Ala Asp Arg Val Leu Asp Val Val Glu Arg Ser Leu Ser Asn Tyr Pro
    145 150 155 160
    Phe Asp Phe Gln Gly Ala Arg Ile Ile Thr Gly Gln Glu Glu Gly Ala
    165 170 175
    Tyr Gly Trp Ile Thr Ile Asn Tyr Leu Leu Gly Lys Phe Ser Gln Lys
    180 185 190
    Thr Arg Trp Phe Ser Ile Val Pro Tyr Glu Thr Asn Asn Gln Glu Thr
    195 200 205
    Phe Gly Ala Leu Asp Leu Gly Gly Ala Ser Thr Gln Val Thr Phe Val
    210 215 220
    Pro Gln Asn Gln Thr Ile Glu Ser Pro Asp Asn Ala Leu Gln Phe Arg
    225 230 235 240
    Leu Tyr Gly Lys Asp Tyr Asn Val Tyr Thr His Ser Phe Leu Cys Tyr
    245 250 255
    Gly Lys Asp Gln Ala Leu Trp Gln Lys Leu Ala Lys Asp Ile Gln Val
    260 265 270
    Ala Ser Asn Glu Ile Leu Arg Asp Pro Cys Phe His Pro Gly Tyr Lys
    275 280 285
    Lys Val Val Asn Val Ser Asp Leu Tyr Lys Thr Pro Cys Thr Lys Arg
    290 295 300
    Phe Glu Met Thr Leu Pro Phe Gln Gln Phe Glu Ile Gln Gly Ile Gly
    305 310 315 320
    Asn Tyr Gln Gln Cys His Gln Ser Ile Leu Glu Leu Phe Asn Thr Ser
    325 330 335
    Tyr Cys Pro Tyr Ser Gln Cys Ala Phe Asn Gly Ile Phe Leu Pro Pro
    340 345 350
    Leu Gln Gly Asp Phe Gly Ala Phe Ser Ala Phe Tyr Phe Val Met Lys
    355 360 365
    Phe Leu Asn Leu Thr Ser Glu Lys Val Ser Gln Glu Lys Val Thr Glu
    370 375 380
    Met Met Lys Lys Phe Cys Ala Gln Pro Trp Glu Glu Ile Lys Thr Ser
    385 390 395 400
    Tyr Ala Gly Val Lys Glu Lys Tyr Leu Ser Glu Tyr Cys Phe Ser Gly
    405 410 415
    Thr Tyr Ile Leu Ser Leu Leu Leu Gln Gly Tyr His Phe Thr Ala Asp
    420 425 430
    Ser Trp Glu His Ile His Phe Ile Gly Lys Ile Gln Gly Ser Asp Ala
    435 440 445
    Gly Trp Thr Leu Gly Tyr Met Leu Asn Leu Thr Asn Met Ile Pro Ala
    450 455 460
    Glu Gln Pro Leu Ser Thr Pro Leu Ser His Ser Thr Tyr Val Phe Leu
    465 470 475 480
    Met Val Leu Phe Ser Leu Val Leu Phe Thr Val Ala Ile Ile Gly Leu
    485 490 495
    Leu Ile Phe His Lys Pro
    500
    <210> SEQ ID NO 39
    <211> LENGTH: 465
    <212> TYPE: PRT
    <213> ORGANISM: Mus musculus
    <400> SEQUENCE: 39
    Met Ala Thr Ser Trp Gly Ala Val Phe Met Leu Ile Ile Ala Cys Val
    1 5 10 15
    Gly Ser Thr Val Phe Tyr Arg Glu Gln Gln Thr Trp Phe Glu Gly Val
    20 25 30
    Phe Leu Ser Ser Met Cys Pro Ile Asn Val Ser Ala Gly Thr Phe Tyr
    35 40 45
    Gly Ile Met Phe Asp Ala Gly Ser Thr Gly Thr Arg Ile His Val Tyr
    50 55 60
    Thr Phe Val Gln Lys Thr Ala Gly Gln Leu Pro Phe Leu Glu Gly Glu
    65 70 75 80
    Ile Phe Asp Ser Val Lys Pro Gly Leu Ser Ala Phe Val Asp Gln Pro
    85 90 95
    Lys Gln Gly Ala Glu Thr Val Gln Glu Leu Leu Glu Val Ala Lys Asp
    100 105 110
    Ser Ile Pro Arg Ser His Trp Glu Arg Thr Pro Val Val Leu Lys Ala
    115 120 125
    Thr Ala Gly Leu Arg Leu Leu Pro Glu Gln Lys Ala Gln Ala Leu Leu
    130 135 140
    Leu Glu Val Glu Glu Ile Phe Lys Asn Ser Pro Phe Leu Val Pro Asp
    145 150 155 160
    Gly Ser Val Ser Ile Met Asp Gly Ser Tyr Glu Gly Ile Leu Ala Trp
    165 170 175
    Val Thr Val Asn Phe Leu Thr Gly Gln Leu His Gly Arg Gly Gln Glu
    180 185 190
    Thr Val Gly Thr Leu Asp Leu Gly Gly Ala Ser Thr Gln Ile Thr Phe
    195 200 205
    Leu Pro Gln Phe Glu Lys Thr Leu Glu Gln Thr Pro Arg Gly Tyr Leu
    210 215 220
    Thr Ser Phe Glu Met Phe Asn Ser Thr Phe Lys Leu Tyr Thr His Ser
    225 230 235 240
    Tyr Leu Gly Phe Gly Leu Lys Ala Ala Arg Leu Ala Thr Leu Gly Ala
    245 250 255
    Leu Glu Ala Lys Gly Thr Asp Gly His Thr Phe Arg Ser Ala Cys Leu
    260 265 270
    Pro Arg Trp Leu Glu Ala Glu Trp Ile Phe Gly Gly Val Lys Tyr Gln
    275 280 285
    Tyr Gly Gly Asn Gln Glu Gly Glu Met Gly Phe Glu Pro Cys Tyr Ala
    290 295 300
    Glu Val Leu Arg Val Val Gln Gly Lys Leu His Gln Pro Glu Glu Val
    305 310 315 320
    Arg Gly Ser Ala Phe Tyr Ala Phe Ser Tyr Tyr Tyr Asp Arg Ala Ala
    325 330 335
    Asp Thr His Leu Ile Asp Tyr Glu Lys Gly Gly Val Leu Lys Val Glu
    340 345 350
    Asp Phe Glu Arg Lys Ala Arg Glu Val Cys Asp Asn Leu Gly Ser Phe
    355 360 365
    Ser Ser Gly Ser Pro Phe Leu Cys Met Asp Leu Thr Tyr Ile Thr Ala
    370 375 380
    Leu Leu Lys Asp Gly Leu Gly Phe Ala Glu Arg His Pro Leu Thr Ala
    385 390 395 400
    His Lys Glu Ser Glu Gln His Arg Asp Trp Leu Gly Leu Gly Gly His
    405 410 415
    Leu Ser Pro Ala Pro Val Ser Gly His His Gln Leu Arg Pro Ser Ser
    420 425 430
    Thr Ser Glu Ala Cys Ile Ser Glu Pro Val Phe Ser Gln Glu Gly Val
    435 440 445
    Asp Ser Glu Thr Phe Ser Asp Leu Ser Gly Lys Ala Trp Pro Glu Thr
    450 455 460
    Arg
    465

Claims (45)

What is claimed is:
1. An isolated polynucleotide encoding an apyrase and/or NDPase and comprising a nucleotide sequence having at least about 80% sequence identity to a human polynucleotide selected from the group consisting of:
(a) a polynucleotide having the nucleotide sequence of SEQ ID NO. 2; and
(b) a polynucleotide having the protein coding nucleotide sequence of the polynucleotide sequence of (a).
2. An isolated polynucleotide encoding an apyrase and comprising a nucleotide sequence having at least about 90% sequence identity to a polynucleotide selected from the group consisting of:
(a) a polynucleotide having the nucleotide sequence of SEQ ID NO. 2; and
(b) a polynucleotide having the protein coding nucleotide sequence of the polynucleotide sequence of (a).
3. An isolated polynucleotide encoding a polypeptide with apyrase and/or NDPase activity, comprising a polynucleotide selected from the group consisting of:
(a) polynucleotides that encode the mature protein coding amino acid sequence of SEQ ID NO. 3.
4. An isolated polynucleotide encoding a polypeptide with apyrase and/or NDPase activity that hybridizes under stringent conditions to the complement of a polynucleotide of SEQ ID NO. 2.
5. The polynucleotide of any one of claims 1 through 4 which is a DNA.
6. The DNA of claim 5 which is a wholly or partially chemically synthesized DNA molecule.
7. An anti-sense polynucleotide which specifically hybridizes with the complement of the polynucleotide of claim 4.
8. The polynucleotide of claim 1 which comprises the nucleotide sequence of SEQ ID NO. 2 or 24 or the mature protein coding portions thereof.
9. An isolated polynucleotide which comprises a complement of the polynucleotide of claim 1.
10. An expression vector comprising the DNA of claim 5.
11. A host cell comprising the DNA of claim 5.
12. A host cell genetically engineered to express the DNA of claim 5.
13. An isolated human polypeptide with apyrase and/or NDPase activity comprising:
(a) the mature protein coding sequence of SEQ ID NO. 3; or
(b) an amino acid sequence having at least about 80% sequence identity to SEQ ID NO. 3.
14. An isolated polypeptide with apyrase and/or NDPase activity comprising:
(a) the CD39-like protein coding sequence of SEQ ID NO. 3; or
(b) an amino acid sequence having at least about 90% sequence identity to SEQ ID NO. 3.
15. The polypeptide of claim 13 or 14 which comprises the amino acid sequence of SEQ ID NO. 3 or 25 or the mature protein portions thereof.
16. The polypeptide of claim 13 or 14 wherein the polypeptide comprises at least one amino acid substitution selected from the group consisting of: D168→T, S170→Q and L175→F.
17. The polypeptide of claim 16 comprising a polypeptide having the amino acid sequence set forth in SEQ ID NO. 7.
18. A method for producing a CD39-like polypeptide comprising the steps of:
(a) growing a culture of cells according to claim 11 under conditions permitting expression of a CD39-like polypeptide; and
(b) isolating the CD39-like polypeptide from the host cell or its growth medium.
19. A composition comprising the polypeptide of claim 13, 14 or 15 and a pharmaceutically acceptable carrier.
20. An antibody specifically immunoreactive with a polypeptide encoded by the polynucleotide according to claim 1.
21. The antibody according to claim 20 which is a monoclonal antibody.
22. A hybridoma which secretes the antibody according to claim 21.
23. An anti-idiotype antibody specifically immunoreactive with the antibody according to claim 21.
24. A method for detecting a polynucleotide of claim 1, 2 or 3 in a sample comprising the steps of:
(a) contacting the sample with a compound that binds to and forms a complex with the polynucleotide for a period sufficient to detect the complex; and
(b) detecting the complex so that if a complex is detected, a polynucleotide of claim 1, 2 or 3 is detected.
25. A method for detecting a polynucleotide of claim 1, 2 or 3 in a sample comprising the steps of:
(a) contacting the sample under stringent hybridization conditions with nucleic acid primers that anneal to a polynucleotide of claim 1, 2 or 3 under such conditions; and
(b) amplifying the polynucleotides of claim 1, 2 or 3 so that if a polynucleotide is amplified, a polynucleotide of claim 1, 2 or 3 is detected.
26. The method of claim 25 wherein the polynucleotide is an RNA molecule that encodes a polypeptide of claim 13 or 14, and the method further comprises reverse transcribing an annealed RNA molecule into a cDNA polynucleotide.
27. A method for detecting a polypeptide of claim 13 or 14 in a sample comprising:
(a) contacting the sample with a compound that binds to and forms a complex with the polypeptide for a period sufficient to detect the complex; and
(b) detecting the complex so that if a complex is detected, a polypeptide of claim 13 or 14 is detected.
28. A method for identifying a compound that binds to a polypeptide of claim 13 or 14 comprising:
(a) contacting a compound with a polypeptide of claim 13 or 14 for a time sufficient to form a polypeptide/compound complex; and
(b) detecting the complex so that if a polypeptide/compound complex is detected, a compound that binds to a polypeptide of claim 13 or 14 is detected.
29. A method for identifying a compound that binds to a polypeptide of claim 13 or 14 comprising:
(a) contacting a compound with a polypeptide of claim 13 or 14, in a cell, for a time sufficient to form a polypeptide/compound complex, wherein the complex drives expression of a reporter gene sequence in the cell, and
(b) detecting the complex by detecting reporter gene sequence expression so that if a polypeptide/compound complex is detected, a compound that binds to a polypeptide of claim 13 or 14 is identified.
30. A method of identifying a modulator compound of a CD39-like polypeptide with apyrase activity comprising the steps of:
(a) contacting the CD39-like polypeptide encoded by the polynucleotide of claim 1, 2 or 3 with a substrate in the presence and absence of a test compound;
(b) comparing apyrase activity of the CD39-like polypeptide in the presence and absence of the test compound; and
(c) identifying the test compound as a modulator compound when biological activity of the CD39-like polypeptide is increased or decreased in the presence of the test compound.
31. A method of identifying a modulator compound of a CD39-like polypeptide with NDPase activity comprising the steps of:
(a) contacting the CD39-like polypeptide encoded by the polynucleotide of claim 1, 2 or 3 with a substrate in the presence and absence of a test compound;
(b) comparing NDPase activity of the CD39-like polypeptide in the presence and absence of the test compound; and
(c) identifying the test compound as a modulator compound when biological activity of the CD39-like polypeptide is increased or decreased in the presence of the test compound.
32. A chimeric polypeptide comprising one or more domains of a CD39-like polypeptide fused to one or more domains of heterologous peptide or polypeptide, e.g., an immunoglobulin constant region.
33. A method of treatment comprising administering to a mammalian subject in need thereof a therapeutic amount of a composition comprising a polypeptide of claim 13 or 14 and a pharmaceutically acceptable carrier.
34. A method of treatment comprising administering to a mammalian subject in need thereof a therapeutic amount of a composition comprising an antibody that specifically binds to a polypeptide of claim 13 or 14 and a pharmaceutically acceptable carrier.
35. A method of inhibiting platelet function comprising administering the polypeptide of claim 13 or 14 to a medium comprising platelets.
36. A method of treating thrombotic diseases comprising administering a therapeutic amount of the polypeptide of claim 13 or 14 to a mammalian subject in need thereof.
37. A method of hydrolyzing nucleotidediphosphates comprising administering the polypeptide of claim 13 or 14 to a medium comprising nucleotidediphospates.
38. A method of inhibiting platelet aggregation in a mammalian subject comprising the step of reducing the ratio of ADP:ATP in said mammalian subject to a less than normal ratio.
39. The method of claim 38 wherein said ratio is reduced by administration of CD39-L4 having the sequence set forth in SEQ ID NO: 3 or a polypeptide having NDPase activity and at least about 90% sequence identity to SEQ ID NO: 3.
40. The method of claim 38 wherein said ratio is reduced by administration of CD39-L66 having the sequence set forth in SEQ ID NO: 25 or a polypeptide having NDPase activity and at least about 90% sequence identity to SEQ ID NO: 25.
41. The method of claim 38 wherein said ratio is reduced by administration of CD39-L2 having the sequence set forth in SEQ ID NO: 27 or a polypeptide having NDPase activity and at least about 90% sequence identity to SEQ ID NO: 27.
42. The method of claim 38 - 41 wherein the ratio of ADP:ATP is reduced systemically in circulation.
43. The method of claim 38 - 41 wherein the ratio of ADP:ATP is reduced locally within an area selected from the group consisting of heart, brain, kidney, lung and limbs.
44. The method of claim 38-41 wherein the ratio of ADP:ATP is reduced without significantly affecting ATP levels.
45. A method of identifying a compound capable of reducing the ratio of ADP:ATP to a less than normal ratio comprising the steps of:
(a) determining apyrase activity of said compound on ATP;
(b) determining apyrase activity of said compounds on ADP; and
(c) selecting a compound that has greater activity with respect to ADP compared to ATP.
US10/092,063 1998-07-16 2002-03-05 Methods and materials relating to novel CD39-like polypeptides Abandoned US20020173005A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/092,063 US20020173005A1 (en) 1998-07-16 2002-03-05 Methods and materials relating to novel CD39-like polypeptides

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US11820598A 1998-07-16 1998-07-16
US12244998A 1998-07-24 1998-07-24
US24444499A 1999-02-04 1999-02-04
US27344799A 1999-03-19 1999-03-19
US09/350,836 US6387645B1 (en) 1998-07-16 1999-07-09 Methods and materials relating to novel CD39-like polypeptides
PCT/US1999/016180 WO2000004041A2 (en) 1998-07-16 1999-07-16 Methods and molecules relating to cd39-like polypeptides
US09/370,265 US6447771B1 (en) 1999-03-19 1999-08-09 Methods and materials relating to novel CD39-like polypeptides
US10/092,063 US20020173005A1 (en) 1998-07-16 2002-03-05 Methods and materials relating to novel CD39-like polypeptides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/370,265 Continuation US6447771B1 (en) 1998-07-16 1999-08-09 Methods and materials relating to novel CD39-like polypeptides

Publications (1)

Publication Number Publication Date
US20020173005A1 true US20020173005A1 (en) 2002-11-21

Family

ID=26956202

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/370,265 Expired - Fee Related US6447771B1 (en) 1998-07-16 1999-08-09 Methods and materials relating to novel CD39-like polypeptides
US10/092,063 Abandoned US20020173005A1 (en) 1998-07-16 2002-03-05 Methods and materials relating to novel CD39-like polypeptides

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/370,265 Expired - Fee Related US6447771B1 (en) 1998-07-16 1999-08-09 Methods and materials relating to novel CD39-like polypeptides

Country Status (1)

Country Link
US (2) US6447771B1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6447771B1 (en) * 1999-03-19 2002-09-10 Hyseq, Inc. Methods and materials relating to novel CD39-like polypeptides
US20090130092A1 (en) 2007-10-30 2009-05-21 Pinsky David J Nucleotide phosphate dissipation as a treatment for vascular disorders
US11345757B2 (en) 2017-07-31 2022-05-31 Trishula Therapeutics, Inc. Anti-CD39 antibodies

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) * 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) * 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4737323A (en) * 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US4837028A (en) * 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4959314A (en) * 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US4965188A (en) * 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US5202231A (en) * 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US5525464A (en) * 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US6350447B1 (en) * 1999-01-29 2002-02-26 Hyseq, Inc. Methods and compositions relating to CD39-like polypeptides and nucleic acids
US6387645B1 (en) * 1998-07-16 2002-05-14 Hyseq, Inc. Methods and materials relating to novel CD39-like polypeptides
US6447771B1 (en) * 1999-03-19 2002-09-10 Hyseq, Inc. Methods and materials relating to novel CD39-like polypeptides

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8822228D0 (en) 1988-09-21 1988-10-26 Southern E M Support-bound oligonucleotides
EP0434792A4 (en) 1989-05-19 1992-05-20 John B. Fenn Multiply charged ions and a method for determining the molecular weight of large molecules
ATE264916T1 (en) 1989-12-22 2004-05-15 Applied Research Systems DNA CONSTRUCTS TO ACTIVATE AND ALTER THE EXPRESSION OF ENDOGENE GENES
CA2092695A1 (en) 1991-05-15 1992-11-16 Oliver Smithies Genomic modifications with homologous dna targeting
TW402639B (en) 1992-12-03 2000-08-21 Transkaryotic Therapies Inc Protein production and protein delivery
AU694146B2 (en) 1993-09-27 1998-07-16 Arch Development Corporation Methods and compositions for efficient nucleic acid sequencing
EP0815252A1 (en) 1995-03-24 1998-01-07 Novartis AG Gene therapy for transplantation and inflammatory or thrombotic conditions
WO1996032471A2 (en) 1995-04-10 1996-10-17 Universite De Sherbrooke Atp-diphosphohydrolases, process of purification thereof and process of producing thereof by recombinant technology
US5620706A (en) 1995-04-10 1997-04-15 Universite De Sherbrooke Polyionic insoluble hydrogels comprising xanthan and chitosan

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4235871A (en) * 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4501728A (en) * 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4959314A (en) * 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683195B1 (en) * 1986-01-30 1990-11-27 Cetus Corp
US4737323A (en) * 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US4965188A (en) * 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4837028A (en) * 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5202231A (en) * 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US5525464A (en) * 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6387645B1 (en) * 1998-07-16 2002-05-14 Hyseq, Inc. Methods and materials relating to novel CD39-like polypeptides
US6350447B1 (en) * 1999-01-29 2002-02-26 Hyseq, Inc. Methods and compositions relating to CD39-like polypeptides and nucleic acids
US6447771B1 (en) * 1999-03-19 2002-09-10 Hyseq, Inc. Methods and materials relating to novel CD39-like polypeptides

Also Published As

Publication number Publication date
US6447771B1 (en) 2002-09-10

Similar Documents

Publication Publication Date Title
US5646040A (en) Mammalian tub gene
US20020187472A1 (en) Steap-related protein
JPH09506250A (en) Protein tyrosine kinase named Rse
CA2316403A1 (en) Mammalian edg-5 receptor homologs
US20030082533A1 (en) Intelectin
JP2003502041A (en) Lipocalin family proteins
US6335013B1 (en) Methods and materials relating to CD39-like polypeptides
AU762392B2 (en) Methods and materials relating to novel CD39-like polypeptides
US6476211B1 (en) Methods and materials relating to CD39-like polypeptides
US20140155288A1 (en) Methods for diagnosing cancer
US5861239A (en) Methods for identifying compounds that modulate mammalian tub protein activity
US6387645B1 (en) Methods and materials relating to novel CD39-like polypeptides
US20020142375A1 (en) Mammalian EDG-7 receptor homologs
US6447771B1 (en) Methods and materials relating to novel CD39-like polypeptides
JP2003525634A (en) Protein named FCTRX and nucleic acid encoding the same
JP2002506875A (en) Methods and compositions for diagnosis and treatment of chromosome 18p-related disorders
US6590089B1 (en) RVP-1 variant differentially expressed in Crohn&#39;s disease
US20030054446A1 (en) Novel retina-specific human proteins C7orf9, C12orf7, MPP4 and F379
US20030167480A1 (en) NDR2-related proteins
US6632617B1 (en) Tumor-associated antigen
US20030165989A1 (en) GPCR diagnostic for brain cancer
CA2422508A1 (en) Atp-binding cassette protein
HUYNH 1. RELATED APPLICATIONS
US20020132238A1 (en) Progesterone receptor complex p23-like protein
US20020137038A1 (en) Intestinal proteins

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION