Genes associated with vascular disease

Information

  • Patent Application
  • 20030166903
  • Publication Number
    20030166903
  • Date Filed
    April 25, 2002
    22 years ago
  • Date Published
    September 04, 2003
    20 years ago
Abstract
The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed in vascular endothelium and which may be used in their entirety or in part to diagnose, to stage, to treat, or to monitor the treatment of a subject with a vascular disorder.
Description


FIELD OF THE INVENTION

[0002] The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed in vascular endothelium and which may be used entirely or in part to diagnose, to stage, to treat, or to monitor the progression or treatment of disorders of the vascular system including atherosclerosis, diabetic retinopathy, endometriosis, obesity, and cancer.



BACKGROUND OF THE INVENTION

[0003] The blood vessel wall is a complex tissue that plays a critical role in numerous physiological processes such as maintaining blood pressure, controlling selective permeability, directing vessel remodeling and angiogenesis, and modulating inflammatory and immune responses. Vessel walls are composed of three morphologically distinct layers, the intima, media, and adventitia, arranged concentrically from the lumenal surface outward. The intima is comprised of an endothelial cell (EC) layer in direct contact with the blood, surrounded by a layer of subendothelial connective tissue, within which lies the internal elastic lamina. The media is comprised of vascular smooth muscle cells (VSMC) and elastic fibrous components. The adventitia is comprised of the external elastic lamina surrounded on the outside by a layer of extracellular matrix components interspersed with fibroblasts and smooth muscle cells.


[0004] During embryonic development, blood vessels are formed de novo by vasculogenesis, the assembly of endothelial precursors. Later, the primitive vascular tree is expanded by angiogenesis, the development of new vessels from preexisting vessels. New vessels are formed both by sprouting of branches (sprouting angiogenesis) and by the partitioning of preexisting vessels via insertion of interstitial tissue columns (intussuseption). In adults, angiogenesis occurs normally during wound healing and during the menstrual cycle when the corpus luteum is formed. A key stimulus for angiogenesis is the production of a signal from hypoxic or otherwise stressed tissues that is detected by receptors on nearby microvascular endothelial cells. The first such signal to be identified was the angiogenesis inducing molecule vascular endothelial growth factor (VEGF).


[0005] Blood vessel structure and function vary considerably in different parts of the vascular tree. For example, vessel diameter and wall thickness decrease, along with changes in wall composition with increasing distance from the heart. Arterial walls are relatively thick compared with their venous counterparts, enabling them to withstand the higher blood pressures and shear forces to which they are exposed. The walls of the largest “elastic arteries” close to the heart have a thick medial layer, rich in elastic fibers which allow it to expand and contract with the heart. These elastic components are lacking in the small arteriole walls which play a major role in systemic blood pressure regulation. At the level of the microvascular capillary beds, where vessels have a diameter of about 7-8 μm, vessels are composed of a simple endothelial cell layer with a basement membrane composed primarily of type IV collagen.


[0006] Different types of capillary walls can be distinguished based on the degree of continuity and basement membrane composition.


[0007] Differences in vessel wall structure and function influence the diseases with which particular vessels are associated. For example, tumor angiogenesis and diabetic retinopathy are pathological conditions associated with abnormal growth of microvascular endothelium. By contrast, atherosclerosis affects the large elastic and muscular arteries such as the aorta and carotid, iliac, coronary, and popliteal arteries, but does not affect small muscular arteries or arterioles, large or small veins, or microvascular tissues.


[0008] Atherosclerosis and the associated coronary artery disease and cerebral stroke represent the most common causes of death in industrialized nations. Atherosclerosis is a progressive disease characterized by a chronic local inflammatory response within the vessel wall of large arteries (Lusis (2000) Nature 407:233-241). Although certain key risk factors have been identified, a full molecular characterization that elucidates the causes and identifies all potential therapeutic targets for this complex disease has not been achieved. Pathological angiogenesis occurs in numerous conditions such as diabetic retinopathy, endometriosis, obesity, and cancer (Carmeliet et al. (2000) Nature 407:249-257). The demonstration that tumor induced angiogenesis is a critical stage in tumor progression, potentially amenable to clinical intervention, has led to a great deal of interest in factors which mediate tumor angiogenesis. Molecules involved in angiogenesis act to stimulate endothelial proliferation, migration, and morphogenesis, and remodel extracellular matrix.


[0009] A number of primary endothelial cell lines are available to study vascular endothelial cell function. Primary cells derived from the endothelia of human aorta (HAEC), coronary artery (HCAEC), iliac artery (HIAEC), brain microvasculature, (HMVECB), neonatal dermal microvasculature (HMVECD), pulmonary artery (HPAEC), umbilical artery (HUAEC), umbilical vein (HUVEC), and uterine myometrium microvasculature (UtMVEC) have all been used as experimental models for investigating in vitro the role of the endothelium in human vascular biology.


[0010] Array technology can provide a simple way to explore the expression of a single polymorphic gene or to produce an expression profile of a large number of related or unrelated genes. Arrays provide a platform for examining which genes are tissue specific, carrying out housekeeping functions, parts of a signaling cascade, or specifically related to a particular genetic predisposition, condition, disease, or disorder. The potential application of gene expression profiling is particularly relevant to improving diagnosis, prognosis, and treatment of disease. For example, both the levels and sequences expressed in vascular endothelium from subjects with atherosclerosis may be compared with the levels and sequences expressed in normal vascular tissue.


[0011] The present invention provides for a combination comprising a plurality of cDNAs for use in detecting changes in expression of genes encoding proteins that are associated with a vascular disorder. The present invention satisfies a need in the art by identifying differentially expressed genes which may be used entirely or in part to diagnose, to stage, to treat, or to monitor the progression or treatment of a subject with a vascular disorder such as atherosclerosis, diabetic retinopathy, endometriosis, obesity, and cancer.



SUMMARY

[0012] The present invention provides a combination comprising a plurality of cDNAs and their complements which are differentially expressed in vascular endothelial tissues and which are selected from SEQ ID NOs:1-292 as presented in the Sequence Listing. In one embodiment, each cDNA is expressed at higher levels in microvascular endothelium, SEQ ID NOs:1-209; in another embodiment, each cDNA is expressed at higher levels in large artery endothelium, SEQ ID NOs:210-292. In one aspect, the combination is useful to diagnose a vascular disorder selected from atherosclerosis, hemangioma, hemangioendothelioma, diabetic retinopathy, warts, pyogenic granulomas, Kaposi's sarcoma, scar keloids, allergic oedema, neoplasms, psoriasis, ulcers, follicular cysts, endometriosis, peritoneal sclerosis, and obesity. In another aspect, the combination is immobilized on a substrate.


[0013] The invention also provides a high throughput method to detect differential expression of one or more of the cDNAs of the combination. The method comprises hybridizing the substrate comprising the combination with the nucleic acids of a sample, thereby forming one or more hybridization complexes, detecting the hybridization complexes, and comparing the hybridization complexes with those of a standard, wherein differences in the size and signal intensity of each hybridization complex indicates differential expression of nucleic acids in the sample. In one aspect, the sample is from a subject with a vascular disorder and differential expression determines an early, mid, and late stage of the disorder.


[0014] The invention further provides a high throughput method of screening a library or a plurality of molecules or compounds to identify a ligand. The method comprises combining the substrate comprising the combination with a library or a plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand. The library or plurality of molecules or compounds are selected from DNA molecules, enhancers, mimetics, peptide nucleic acids, proteins, repressors, regulatory proteins, RNA molecules, and transcription factors. The invention additionally provides a method for purifying a ligand, the method comprising combining a cDNA of the invention with a sample under conditions which allow specific binding, recovering the bound cDNA, and separating the cDNA from the ligand, thereby obtaining purified ligand.


[0015] The invention provides an isolated cDNA selected from SEQ ID NOs:36, 51, 61, 83, 87, 110, 111, 136, 159, 198, and 244 as presented in the Sequence Listing. The invention also provides a vector comprising the cDNA, a host cell comprising the vector, and a method for producing a protein comprising culturing the host cell under conditions for the expression of a protein and recovering the protein from the host cell culture. The invention provides a method for using an isolated cDNA to detect differential gene expression in a sample, the method comprising hybridizing a cDNA having a nucleotide sequence selected from SEQ ID NOs:36, 51, 61, 83, 87, 110, 111, 136, 159, 198, and 244 with a sample containing nucleic acids, thereby forming a hybridization complex, and comparing hybridization complex formation with complex formation in a normal standard, wherein differences in complex formation indicate differential expression of the nucleic acid in the sample. In one aspect, the sample is from vascular tissue or a patient with a vascular disorder. In another aspect, the nucleic acids of the sample are amplified prior to hybridization.


[0016] The present invention provides a purified protein encoded and produced by a cDNA of the invention. The invention also provides a high-throughput method for using a protein to screen a library or a plurality of molecules or compounds to identify a ligand. The method comprises combining the protein or a portion thereof with the library or plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds the protein. The library or plurality of molecules or compounds is selected from agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA molecules, and ribozymes.


[0017] The invention provides a method for using the protein to produce an antibody which specifically binds the protein. The method for preparing a polyclonal antibody comprises immunizing a animal with protein under conditions to elicit an antibody response, isolating animal antibodies, attaching the protein to a substrate, contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein, dissociating the antibodies from the protein, thereby obtaining purified polyclonal antibodies. The method for preparing a monoclonal antibodies comprises immunizing a animal with a protein under conditions to elicit an antibody response, isolating antibody producing cells from the animal, fusing the antibody producing cells with immortalized cells in culture to form monoclonal antibody producing hybridoma cells, culturing the hybridoma cells, and isolating monoclonal antibodies from culture.


[0018] The invention provides purified antibodies which bind specifically to a protein. The invention also provides a method for using an antibody to detect expression of a protein in a sample, the method comprising combining the antibody with a sample under conditions for formation of antibody:protein complexes, and detecting complex formation, wherein complex formation indicates expression of the protein in the sample. In one aspect, the amount of complex formation when compared to standards is diagnostic of vascular disorders.


[0019] The invention provides a method for immunopurification of a protein comprising attaching an antibody to a substrate, exposing the antibody to a sample containing protein under conditions to allow antibody:protein complexes to form, dissociating the protein from the complex, and collecting purified protein. The invention also provides an array upon which a cDNA encoding a protein, the protein, or an antibody which specifically binds the protein are immobilized. The invention also provides a composition comprising a cDNA, a protein, an antibody, or a ligand which has agonistic or antagonistic activity.



DESCRIPTION OF THE COMPACT DISC-RECORDABLE (CD-R)

[0020] CD-R 1 is labeled: “PA-0049 US, Copy 1,” was created on Apr. 25, 2002 and contains: the Sequence Listing formatted in plain ASCII text. The file for the Sequence Listing is entitled pa0049sl.txt, created on Apr. 25, 2002 and is 716 KB in size.


[0021] CD-R 2 is an exact copy of CD-R 1. CD-R 2 is labeled: “PA-0049 US, Copy 2,” and was created on Apr. 25, 2002.


[0022] The CD-R labeled as: “PA-0049 US, CRF,” contains the Sequence Listing formatted in plain ASCII text. The file for the Sequence Listing is entitled pa0049sl.txt, was created on Apr. 25, 2002 and is 716 KB in size.


[0023] The content of the Sequence Listing named above and as described below, submitted in duplicate on two (2) CD-Rs (labeled “PA-0049 US, Copy 1” and “PA-0049 US, Copy 2”), and the CRF (labeled “PA-0049 US, CRF”) containing the Sequence Listing, are incorporated by reference herein, in their entirety.]



DESCRIPTION OF THE SEQUENCE LISTING AND TABLES

[0024] A portion of the disclosure of this patent document contains material that is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the patent disclosure, as it appears in the Patent and Trademark Office patent file or records, but otherwise reserves all copyright rights whatsoever.


[0025] The Sequence Listing is a compilation of cDNAs obtained by sequencing and extending clone inserts. Each sequence is identified by a sequence identification number (SEQ ID NO) and by the Incyte template number (Incyte ID) from which it was obtained.


[0026] Tables 1, 2, and 3 show the differential expression of clones representing the cDNAs of the present invention. Each clone in Tables 1 and 2 represents a gene expressed at higher levels in microvascular endothelium relative to non-microvascular endothelium. Each clone in Table 3 represents a gene expressed at higher levels in large artery endothelium relative to microvascular and umbilical vein endothelium. Ratios are reported as log base 2; negative values indicate greater expression in the specific endothelial sample relative to the control. In Tables 1 and 3, column 1 shows the Clone ID for the clone present on the microarray and columns 2-10 show the differential expression value for the clone in specific regions of vascular endothelium relative to a vascular endothelium pooled control. In Table 2, column 1 shows the Clone ID for the clone present on the microarray, column 2 shows the average expression of the cDNA in microvascular endothelium and umbilical vein relative to the pooled control, column 3 shows the average expression in large artery endothelium relative to the pooled control, and column 4 shows the difference between microvascular/umbilical vein and large artery expression.


[0027] Table 4 shows the region of each cDNA encompassed by the clone present on a microarray and identified as differentially expressed. Columns 1 and 2 show the SEQ ID NO and Template ID, respectively. Column 3 shows the Clone ID and columns 4 and 5 show the first residue (START) and last residue (STOP) encompassed by the clone on the template.


[0028] Table 5 lists the functional annotation of the cDNAs of the present invention. Columns 1 and 2 show the SEQ ID NO and Template ID, respectively. Columns 3, 4, and 5 show the nearest GenBank homolog (hit; GI Number), probability score (E-value), and functional annotation, respectively, as determined by BLAST analysis (version 2.0 using default parameters; Altschul et al. (1997) Nucleic Acids Res 25:3389-3402; Altschul (1993) J Mol Evol 36:290-300; and Altschul et al. (1990) J Mol Biol 215:403-410) of the cDNA against GenBank (release 122; National Center for Biotechnology Information (NCBI), Bethesda Md.).



DESCRIPTION OF THE INVENTION

[0029] Definitions


[0030] “Antibody” refers to intact immunoglobulin molecule, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab)2 fragment, an Fv fragment; and an antibody-peptide fusion protein.


[0031] “Antigenic determinant” refers to an antigenic or immunogenic epitope, structural feature, or region of an oligopeptide, peptide, or protein which is capable of inducing formation of an antibody which specifically binds the protein. Biological activity is not a prerequisite for immunogenicity.


[0032] “Array” refers to an ordered arrangement of at least two cDNAs, proteins, or antibodies on a substrate. At least one of the cDNAs, proteins, or antibodies represents a control or standard, and the other cDNA, protein, or antibody of diagnostic or therapeutic interest. The arrangement of at least two and up to about 40,000 cDNAs, proteins, or antibodies on the substrate assures that the size and signal intensity of each labeled complex, formed between each cDNA and at least one nucleic acid, each protein and at least one ligand or antibody, or each antibody and at least one protein to which the antibody specifically binds, is individually distinguishable.


[0033] A “combination” comprises at least two and up to about 584 sequences selected from the group consisting of SEQ ID NOs:1-292 and their complements as presented in the Sequence Listing.


[0034] “cDNA” refers to an isolated polynucleotide, nucleic acid, or a fragment thereof, that contains from about 400 to about 12,000 nucleotides. It may have originated recombinantly or synthetically, may be double-stranded or single-stranded, represents coding and noncoding 3′ or 5′ sequence, generally lacks introns and may be purified or combined with carbohydrate, lipids, protein or inorganic elements or substances.


[0035] The phrase “cDNA encoding a protein” refers to a nucleic acid whose sequence closely aligns with sequences that encode conserved regions, motifs or domains identified by employing analyses well known in the art. These analyses include BLAST (Basic Local Alignment Search Tool; Altschul, supra; Altschul (1990) supra) which provides identity within the conserved region. Brenner et al. (1998; Proc Natl Acad Sci 95:6073-6078) who analyzed BLAST for its ability to identify structural homologs by sequence identity found 30% identity is a reliable threshold for sequence alignments of at least 150 residues and 40% is a reasonable threshold for alignments of at least 70 residues (Brenner, page 6076, column 2).


[0036] “Derivative” refers to a cDNA or a protein that has been subjected to a chemical modification. Derivatization of a cDNA can involve substitution of a nontraditional base such as queosine or of an analog such as hypoxanthine. These substitutions are well known in the art. Derivatization of a protein involves the replacement of a hydrogen by an acetyl, acyl, alkyl, amino, formyl, or morpholino group. Derivative molecules retain the biological activities of the naturally occurring molecules but may confer longer lifespan or enhanced activity.


[0037] “Differential expression” refers to an increased or upregulated or a decreased or downregulated expression as detected by absence, presence, or at least two-fold change in the amount of transcribed messenger RNA or translated protein in a sample.


[0038] “Disorder” refers to conditions, diseases or syndromes of the vascular endothelium including disorders of increased vascularization such as atherosclerosis, hemangioma, hemangioendothelioma, diabetic retinopathy, warts, pyogenic granulomas, Kaposi's sarcoma, scar keloids, allergic oedema, neoplasms, follicular cysts, endometriosis, peritoneal sclerosis, and obesity; disorders of insufficient vascularization such as ulcers; and disorders of abnormal remodeling such as psoriasis.


[0039] An “expression profile” is a representation of gene expression in a sample. A nucleic acid expression profile is produced using sequencing, hybridization, or amplification technologies and mRNAs or cDNAs from a sample. A protein expression profile, although time delayed, mirrors the nucleic acid expression profile and uses PAGE, ELISA, FACS, or arrays and labeling moieties or antibodies to detect expression in a sample. The nucleic acids, proteins, or antibodies may be used in solution or attached to a substrate, and their detection is based on methods and labeling moieties well known in the art.


[0040] “Fragment” refers to a chain of consecutive nucleotides from about 60 to about 5000 base pairs in length. Fragments may be used in PCR, hybridization or array technologies to identify related nucleic acids and in binding assays to screen for a ligand. Such ligands are useful as therapeutics to regulate replication, transcription or translation.


[0041] A “hybridization complex” is formed between a cDNA and a nucleic acid of a sample when the purines of one molecule hydrogen bond with the pyrimidines of the complementary molecule, e.g., 5′-A-G-T-C-3′ base pairs with 3′-T-C-A-G-5′. The degree of complementarity and the use of nucleotide analogs affect the efficiency and stringency of hybridization reactions.


[0042] “Identity” as applied to sequences, refers to the quantification (usually percentage) of nucleotide or residue matches between at least two sequences aligned using a standardized algorithm such as Smith-Waterman alignment (Smith and Waterman (1981) J Mol Biol 147:195-197), CLUSTALW (Thompson et al. (1994) Nucleic Acids Res 22:4673-4680), or BLAST2 (Altschul (1997) supra). BLAST2 may be used in a standardized and reproducible way to insert gaps in one of the sequences in order to optimize alignment and to achieve a more meaningful comparison between them. “Similarity” as applied to proteins uses the same algorithms but takes into account conservative substitutions of nucleotides or residues.


[0043] “Isolated” or “purified” refers to any molecule or compound that is separated from its natural environment and is from about 60% free to about 90% free from other components with which it is naturally associated.


[0044] “Labeling moiety” refers to any reporter molecule whether a visible or radioactive label, stain or dye that can be attached to or incorporated into a cDNA or protein. Visible labels and dyes include but are not limited to anthocyanins, β glucuronidase, BIODIPY, Coomassie blue, Cy3 and Cy5, digoxigenin, FITC, green fluorescent protein, luciferase, spyro red, silver, and the like. Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like.


[0045] “Large artery endothelium” includes vascular endothelium obtained from aorta, coronary artery, pulmonary artery, and iliac artery.


[0046] “Ligand” refers to any agent, molecule, or compound which will bind specifically to a complementary site on a cDNA molecule or polynucleotide, or to an epitope or a protein. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic or organic substances including nucleic acids, proteins, carbohydrates, fats, and lipids.


[0047] “Microvascular endothelium” includes vascular endothelium obtained from neonatal dermis and uterine myometrium.


[0048] “Neoplasms” may include adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, colon, esophagus, gall bladder, ganglia, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, rectum, salivary glands, skin, small intestine, spleen, stomach, testis, thymus, thyroid, and uterus.


[0049] “Non-microvascular endothelium” includes large artery endothelium and vascular endothelium obtained from umbilical artery and vein.


[0050] “Oligonucleotide” refers a single stranded molecule from about 18 to about 60 nucleotides in length which may be used in hybridization or amplification technologies or in regulation of replication, transcription or translation. Equivalent terms include amplimer, primer, and oligomer.


[0051] “Portion” refers to any part of a protein used for any purpose which retains at least one biological or antigenic characteristic of a native protein, but especially, to an epitope for the screening of ligands or for production of antibodies.


[0052] “Post-translational modification” of a protein can involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and the like. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cellular location, cell type, pH, enzymatic milieu, and the like.


[0053] “Probe” refers to a cDNA that hybridizes to at least one nucleic acid in a sample. Where targets are single stranded, probes are complementary single strands. Probes can be labeled for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.


[0054] “Protein” refers to a polypeptide or any portion thereof. An “oligopeptide” is an amino acid sequence from about five residues to about 15 residues that is used as part of a fusion protein to produce an antibody.


[0055] “Sample” is used in its broadest sense as containing nucleic acids, proteins, antibodies, and the like. A sample may comprise a bodily fluid such as ascites, blood, lymph, saliva, semen, spinal, sputum, tears, and urine; the soluble fraction of a cell preparation, or an aliquot of media in which cells were grown; a chromosome, an organelle, or membrane isolated or extracted from a cell; genomic DNA, RNA, or cDNA in solution or bound to a substrate; a cell; a tissue or tissue biopsy; a tissue print; buccal cells, skin, a hair or its follicle; and the like.


[0056] “Specific binding” refers to a special and precise interaction between two molecules which is dependent upon their structure, particularly their molecular side groups. For example, the intercalation of a regulatory protein into the major groove of a DNA molecule, the hydrogen bonding along the backbone between two single stranded nucleic acids, or the binding between an epitope of a protein and an agonist, antagonist, or antibody.


[0057] “Substrate” refers to any rigid or semi-rigid support to which cDNAs or proteins are bound and includes membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, capillaries or other tubing, plates, polymers, and microparticles with a variety of surface forms including wells, trenches, pins, channels and pores.


[0058] A “transcript image” (TI) is a profile of gene transcription activity in a particular tissue at a particular time. TI provides assessment of the relative abundance of expressed polynucleotides in the cDNA libraries of an EST database as described in U.S. Pat. No. 5,840,484, incorporated herein by reference.


[0059] “Variant” refers to molecules that are recognized variations of a cDNA or a protein encoded by the cDNA. Splice variants may be determined by BLAST score, wherein the score is at least 100, and most preferably at least 400. Allelic variants have a high percent identity to the cDNAs and may differ by about three bases per hundred bases. “Single nucleotide polymorphism” (SNP) refers to a change in a single base as a result of a substitution, insertion or deletion. The change may be conservative (purine for purine) or non-conservative (purine to pyrimidine) and may or may not result in a change in an encoded amino acid.


[0060] The Invention


[0061] The present invention provides for a combination comprising a plurality of cDNAs and their complements, SEQ ID NOs:1-292, which may be used to diagnose, to stage, to treat, or to monitor the progression or treatment of a vascular disorder. These cDNAs represent known and novel genes differentially expressed in vascular endothelium from microvascular and non-microvascular tissues. The combination may be used in its entirety or in part, as subsets of cDNAs expressed at higher levels in microvascular endothelium, SEQ ID NOs:1-209, or of cDNAs expressed at higher levels in large artery endothelium, SEQ ID NOs:210-292. SEQ ID NOs:36, 51, 61, 83, 87, 110, 111, 136, 159, 198, and 244 represent novel cDNAs differentially expressed in vascular endothelium. Since the novel cDNAs were identified solely by their differential expression, it is not essential to know a priori the name, structure, or 40 function of the gene or its encoded protein. The usefulness of the novel cDNAs exists in their immediate value as diagnostics for vascular disorders.


[0062] Tables 1, 2, and 3 show the differential expression of clones representing the cDNAs of the present invention on microarrays. Each clone in Tables 1 and 2 represents a gene expressed at higher levels in microvascular endothelium relative to non-microvascular endothelium. Each clone in Table 3 represents a gene expressed at higher levels in large artery endothelium relative to microvascular endothelium and umbilical vein. Ratios are reported as log base 2; negative values indicate greater expression in the specific endothelial sample relative to the control.


[0063] In Tables 1 and 3, column 1 shows the clone ID and columns 2-10 show the differential expression value for the clone in specific regions of vascular endothelium relative to a vascular endothelium pooled control. The vascular endothelium represented in columns 2-10 are aorta (HAEC), coronary artery (HCAEC), pulmonary artery (HPAEC), iliac artery (HIAEC), uterine myometrium microvasculature (UtMVEC), neonatal dermal microvasculature (HMVECD), umbilical artery (HUAEC), and umbilical vein (HUVEC; three separate samples), respectively. In Table 2, column 1 shows the Clone ID for the clone present on the microarray, column 2 shows the average expression of the cDNA in microvascular and umbilical vein endothelium relative to the pooled control, column 3 shows the average expression in large artery endothelium relative to the pooled control, and column 4 shows the difference between microvascular/umbilical vein and large artery expression.


[0064] Table 4 shows the region of each cDNA encompassed by the clone present on a microarray and identified as differentially expressed. Columns 1 and 2 show the SEQ ID NO and template ID, respectively. Column 3 shows the clone ID and columns 4 and 5 show the first residue (START) and last residue (STOP) encompassed by the clone on the template.


[0065] Table 5 lists the functional annotation of the cDNAs of the present invention. Columns 1 and 2 show the SEQ ID NO and template ID, respectively. Columns 3, 4, and 5 show the GenBank homolog (hit), probability score (E-value), and functional annotation, respectively, as determined by BLAST analysis (version 2.0 using default parameters; Altschul (1997) supra) of the cDNA against GenBank (release 122; NCBI).


[0066] The cDNAs of the invention define a differential expression pattern against which to compare the expression pattern of biopsied and/or in vitro treated vascular tissues. Experimentally, differential expression of the cDNAs can be evaluated by methods including, but not limited to, differential display by spatial immobilization or by gel electrophoresis, genome mismatch scanning, representational discriminant analysis, clustering, transcript imaging and array technologies. These methods may be used alone or in combination.


[0067] The combination may be arranged on a substrate and hybridized with tissues from subjects with diagnosed vascular disorders to identify those sequences which are differentially expressed in, e.g., both atherosclerosis and other vascular disorders. This allows identification of those sequences of highest diagnostic and potential therapeutic value. In one embodiment, an additional set of cDNAs, such as cDNAs encoding signaling molecules, are arranged on the substrate with the combination. Such combinations may be useful in the elucidation of pathways which are affected in a particular disorder or to identify new, coexpressed, candidate, therapeutic molecules.


[0068] In another embodiment, the combination can be used for large scale genetic or gene expression analysis of a large number of novel, nucleic acids. These samples are prepared by methods well known in the art and are from mammalian cells or tissues which are in a certain stage of development; have been treated with a known molecule or compound, such as a cytokine, growth factor, a drug, and the like; or have been extracted or biopsied from a mammal with a known or unknown condition, disorder, or disease before or after treatment. The sample nucleic acids are hybridized to the combination for the purpose of defining a novel gene profile associated with that developmental stage, treatment, or disorder.


[0069] cDNAs and Their Uses


[0070] cDNAs can be prepared by a variety of synthetic or enzymatic methods well known in the art. cDNAs can be synthesized, in whole or in part, using chemical methods well known in the art (Caruthers et al. (1980) Nucleic Acids Symp Ser (7):215-233). Alternatively, cDNAs can be produced enzymatically or recombinantly, by in vitro or in vivo transcription.


[0071] Nucleotide analogs can be incorporated into cDNAs by methods well known in the art. The only requirement is that the incorporated analog must base pair with native purines or pyrimidines. For example, 2, 6-diaminopurine can substitute for adenine and form stronger bonds with thymidine than those between adenine and thymidine. A weaker pair is formed when hypoxanthine is substituted for guanine and base pairs with cytosine. Additionally, cDNAs can include nucleotides that have been derivatized chemically or enzymatically.


[0072] cDNAs can be synthesized on a substrate. Synthesis on the surface of a substrate may be accomplished using a chemical coupling procedure and a piezoelectric printing apparatus as described by Baldeschweiler et al. (PCT publication WO95/251116). Alternatively, the cDNAs can be synthesized on a substrate surface using a self-addressable electronic device that controls when reagents are added as described by Heller et al. (U.S. Pat. No. 5,605,662). cDNAs can be synthesized directly on a substrate by sequentially dispensing reagents for their synthesis on the substrate surface or by dispensing preformed DNA fragments to the substrate surface. Typical dispensers include a micropipette delivering solution to the substrate with a robotic system to control the position of the micropipette with respect to the substrate. There can be a multiplicity of dispensers so that reagents can be delivered to the reaction regions efficiently.


[0073] cDNAs can be immobilized on a substrate by covalent means such as by chemical bonding procedures or UV irradiation. In one method, a cDNA is bound to a glass surface which has been modified to contain epoxide or aldehyde groups. In another method, a cDNA is placed on a polylysine coated surface and UV cross-linked to it as described by Shalon et al. (WO95/35505). In yet another method, a cDNA is actively transported from a solution to a given position on a substrate by electrical means (Heller, supra). cDNAs do not have to be directly bound to the substrate, but rather can be bound to the substrate through a linker group. The linker groups are typically about 6 to 50 atoms long to provide exposure of the attached cDNA. Preferred linker groups include ethylene glycol oligomers, diamines, diacids and the like. Reactive groups on the substrate surface react with a terminal group of the linker to bind the linker to the substrate. The other terminus of the linker is then bound to the cDNA. Alternatively, polynucleotides, plasmids or cells can be arranged on a filter. In the latter case, cells are lysed, proteins and cellular components degraded, and the DNA is coupled to the filter by UV cross-linking.


[0074] The cDNAs may be used for a variety of purposes. For example, the combination of the invention may be used on an array. The array, in turn, can be used in high-throughput methods for detecting a related polynucleotide in a sample, screening a plurality of molecules or compounds to identify a ligand, diagnosing atherosclerosis, or inhibiting or inactivating a therapeutically relevant gene related to the cDNA.


[0075] When the cDNAs of the invention are employed on a microarray, the cDNAs are arranged in an ordered fashion so that each cDNA is present at a specified location. Because the cDNAs are at specified locations on the substrate, the hybridization patterns and intensities, which together create a unique expression profile, can be interpreted in terms of expression levels of particular genes and can be correlated with a particular metabolic process, condition, disorder, disease, stage of disease, or treatment.


[0076] Hybridization


[0077] The cDNAs or fragments or complements thereof may be used in various hybridization technologies. The cDNAs may be labeled using a variety of reporter molecules by either PCR, recombinant, or enzymatic techniques. For example, a commercially available vector containing the cDNA is transcribed in the presence of an appropriate polymerase, such as T7 or SP6 polymerase, and at least one labeled nucleotide. Commercial kits are available for labeling and cleanup of such cDNAs. Radioactive (Amersham Bisociences (APB), Piscataway N.J.), fluorescent (Qiagen-Operon, Alameda Calif.), and chemiluminescent labeling (Promega, Madison Wis.) are well known in the art.


[0078] A cDNA may represent the complete coding region of an mRNA or be designed or derived from unique regions of the mRNA or genomic molecule, an intron, a 3′ untranslated region, or from a conserved motif. The cDNA is at least 18 contiguous nucleotides in length and is usually single stranded. Such a cDNA may be used under hybridization conditions that allow binding only to an identical sequence, a naturally occurring molecule encoding the same protein, or an allelic variant. Discovery of related human and mammalian sequences may also be accomplished using a pool of degenerate cDNAs and appropriate hybridization conditions. Generally, a cDNA for use in Southern or northern hybridizations may be from about 400 to about 6000 nucleotides long. Such cDNAs have high binding specificity in solution-based or substrate-based hybridizations. An oligonucleotide, a fragment of the cDNA, may be used to detect a polynucleotide in a sample using PCR.


[0079] The stringency of hybridization is determined by G+C content of the cDNA, salt concentration, and temperature. In particular, stringency is increased by reducing the concentration of salt or raising the hybridization temperature. In solutions used for some membrane based hybridizations, addition of an organic solvent such as formamide allows the reaction to occur at a lower temperature. Hybridization may be performed with buffers, such as 5×saline sodium citrate (SSC) with 1% sodium dodecyl sulfate (SDS) at 60° C., that permit the formation of a hybridization complex between nucleic acid sequences that contain some mismatches. Subsequent washes are performed with buffers such as 0.2×SSC with 0.1% SDS at either 45° C. (medium stringency) or 65°-68° C. (high stringency). At high stringency, hybridization complexes will remain stable only where the nucleic acids are completely complementary. In some membrane-based hybridizations, preferably 35% or most preferably 50%, formamide may be added to the hybridization solution to reduce the temperature at which hybridization is performed. Background signals may be reduced by the use of detergents such as Sarkosyl or TRITON X-100 (Sigma-Aldrich, St. Louis Mo.) and a blocking agent such as denatured salmon sperm DNA. Selection of components and conditions for hybridization are well known to those skilled in the art and are reviewed in Ausubel et al. (1997, Short Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., Units 2.8-2.11, 3.18-3.19 and 4-6-4.9).


[0080] Dot-blot, slot-blot, low density and high density arrays are prepared and analyzed using methods known in the art. cDNAs from about 18 consecutive nucleotides to about 5000 consecutive nucleotides in length are contemplated by the invention and used in array technologies. The preferred number of cDNAs on an array is at least about 100,000, a more preferred number is at least about 40,000, an even more preferred number is at least about 10,000, and a most preferred number is at least about 600 to about 800. The array may be used to monitor the expression level of large numbers of genes simultaneously and to identify genetic variants, mutations, and SNPs. Such information may be used to determine gene function; to understand the genetic basis of a disorder; to diagnose a disorder; and to develop and monitor the activities of therapeutic agents being used to control or cure a disorder. (See, e.g., U.S. Pat. No. 5,474,796; WO95/11995; WO95/35505; U.S. Pat. No. 5,605,662; and U.S. Pat. No. 5,958,342.)


[0081] Screening and Purification Assays


[0082] A cDNA may be used to screen a library or a plurality of molecules or compounds for a ligand which specifically binds the cDNA. Ligands may be DNA molecules, RNA molecules, peptide nucleic acid molecules, peptides, proteins such as transcription factors, promoters, enhancers, repressors, and other proteins that regulate replication, transcription, or translation of the polynucleotide in the biological system. The assay involves combining the cDNA or a fragment thereof with the molecules or compounds under conditions that allow specific binding and detecting the bound cDNA to identify at least one ligand that specifically binds the cDNA.


[0083] In one embodiment, the cDNA may be incubated with a library of isolated and purified molecules or compounds and binding activity determined by methods such as a gel-retardation assay (U.S. Pat. No. 6,010,849) or a reticulocyte lysate transcriptional assay. In another embodiment, the cDNA may be incubated with nuclear extracts from biopsied and/or cultured cells and tissues. Specific binding between the cDNA and a molecule or compound in the nuclear extract is initially determined by gel shift assay. Protein binding may be confirmed by raising antibodies against the protein and adding the antibodies to the gel-retardation assay where specific binding will cause a supershift in the assay.


[0084] In another embodiment, the cDNA may be used to purify a molecule or compound using affinity chromatography methods well known in the art. In one embodiment, the cDNA is chemically reacted with cyanogen bromide groups on a polymeric resin or gel. Then a sample is passed over and reacts with or binds to the cDNA. The molecule or compound which is bound to the cDNA may be released from the cDNA by increasing the salt concentration of the flow-through medium and collected.


[0085] The cDNA may be used to purify a ligand from a sample. A method for using a cDNA to purify a ligand would involve combining the cDNA or a fragment thereof with a sample under conditions to allow specific binding, recovering the bound cDNA, and using an appropriate agent to separate the cDNA from the purified ligand.


[0086] Protein Production and Uses


[0087] The full length cDNAs or fragment thereof may be used to produce purified proteins using recombinant DNA technologies described herein and taught in Ausubel (supra; Units 16.1-16.62). One of the advantages of producing proteins by these procedures is the ability to obtain highly-enriched sources of the proteins thereby simplifying purification procedures.


[0088] The proteins may contain amino acid substitutions, deletions or insertions made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. Such substitutions may be conservative in nature when the substituted residue has structural or chemical properties similar to the original residue (e.g., replacement of leucine with isoleucine or valine) or they may be nonconservative when the replacement residue is radically different (e.g., a glycine replaced by a tryptophan). Computer programs included in LASERGENE software (DNASTAR, Madison Wis.) and algorithms included in RasMol software (University of Massachusetts, Amherst Mass.) may be used to help determine which and how many amino acid residues in a particular portion of the protein may be substituted, inserted, or deleted without abolishing biological or immunological activity.


[0089] Expression of Encoded Proteins


[0090] Expression of a particular cDNA may be accomplished by cloning the cDNA into a vector and transforming this vector into a host cell. The cloning vector used for the construction of cDNA libraries in the LIFESEQ databases (Incyte Genomics, Palo Alto Calif.) may also be used for expression. Such vectors usually contain a promoter and a polylinker useful for cloning, priming, and transcription. An exemplary vector may also contain the promoter for β-galactosidase, an amino-terminal methionine and the subsequent seven amino acid residues of β-galactosidase. The vector may be transformed into competent E. coli cells. Induction of the isolated bacterial strain with isopropylthiogalactoside (IPTG) using standard methods will produce a fusion protein that contains an N terminal methionine, the first seven residues of β-galactosidase, about 15 residues of linker, and the protein encoded by the cDNA.


[0091] The cDNA may be shuttled into other vectors known to be useful for expression of protein in specific hosts. Oligonucleotides containing cloning sites and fragments of DNA sufficient to hybridize to stretches at both ends of the cDNA may be chemically synthesized by standard methods. These primers may then be used to amplify the desired fragments by PCR. The fragments may be digested with appropriate restriction enzymes under standard conditions and isolated using gel electrophoresis. Alternatively, similar fragments are produced by digestion of the cDNA with appropriate restriction enzymes and filled in with chemically synthesized oligonucleotides. Fragments of the coding sequence from more than one gene may be ligated together and expressed.


[0092] Signal sequences that dictate secretion of soluble proteins are particularly desirable as component parts of a recombinant sequence. For example, a chimeric protein may be expressed that includes one or more additional purification-facilitating domains. Such domains include, but are not limited to, metal-chelating domains that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex, Seattle Wash.). The inclusion of a cleavable-linker sequence such as ENTEROKINASEMAX (Invitrogen, San Diego Calif.) between the protein and the purification domain may also be used to recover the protein.


[0093] Suitable host cells may include, but are not limited to, mammalian cells such as Chinese Hamster Ovary (CHO) and human 293 cells, insect cells such as Sf9 cells, plant cells such as Nicotiana tabacum, yeast cells such as Saccharomyces cerevisiae, and bacteria such as E. coli. For each of these cell systems, a useful vector may also include an origin of replication and one or two selectable markers to allow selection in bacteria as well as in a transformed eukaryotic host. Vectors for use in eukaryotic host cells may require the addition of 3′ poly(A) tail if the cDNA lacks poly(A).


[0094] Additionally, the vector may contain promoters or enhancers that increase gene expression. Many promoters are known and used in the art. Most promoters are host specific and exemplary promoters includes SV40 promoters for CHO cells; T7 promoters for bacterial hosts; viral promoters and enhancers for plant cells; and PGH promoters for yeast. Adenoviral vectors with the rous sarcoma virus enhancer or retroviral vectors with long terminal repeat promoters may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of secreted soluble protein may be recovered from the conditioned medium and analyzed using chromatographic methods well known in the art. An alternative method for the production of large amounts of secreted protein involves the transformation of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, and the like.


[0095] In addition to recombinant production, proteins or portions thereof may be produced manually, using solid-phase techniques (Stewart et al. (1969) Solid-Phase Peptide Synthesis, W H Freeman, San Francisco Calif.; Merrifield (1963) J Am Chem Soc 5:2149-2154), or using machines such as the 431A peptide synthesizer (Applied Biosystems (ABI), Foster City Calif.). Proteins produced by any of the above methods may be used as pharmaceutical compositions to treat disorders associated with null or inadequate expression of the genomic sequence.


[0096] Screening and Purification Assays


[0097] A protein or a portion thereof encoded by the cDNA may be used to screen a library or a plurality of molecules or compounds for a ligand with specific binding affinity or to purify a molecule or compound from a sample. The protein or portion thereof employed in such screening may be free in solution, affixed to an abiotic or biotic substrate, or located intracellularly. For example, viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a protein on their cell surface can be used in screening assays. The cells are screened against a library or a plurality of ligands and the specificity of binding or formation of complexes between the expressed protein and the ligand may be measured. The ligands may be agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA molecules, ribozymes or any other test molecule or compound that specifically binds the protein. An exemplary assay involves combining the mammalian protein or a portion thereof with the molecules or compounds under conditions that allow specific binding and detecting the bound protein to identify at least one ligand that specifically binds the protein.


[0098] This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding the protein specifically compete with a test compound capable of binding to the protein or oligopeptide or fragment thereof. One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in U.S. Pat. No. 5,876,946. Molecules or compounds identified by screening may be used in a model system to evaluate their toxicity, diagnostic, or therapeutic potential.


[0099] The protein may be used to purify a ligand from a sample. A method for using a protein to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified ligand.


[0100] Production of Antibodies


[0101] A protein encoded by a cDNA of the invention may be used to produce specific antibodies. Antibodies may be produced using an oligopeptide or a portion of the protein with inherent immunological activity. Methods for producing antibodies include: 1) injecting an animal, usually goats, rabbits, or mice, with the protein, or an amtigenic determinant or an oligopeptide thereof, to induce an immune response; 2) engineering hybridomas to produce monoclonal antibodies; 3) inducing in vivo production in the lymphocyte population; or 4) screening libraries of recombinant immunoglobulins. Recombinant immunoglobulins may be produced as taught in U.S. Pat. No. 4,816,567.


[0102] Antibodies produced using the proteins of the invention are useful for the diagnosis of prepathologic disorders as well as the diagnosis of chronic or acute diseases characterized by abnormalities in the expression, amount, or distribution of the protein. A variety of protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies specific for proteins are well known in the art. Immunoassays typically involve the formation of complexes between a protein and its specific binding molecule or compound and the measurement of complex formation. Immunoassays may employ a two-site, monoclonal-based assay that utilizes monoclonal antibodies reactive to two noninterfering epitopes on a specific protein or a competitive binding assay (Pound (1998) Immunochemical Protocols, Humana Press, Totowa N.J.).


[0103] Immunoassay procedures may be used to quantify expression of the protein in cell cultures, in subjects with a particular disorder or in model animal systems under various conditions. Increased or decreased production of proteins as monitored by immunoassay may contribute to knowledge of the cellular activities associated with developmental pathways, engineered conditions or diseases, or treatment efficacy. The quantity of a given protein in a given tissue may be determined by performing immunoassays on freeze-thawed detergent extracts of biological samples and comparing the slope of the binding curves to binding curves generated by purified protein.


[0104] Antibody Arrays


[0105] In an alternative to yeast two hybrid system analysis of proteins, an antibody array can be used to study protein-protein interactions and phosphorylation. A variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until protein:antibody complexes are formed. Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest. In the alternative, a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti-DIG antibody which reveals where the protein of interest forms a complex. The identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.


[0106] Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt et al. (2000) Nat Biotechnol 18:989-94).


[0107] Labeling of Molecules for Assay


[0108] A wide variety of reporter molecules and conjugation techniques are known by those skilled in the art and may be used in various cDNA, polynucleotide, protein, peptide or antibody assays. Synthesis of labeled molecules may be achieved using commercial kits for incorporation of a labeled nucleotide such as 32P-dCTP, Cy3-dCTP or Cy5-dCTP or amino acid such as 35S-methionine. Polynucleotides, cDNAs, proteins, or antibodies may be directly labeled with a reporter molecule by chemical conjugation to amines, thiols and other groups present in the molecules using reagents such as BIODIPY or FITC (Molecular Probes, Eugene Oreg.).


[0109] The proteins and antibodies may be labeled for purposes of assay by joining them, either covalently or noncovalently, with a reporter molecule that provides for a detectable signal. A wide variety of labels and conjugation techniques are known and have been reported in the scientific and patent literature including, but not limited to U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.


[0110] Diagnostics


[0111] The cDNAs, or fragments thereof, may be used to detect and quantify differential gene expression; absence, presence, or excess expression of mRNAs; or to monitor mRNA levels during therapeutic intervention. Disorders associated with altered expression include disorders of increased vascularization such as atherosclerosis, hemangioma, hemangioendothelioma, warts, pyogenic granulomas, Kaposi's sarcoma, scar keloids, allergic oedema, neoplasms, follicular cysts, endometriosis, peritoneal sclerosis, diabetic retinopathy, and obesity; disorders of insufficient vascularization such as ulcers; and disorders of abnormal remodeling such as psoriasis. These cDNAs can also be utilized as markers of treatment efficacy against the disorders noted above and other disorders, conditions, and diseases over a period ranging from several days to months. The diagnostic assay may use hybridization or amplification technology to compare gene expression in a biological sample from a patient to standard samples in order to detect altered gene expression. Qualitative or quantitative methods for this comparison are well known in the art.


[0112] For example, the cDNA may be labeled by standard methods and added to a biological sample from a patient under conditions for hybridization complex formation. After an incubation period, the sample is washed and the amount of label (or signal) associated with hybridization complexes is quantified and compared with a standard value. If the amount of label in the patient sample is significantly altered in comparison to the standard value, then the presence of the associated condition, disease or disorder is indicated.


[0113] In order to provide a basis for the diagnosis of a condition, disease or disorder associated with gene expression, a normal or standard expression profile is established. This may be accomplished by combining a biological sample taken from normal subjects, either animal or human, with a probe under conditions for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained using normal subjects with values from an experiment in which a known amount of a purified target sequence is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a particular condition, disease, or disorder. Deviation from standard values toward those associated with a particular condition is used to diagnose that condition.


[0114] Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies and in clinical trial or to monitor the treatment of an individual patient. Once the presence of a condition is established and a treatment protocol is initiated, diagnostic assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in a normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.


[0115] Gene Expression Profiles


[0116] A gene expression profile comprises a plurality of cDNAs and a plurality of detectable hybridization complexes, wherein each complex is formed by hybridization of one or more probes to one or more complementary nucleic acids in a sample. The cDNAs of the invention are used as elements on a array to analyze gene expression profiles. In one embodiment, the array is used to monitor the progression of disease. Researchers and clinicians can catalog the differences in gene expression between healthy and diseased tissues or cells. By analyzing changes in patterns of gene expression, disease can be diagnosed at earlier stages before the patient is symptomatic. The invention can be used to formulate a prognosis and to design a treatment regimen. The invention can also be used to monitor the efficacy of treatment. For treatments with known side effects, the array is employed to improve the treatment regimen. A dosage is established that causes a change in genetic expression patterns indicative of successful treatment. Expression patterns associated with the onset of undesirable side effects are avoided. This approach may be more sensitive and rapid than waiting for the patient to show inadequate improvement, or to manifest side effects, before altering the course of treatment.


[0117] Experimentally, expression profiles can also be evaluated by methods including, but not limited to, differential display by spatial immobilization or by gel electrophoresis, genome mismatch scanning, representational discriminant analysis, transcript imaging, and by protein or antibody arrays. Expression profiles produced by these methods may be used alone or in combination. The correspondence between mRNA and protein expression has been discussed by Zweiger (2001, Transducing the Genome. McGraw-Hill, San Francisco, Calif.) and Glavas et al. (2001; T cell activation upregulates cyclic nucleotide phosphodiesterases 8A1 and 7A3, Proc Natl Acad Sci 98:6319-6342) among others.


[0118] In another embodiment, animal models which mimic a human disease can be used to characterize expression profiles associated with a particular condition, disorder or disease; or treatment of the condition, disorder or disease. Novel treatment regimens may be tested in these animal models using arrays to establish and then follow expression profiles over time. In addition, arrays may be used with cell cultures or tissues removed from animal models to rapidly screen large numbers of candidate drug molecules, looking for ones that produce an expression profile similar to those of known therapeutic drugs, with the expectation that molecules with the same expression profile will likely have similar therapeutic effects. Thus, the invention provides the means to rapidly determine the molecular mode of action of a drug.


[0119] Assays Using Antibodies


[0120] Antibodies directed against epitopes on a protein encoded by a cDNA of the invention may be used in assays to quantify the amount of protein found in a particular human cell. Such assays include methods utilizing the antibody and a label to detect expression level under normal or disease conditions.


[0121] The antibodies may be used with or without modification, and labeled by joining them, either covalently or noncovalently, with a labeling moiety.


[0122] Protocols for detecting and measuring protein expression using either polyclonal or monoclonal antibodies are well known in the art. Examples include ELISA, RIA, fluorescent activated cell sorting (FACS) and arrays. Such immunoassays typically involve the formation of complexes between the protein and its specific antibody and the measurement of such complexes. These and other assays are described in Pound (supra).


[0123] Therapeutics


[0124] The cDNAs and fragments thereof can be used in gene therapy. cDNAs can be delivered ex vivo to target cells, such as cells of bone marrow. Once stable integration and transcription and or translation are confirmed, the bone marrow may be reintroduced into the subject. Expression of the protein encoded by the cDNA may correct a disorder associated with mutation of a normal sequence, reduction or loss of an endogenous target protein, or overepression of an endogenous or mutant protein. Alternatively, cDNAs may be delivered in vivo using vectors such as retrovirus, adenovirus, adeno-associated virus, herpes simplex virus, and bacterial plasmids. Non-viral methods of gene delivery include cationic liposomes, polylysine conjugates, artificial viral envelopes, and direct injection of DNA (Anderson (1998) Nature 392:25-30; Dachs et al. (1997) Oncol Res 9:313-325; Chu et al. (1998) J Mol Med 76(3-4):184-192; Weiss et al. (1999) Cell Mol Life Sci 55(3):334-358; Agrawal (1996) Antisense Therapeutics, Humana Press, Totowa N.J.; and August et al. (1997) Gene Therapy (Advances in Pharmacolo, Vol. 40), Academic Press, San Diego Calif.).


[0125] In addition, expression of a particular protein can be regulated through the specific binding of a fragment of a cDNA to a genomic sequence or an mRNA which encodes the protein or directs its transcription or translation. The cDNA can be modified or derivatized to any RNA-like or DNA-like 5 material including peptide nucleic acids, branched nucleic acids, and the like. These sequences can be produced biologically by transforming an appropriate host cell with a vector containing the sequence of interest.


[0126] Molecules which regulate the activity of the cDNA or encoded protein are useful as therapeutics for vascular disorders such as atherosclerosis, diabetic retinopathy, neoplasms, and obesity. Such molecules include agonists which increase the expression or activity of the polynucleotide or encoded protein, respectively; or antagonists which decrease expression or activity of the polynucleotide or encoded protein, respectively. In one aspect, an antibody which specifically binds the protein may be used directly as an antagonist or indirectly as a delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express the protein. Additionally, any of the proteins, or their ligands, or complementary nucleic acid sequences may be administered as pharmaceutical compositions or in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to affect the treatment or prevention of the conditions and disorders associated with an immune response. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects. Further, the therapeutic agents may be combined with pharmaceutically-acceptable carriers including excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration used by doctors and pharmacists may be found in the latest edition of Remington's Pharmaceutical Sciences (Mack Publishing, Easton Pa.).


[0127] Model Systems


[0128] Animal models may be used as bioassays where they exhibit a phenotypic response similar to that of humans and where exposure conditions are relevant to human exposures. Mammals are the most common models, and most infectious agent, cancer, drug, and toxicity studies are performed on rodents such as rats or mice because of low cost, availability, lifespan, reproductive potential, and abundant reference literature. Inbred and outbred rodent strains provide a convenient model for investigation of the physiological consequences of underexpression or overexpression of genes of interest and for the development of methods for diagnosis and treatment of diseases. A mammal inbred to overexpress a particular gene (for example, secreted in milk) may also serve as a convenient source of the protein expressed by that gene.


[0129] Transgenic Animal Models


[0130] Transgenic rodents that overexpress or underexpress a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents. (See, e.g., U.S. Pat. No. 5,175,383 and U.S. Pat. No. 5,767,337.) In some cases, the introduced gene may be activated at a specific time in a specific tissue type during fetal or postnatal development. Expression of the transgene is monitored by analysis of phenotype, of tissue-specific mRNA expression, or of serum and tissue protein levels in transgenic animals before, during, and after challenge with experimental drug therapies.


[0131] Embryonic Stem Cells


[0132] Embryonic (ES) stem cells isolated from rodent embryos retain the potential to form embryonic tissues. When ES cells such as the mouse 129/SvJ cell line are placed in a blastocyst from the C57BL/6 mouse strain, they resume normal development and contribute to tissues of the live-born animal. ES cells are preferred for use in the creation of experimental knockout and knockin animals. The method for this process is well known in the art and the steps are: the cDNA is introduced into a vector, the vector is transformed into ES cells, transformed cells are identified and microinjected into mouse cell blastocysts, blastocysts are surgically transferred to pseudopregnant dams. The resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains.


[0133] Knockout Analysis


[0134] In gene knockout analysis, a region of a gene is enzymatically modified to include a non-natural intervening sequence such as the neomycin phosphotransferase gene (neo; Capecchi (1989) Science 244:1288-1292). The modified gene is transformed into cultured ES cells and integrates into the endogenous genome by homologous recombination. The inserted sequence disrupts transcription and translation of the endogenous gene.


[0135] Knockin Analysis


[0136] ES cells can be used to create knockin humanized animals or transgenic animal models of human diseases. With knockin technology, a region of a human gene is injected into animal ES cells, and the human sequence integrates into the animal cell genome. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on the progression and treatment of the analogous human condition.


[0137] As described herein, the uses of the cDNAs, provided in the Sequence Listing of this application, and their encoded proteins are exemplary of known techniques and are not intended to reflect any limitation on their use in any technique that would be known to the person of average skill in the art.


[0138] Furthermore, the cDNAs provided in this application may be used in molecular biology techniques that have not yet been developed, provided the new techniques rely on properties of nucleotide sequences that are currently known to the person of ordinary skill in the art, e.g., the triplet genetic code, specific base pair interactions, and the like. Likewise, reference to a method may include combining more than one method for obtaining or assembling full length cDNA sequences that will be known to those skilled in the art. It is also to be understood that this invention is not limited to the particular methodology, protocols, and reagents described, as these may vary. It is also understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. The examples below are provided to illustrate the subject invention and are not included for the purpose of limiting the invention.







EXAMPLES

[0139] I Construction of cDNA Libraries


[0140] RNA was purchased from Clontech Laboratories (Palo Alto Calif.) or isolated from various tissues. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL reagent (Invitrogen). The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated with either isopropanol or ethanol and sodium acetate, or by other routine methods.


[0141] Phenol extraction and precipitation of RNA were repeated as necessary to increase RNA purity. In most cases, RNA was treated with DNAse. For most libraries, poly(A) RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (Qiagen, Valencia Calif.), or an OLIGOTEX mRNA purification kit (Qiagen). Alternatively, poly(A) RNA was isolated directly from tissue lysates using other kits, including the POLY(A)PURE mRNA purification kit (Ambion, Austin Tex.).


[0142] In some cases, Stratagene (La Jolla Calif.) was provided with RNA and constructed the corresponding cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen) using the recommended procedures or similar methods known in the art. (See Ausubel, supra, Units 5.1 through 6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (APB) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of the pBLUESCRIPT phagemid (Stratagene), pSPORT1 plasmid (Invitrogen), or pINCY plasmid (Incyte Genomics). Recombinant plasmids were transformed into XL1-BLUE, XL1-BLUEMRF, or SOLR competent E. coli cells (Stratagene) or DH5a, DH10B, or ELECTROMAX DH10B competent E. coli cells (Invitrogen).


[0143] In some cases, libraries were superinfected with a 5×excess of the helper phage, M13K07, according to the method of Vieira et al. (1987, Methods Enzymol 153:3-11) and normalized or subtracted using a methodology adapted from Soares (1994, Proc Natl Acad Sci 91:9228-9232), Swaroop et al. (1991, Nucleic Acids Res 19:1954), and Bonaldo et al. (1996, Genome Res 6:791-806). The modified Soares normalization procedure was utilized to reduce the repetitive cloning of highly expressed high abundance cDNAs while maintaining the overall sequence complexity of the library. Modification included significantly longer hybridization times which allowed for increased gene discovery rates by biasing the normalized libraries toward those infrequently expressed low-abundance cDNAs which are poorly represented in a standard transcript image (Soares, supra).


[0144] II Isolation and Sequencing of cDNA Clones


[0145] Plasmids were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using one of the following: the Magic or WIZARD MINIPREPS DNA purification system (Promega); the AGTC MINIPREP purification kit (Edge BioSystems, Gaithersburg Md.); the QIAWELL 8, QIAWELL 8 Plus, or QIAWELL 8 Ultra plasmid purification systems, or the REAL PREP 96 plasmid purification kit (Qiagen). Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4° C.


[0146] Alternatively, plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao (1994) Anal Biochem 216:1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).


[0147] cDNA sequencing reactions were processed using standard methods or high-throughput instrumentation such as the CATALYST 800 thermal cycler (ABI) or the DNA ENGINE thermal cycler (MJ Research, Watertown Mass.) in conjunction with the HYDRA microdispenser (Robbins Scientific, Sunnyvale Calif.) or the MICROLAB 2200 system (Hamilton, Reno Nev.). cDNA sequencing reactions were prepared using reagents provided by APB or supplied in sequencing kits such as the PRISM BIGDYE cycle sequencing kit (ABI). Electrophoretic separation of cDNA sequencing reactions and detection of labeled cDNAs were carried out using the MEGABACE 1000 DNA sequencing system (APB); the PRISM 373 or 377 sequencing systems (ABI) in conjunction with standard protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, supra, Unit 7.7).


[0148] III Extension of cDNA Sequences


[0149] Nucleic acid sequences were extended using the cDNA clones and oligonucleotide primers. One primer was synthesized to initiate 5′ extension of the known fragment, and the other, to initiate 3′ extension of the known fragment. The initial primers were designed using OLIGO primer analysis software (Molecular Biology Insights, Cascade Colo.), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68° C. to about 72° C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.


[0150] Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed. Preferred libraries are ones that have been size-selected to include larger cDNAs. Also, random primed libraries are preferred because they will contain more sequences with the 5′ and upstream regions of genes. A randomly primed library is particularly useful if an oligo d(T) library does not yield a full-length cDNA.


[0151] High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the DNA ENGINE thermal cycler (MJ Research). The reaction mix contained DNA template, 200 mmol of each primer, reaction buffer containing Mg2+, (NH4)2SO4, and β-mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Invitrogen), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 60° C., 1 min; Step 4: 68° C., 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68° C., 5 min; Step 7: storage at 4° C. In the alternative, the parameters for primer pair T7 and SK+ (Stratagene) were as follows: Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 57° C., 1 min; Step 4: 68° C., 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68° C., 5 min; Step 7: storage at 4° C.


[0152] The concentration of DNA in each well was determined by dispensing 100 μl PICOGREEN reagent (0.25% reagent in 1×TE, v/v; Molecular Probes) and 0.5 μl of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton Mass.) and allowing the DNA to bind to the reagent. The plate was scanned in a FLUOROSKAN II (Labsystems Oy) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 μl to 10 μl aliquot of the reaction mixture was analyzed by electrophoresis on a 1% agarose mini-gel to determine which reactions were successful in extending the sequence.


[0153] The extended nucleic acids were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison Wis.), and sonicated or sheared prior to religation into pUC18 vector (APB). For shotgun sequencing, the digested nucleic acids were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with AGARACE enzyme (Promega). Extended clones were religated using T4 DNA ligase (New England Biolabs, Beverly Mass.) into pUC18 vector (APB), treated with Pfu DNA polymerase (Stratagene) to fill-in restriction site overhangs, and transformed into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37° C. in 384-well plates in LB/2×carbenicillin liquid media.


[0154] The cells were lysed, and DNA was amplified by PCR using Taq DNA polymerase (APB) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 60° C., 1 min; Step 4: 72° C., 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72° C., 5 min; Step 7: storage at 4° C. DNA was quantified using PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions described above. Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the PRISM BIGDYE terminator cycle sequencing kit (ABI).


[0155] IV Assembly and Analysis of Sequences


[0156] The nucleic acid sequences of the cDNAs presented in the Sequence Listing may contain occasional sequencing errors and unidentified nucleotides (N) that reflect state-of-the-art technology at the time the cDNA was sequenced. Occasional sequencing errors and Ns may be resolved and SNPs verified either by resequencing the cDNA or using algorithms to compare multiple sequences; these techniques are well known to those skilled in the art who wish to practice the invention. The sequences may be analyzed using a variety of algorithms described in Ausubel (supra, unit 7.7) and in Meyers (1995; Molecular Biology and Biotechnology, Wiley VCH, New York N.Y., pp. 856-853).


[0157] Component nucleotide sequences from chromatograms were subjected to PHRED analysis (Phil Green, University of Washington, Seattle Wash.) and assigned a quality score. The sequences having at least a required quality score were subject to various pre-processing algorithms to eliminate low quality 3′ ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, bacterial contamination sequences, and sequences smaller than 50 base pairs. Sequences were screened using the BLOCK 2 program (Incyte Genomics), a motif analysis program based on sequence information contained in the SWISS-PROT and PROSITE databases (Bairoch et al. (1997) Nucleic Acids Res 25:217-221; Attwood et al. (1997) J Chem Inf Comput Sci 37:417-424).


[0158] Processed sequences were subjected to assembly procedures in which the sequences were assigned to bins, one sequence per bin. Sequences in each bin were assembled to produce consensus sequences, templates. Subsequent new sequences were added to existing bins using BLAST (Altschul (1990, supra); Altschul (1993, supra); Karlin et al. (1988) Proc Natl Acad Sci 85:841-845), BLASTn (vers.1.4, WashU), and CROSSMATCH software (Green, supra). Candidate pairs were identified as all BLAST hits having a quality score greater than or equal to 150. Alignments of at least 82% local identity were accepted into the bin. The component sequences from each bin were assembled using PHRAP (Green, supra). Bins with several overlapping component sequences were assembled using DEEP PHRAP (Green, supra).


[0159] Bins were compared against each other, and those having local similarity of at least 82% were combined and reassembled. Reassembled bins having templates of insufficient overlap (less than 95% local identity) were re-split. Assembled templates were also subjected to analysis by STITCHER/EXON MAPPER algorithms which analyzed the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced genes across tissue types, disease states, and the like. These resulting bins were subjected to several rounds of the above assembly procedures to generate the template sequences found in the LIFESEQ GOLD database (Incyte Genomics).


[0160] The assembled templates were annotated using the following procedure. Template sequences were analyzed using BLASTn (vers. 2.0, NCBI) versus GBpri (GenBank vers. 116). “Hits” were defined as an exact match having from 95% local identity over 200 base pairs through 100% local identity over 100 base pairs, or a homolog match having an E-value equal to or greater than 1×10−8. (The “E-value” quantifies the statistical probability that a match between two sequences occurred by chance). The hits were subjected to frameshift FASTx versus GENPEPT (GenBank version 109). In this analysis, a homolog match was defined as having an E-value of 1×10−8. The assembly method used above was described in U.S. Ser. No. 09/276,534, filed Mar. 25, 1999, and the LIFESEQ GOLD user manual (Incyte Genomics).


[0161] Following assembly, template sequences were subjected to motif, BLAST, Hidden Markov Model (HMM; Pearson and Lipman (1988) Proc Natl Acad Sci 85:2444-2448; Smith and Waterman, supra), and functional analyses, and categorized in protein hierarchies using methods described in U.S. Ser. No. 08/812,290, filed Mar. 6, 1997; U.S. Ser. No. 08/947,845, filed Oct. 9, 1997; U.S. Pat. No. 5,953,727; and U.S. Ser. No. 09/034,807, filed Mar. 4, 1998. Template sequences may be further queried against public databases such as the GenBank rodent, mammalian, vertebrate, eukaryote, prokaryote, and human EST databases.


[0162] V Selection of Sequences, Microarray Preparation and Use


[0163] Incyte clones represent template sequences derived from the LIFESEQ GOLD assembled human sequence database (Incyte Genomics). In cases where more than one clone was available for a particular template, the 5′-most clone in the template was used on the microarray. The HUMAN GENOME GEM series 1-5 microarrays (Incyte Genomics) contain 45,320 array elements which represent 22,632 annotated clusters and 22,688 unannotated clusters. For the UNIGEM series microarrays (Incyte Genomics), Incyte clones were mapped to non-redundant Unigene clusters (Unigene database (build 46), NCBI; Shuler (1997) J Mol Med 75:694-698), and the 5′ clone with the strongest BLAST alignment (at least 90% identity and 100 bp overlap) was chosen, verified, and used in the construction of the microarray. The UNIGEM V 2.0 microarray (Incyte Genomics) contains 8,502 array elements which represent 8,372 annotated genes and 130 unannotated clusters. Table 5 shows the GenBank annotations (where available) for SEQ ID NOs:1-292 of this invention as produced by BLAST analysis.


[0164] To construct microarrays, cDNAs were amplified from bacterial cells using primers complementary to vector sequences flanking the cDNA insert. Thirty cycles of PCR increased the initial quantity of cDNAs from 1-2 ng to a final quantity greater than 5 μg. Amplified cDNAs were then purified using SEPHACRYL-400 columns (APB). Purified cDNAs were immobilized on polymer-coated glass slides. Glass microscope slides (Corning, Corning N.Y.) were cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments. Glass slides were etched in 4% hydrofluoric acid (VWR Scientific Products, West Chester Pa.), washed thoroughly in distilled water, and coated with 0.05% aminopropyl silane (Sigma-Aldrich) in 95% ethanol. Coated slides were cured in a 110° C. oven. cDNAs were applied to the coated glass substrate using a procedure described in U.S. Pat. No. 5,807,522. One microliter of the cDNA at an average concentration of 100 ng/ul was loaded into the open capillary printing element by a high-speed robotic apparatus which then deposited about 5 nl of cDNA per slide.


[0165] Microarrays were UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene), and then washed at room temperature once in 0.2% SDS and three times in distilled water. Non-specific binding sites were blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (Tropix, Bedford Mass.) for 30 minutes at 60° C. followed by washes in 0.2% SDS and distilled water as before.


[0166] VI Preparation of Samples


[0167] Human coronary artery endothelial cells (HCAEC), human iliac artery endothelial cells (HIAEC), human pulmonary artery endothelial cells (HPAEC), human aortic endothelial cells (HAEC), human umbilical artery endothelial cells (HUAEC), human uterine myometium microvasculature (UtMVEC), human dermal microvasculature (HMVECD), and human umbilical vein endothelial cells (HUVEC) were obtained from Clonetics Corporation (San Diego, Calif.). Cells were grown in EGM-2 medium at 37° C., 5% CO2 using procedures supplied by Clonetics. All cells were grown to approximately 85% confluency and harvested without treatment.


[0168] Isolation and Labeling of Sample cDNAs


[0169] Cells were harvested and lysed in 1 ml of TRIZOL reagent (5×106 cells/ml; Invitrogen). The lysates were vortexed thoroughly and incubated at room temperature for 2-3 minutes and extracted with 0.5 ml chloroform. The extract was mixed, incubated at room temperature for 5 minutes, and centrifuged at 16,000×g for 15 minutes at 4° C. The aqueous layer was collected, and an equal volume of isopropanol was added. Samples were mixed, incubated at room temperature for 10 minutes, and centrifuged at 16,000×g for 20 minutes at 4° C. The supernatant was removed, and the RNA pellet was washed with 1 ml of 70% ethanol, centrifuged at 16,000×g at 4° C., and resuspended in RNAse-free water. The concentration of the RNA was determined by measuring the optical density at 260 nm. Poly(A) RNA was prepared using an OLIGOTEX mRNA kit (Qiagen) with the following modifications: OLIGOTEX beads were washed in tubes instead of on spin columns, resuspended in elution buffer, and then loaded onto spin columns to recover mRNA. To obtain maximum yield, the mRNA was eluted twice. The pooled control was prepared using an equal amount of mRNA from HCAEC, HUAEC, UtMVEC, HIAEC, HMVECD, and HUVEC.


[0170] Each poly(A) RNA sample was reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/μl oligo-d(T) primer (21mer), 1×first strand buffer, 0.03 units/ul RNAse inhibitor, 500 uM dATP, 500 uM dGTP, 500 uM dTTP, 40 uM dCTP, and 40 uM either dCTP-Cy3 or dCTP-Cy5 (APB). The reverse transcription reaction was performed in a 25 ml volume containing 200 ng poly(A) RNA using the GEMBRIGHT kit (Incyte Genomics). Specific control poly(A) RNAs (YCFRO6, YCFR45, YCFR67, YCFR85, YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were synthesized by in vitro transcription from non-coding yeast genomic DNA (W. Lei, unpublished). As quantitative controls, control mRNAs (YCFRO6, YCFR45, YCFR67, and YCFR85) at 0.002 ng, 0.02 ng, 0.2 ng, and 2 ng were diluted into reverse transcription reaction at ratios of 1:100,000,1:10,000, 1:1000, 1:100 (w/w) to sample mRNA, respectively. To sample differential expression patterns, control rmRNAs (YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were diluted into reverse transcription reaction at ratios of 1:3, 3:1, 1:10, 10:1, 1:25, 25:1 (w/w) to sample mRNA. Reactions were incubated at 37° C. for 2 hr, treated with 2.5 ml of 0.5M sodium hydroxide, and incubated for 20 minutes at 85° C. to the stop the reaction and degrade the RNA. cDNAs were purified using two successive CHROMA SPIN 30 gel filtration spin columns (Clontech). Cy3- and CyS-labeled reaction samples were combined as described below and ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The cDNAs were then dried to completion using a SpeedVAC system (Savant Instruments, Holbrook N.Y.) and resuspended in 14 μl 5×SSC, 0.2% SDS.


[0171] VII Hybridization and Detection


[0172] Hybridization reactions contained 9 μl of sample mixture containing 0.2 μg each of Cy3-labeled pooled control and Cy5 labeled source-specific vascular endothelium cDNA synthesis products in 5×SSC, 0.2% SDS hybridization buffer. The mixture was heated to 65° C. for 5 minutes and was aliquoted onto the microarray surface and covered with an 1.8 cm2 coverslip. The microarrays were transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber was kept at 100% humidity internally by the addition of 140 μl of 5×SSC in a corner of the chamber. The chamber containing the microarrays was incubated for about 6.5 hours at 60° C. The microarrays were washed for 10 min at 45° C. in low stringency wash buffer (1×SSC, 0.1% SDS), three times for 10 minutes each at 45° C. in high stringency wash buffer (0.1×SSC), and dried.


[0173] Reporter-labeled hybridization complexes were detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Santa Clara Calif.) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of CyS. The excitation laser light was focused on the microarray using a 20×microscope objective (Nikon, Melville N.Y.). The slide containing the microarray was placed on a computer-controlled X-Y stage on the microscope and raster-scanned past the objective. The 1.8 cm×1.8 cm microarray used in the present example was scanned with a resolution of 20 micrometers.


[0174] In two separate scans, the mixed gas multiline laser excited the two fluorophores sequentially. Emitted light was split, based on wavelength, into two photomultiplier tube detectors (PMT R1477; Hamamatsu Photonics Systems, Bridgewater N.J.) corresponding to the two fluorophores. Appropriate filters positioned between the microarray and the photomultiplier tubes were used to filter the signals. The emission maxima of the fluorophores used were 565 nm for Cy3 and 650 nm for Cy5. Each microarray was typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus was capable of recording the spectra from both fluorophores simultaneously.


[0175] The sensitivity of the scans was calibrated using the signal intensity generated by a cDNA control species. Samples of the calibrating cDNA were separately labeled with the two fluorophores and identical amounts of each were added to the hybridization mixture. A specific location on the microarray contained a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000.


[0176] The output of the photomultiplier tube was digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood, Mass.) installed in an IBM-compatible PC computer. The digitized data were displayed as an image where the signal intensity was mapped using a linear 20-color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal). The data was also analyzed quantitatively. Where two different fluorophores were excited and measured simultaneously, the data were first corrected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using each fluorophore's emission spectrum.


[0177] A grid was superimposed over the fluorescence signal image such that the signal from each spot was centered in each element of the grid. The fluorescence signal within each element was then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis was the GEMTOOLS gene expression analysis program (Incyte Genomics). Significance was defined as signal to background ratio exceeding 2× and area hybridization exceeding 40%.


[0178] VIII Data Analysis and Results


[0179] Array elements that exhibited at least 1.5-fold change in expression in one or more vascular endothelial sample, a signal intensity over 250 units, a signal-to-background ratio of at least 2.5, and an element spot size of at least 40% were identified as differentially expressed using the GEMTOOLS program (Incyte Genomics). Differential expression values were converted to log base 2 scale; negative values indicate higher expression in the specific endothelial sample relative to the pooled control. Expression in large arteries (HCAEC, HIAEC, HPAEC, HAEC) was compared with expression in microvasculature (UtMVEC, HMVECD). The clones representing cDNAs that are expressed at higher levels in microvasculature are shown in Tables 1 and 2. The clones representing cDNAs that are expressed at higher levels in large artery endothelium are shown in Table 3.


[0180] IX Further Characterization of Differentially Expressed cDNAs and Proteins


[0181] Clones were blasted against the LIFESEQ Gold 5.1 database (Incyte Genomics) and an Incyte template and its sequence variants were chosen for each clone. Table 4 provides a map between the clones present on microarrays and identified in Tables 1, 2, and 3, and the cDNAs identified in the Sequence Listing. The template and variant sequences were blasted against GenBank database to acquire annotation. The nucleotide sequences were translated into amino acid sequence which was blasted against the GenPept and other protein databases to acquire annotation and characterization, i.e., structural motifs. Table 5 shows the annotations provided by BLAST analysis for each cDNA of the present invention.


[0182] Percent sequence identity can be determined electronically for two or more amino acid or nucleic acid sequences using the MEGALIGN program, a component of LASERGENE software (DNASTAR). The percent identity between two amino acid sequences is calculated by dividing the length of sequence A, minus the number of gap residues in sequence A, minus the number of gap residues in sequence B, into the sum of the residue matches between sequence A and sequence B, times one hundred. Gaps of low or of no homology between the two amino acid sequences are not included in determining percentage identity.


[0183] Sequences with conserved protein motifs may be searched using the BLOCKS search program. This program analyses sequence information contained in the Swiss-Prot and PROSITE databases and is useful for determining the classification of uncharacterized proteins translated from genomic or cDNA sequences (Bairoch (supra); Attwood (supra). PROSITE database is a useful source for identifying functional or structural domains that are not detected using motifs due to extreme sequence divergence. Using weight matrices, these domains are calibrated against the SWISS-PROT database to obtain a measure of the chance distribution of the matches.


[0184] The PRINTS database can be searched using the BLIMPS search program to obtain protein family “fingerprints”. The PRINTS database complements the PROSITE database by exploiting groups of conserved motifs within sequence alignments to build characteristic signatures of different protein families. For both BLOCKS and PRINTS analyses, the cutoff scores for local similarity were: >1300=strong, 1000-1300=suggestive; for global similarity were: p<exp-3; and for strength (degree of correlation) were: >1300=strong, 1000-1300=weak.


[0185] X Other Hybridization Technologies and Analyses


[0186] Other hybridization technologies utilize a variety of substrates such as nylon membranes, capillary tubes, etc. Arranging cDNAs on polymer coated slides is described in Example V; sample cDNA preparation and hybridization and analysis using polymer coated slides is described in examples VI and VII, respectively.


[0187] The cDNAs are applied to a membrane substrate by one of the following methods. A mixture of cDNAs is fractionated by gel electrophoresis and transferred to a nylon membrane by capillary transfer. Alternatively, the cDNAs are individually ligated to a vector and inserted into bacterial host cells to form a library. The cDNAs are then arranged on a substrate by one of the following methods. In the first method, bacterial cells containing individual clones are robotically picked and arranged on a nylon membrane. The membrane is placed on LB agar containing selective agent (carbenicillin, kanamycin, ampicillin, or chloramphenicol depending on the vector used) and incubated at 37° C. for 16 hr. The membrane is removed from the agar and consecutively placed colony side up in 10% SDS, denaturing solution (1.5 M NaCl, 0.5 M NaOH), neutralizing solution (1.5 M NaCl, 1 M Tris, pH 8.0), and twice in 2×SSC for 10 min each. The membrane is then UV irradiated in a STRATALINKER UV-crosslinker (Stratagene).


[0188] In the second method, cDNAs are amplified from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR amplification increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 μg. Amplified nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are arranged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobilized by denaturation, neutralization, and UV irradiation as described above.


[0189] Hybridization probes derived from cDNAs of the Sequence Listing are employed for screening cDNAs, mRNAs, or genomic DNA in membrane-based hybridizations. Probes are prepared by diluting the cDNAs to a concentration of 40-50 ng in 45 μl TE buffer, denaturing by heating to 100° C. for five min and briefly centrifuging. The denatured cDNA is then added to a REDIPRIME tube (APB), gently mixed until blue color is evenly distributed, and briefly centrifuged. Five microliters of [32P]dCTP is added to the tube, and the contents are incubated at 37° C. for 10 min. The labeling reaction is stopped by adding 5 μl of 0.2M EDTA, and probe is purified from unincorporated nucleotides using a PROBEQUANT G-50 microcolumn (APB). The purified probe is heated to 100° C. for five min and then snap cooled for two min on ice.


[0190] Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and 1×high phosphate buffer (0.5 M NaCl, 0.1 M Na2HPO4, 5 mM EDTA, pH 7) at 55° C. for two hr. The probe, diluted in 15 ml fresh hybridization solution, is then added to the membrane. The membrane is hybridized with the probe at 55° C. for 16 hr. Following hybridization, the membrane is washed for 15 min at 25° C. in 1 mM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 min each at 25° C. in 1 mM Tris (pH 8.0). To detect hybridization complexes, XOMAT-AR film (Eastman Kodak, Rochester N.Y.) is exposed to the membrane overnight at −70° C., developed, and examined.


[0191] XI Expression of the Encoded Protein


[0192] Expression and purification of a protein encoded by a cDNA of the invention is achieved using bacterial or virus-based expression systems. For expression in bacteria, cDNA is subcloned into a vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcription. Examples of such promoters include, but are not limited to, the trp-lac (tac) hybrid promoter and the T5 or T7 bacteriophage promoter in conjunction with the lac operator regulatory element.


[0193] Recombinant vectors are transformed into bacterial hosts, such as BL21 (DE3). Antibiotic resistant bacteria express the protein upon induction with IPTG. Expression in eukaryotic cells is achieved by infecting Spodoptera frugiperda (Sf9) insect cells with recombinant baculovirus, Autographica californica nuclear polyhedrosis virus. The polyhedrin gene of baculovirus is replaced with the cDNA by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates.


[0194] Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of transcription. For ease of purification, the protein is synthesized as a fusion protein with glutathione-S-transferase (GST; APB) or a similar alternative such as FLAG. The fusion protein is purified on immobilized glutathione under conditions that maintain protein activity and antigenicity. After purification, the GST moiety is proteolytically cleaved from the protein with thrombin. A fusion protein with FLAG, an 8-amino acid peptide, is purified using commercially available monoclonal and polyclonal anti-FLAG antibodies (Eastman Kodak, Rochester N.Y.).


[0195] XII Production of Specific Antibodies


[0196] A denatured protein from a reverse phase HPLC separation is obtained in quantities up to 75 mg. This denatured protein is used to immunize mice or rabbits following standard protocols. About 100 μg is used to immunize a mouse, while up to 1 mg is used to immunize a rabbit. The denatured protein is radioiodinated and incubated with murine B-cell hybridomas to screen for monoclonal antibodies. About 20 mg of protein is sufficient for labeling and screening several thousand clones.


[0197] In another approach, the amino acid sequence translated from a cDNA of the invention is analyzed using PROTEAN software (DNASTAR) to determine antigenic determinants of the protein. The optimal sequences for immunization are usually at the C-terminus, the N-terminus, and those intervening, hydrophilic regions of the protein that are likely to be exposed to the external environment when the protein is in its natural conformation. Typically, oligopeptides about 15 residues in length are synthesized using an 431 peptide synthesizer (ABI) using Fmoc-chemistry and then coupled to keyhole limpet hemocyanin (KLH; Sigma-Aldrich) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester. If necessary, a cysteine may be introduced at the N-terminus of the peptide to permit coupling to KLH. Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated goat anti-rabbit IgG.


[0198] Hybridomas are prepared and screened using standard techniques. Hybridomas of interest are detected by screening with radioiodinated protein to identify those fusions producing a monoclonal antibody specific for the protein. In a typical protocol, wells of 96 well plates (FAST, Becton-Dickinson, Palo Alto Calif.) are coated with affinity-purified, specific rabbit-anti-mouse (or suitable anti-species Ig) antibodies at 10 mg/ml. The coated wells are blocked with 1% BSA and washed and exposed to supernatants from hybridomas. After incubation, the wells are exposed to radiolabeled protein at 1 mg/ml. Clones producing antibodies bind a quantity of labeled protein that is detectable above background.


[0199] Such clones are expanded and subjected to 2 cycles of cloning at 1 cell/3 wells. Cloned hybridomas are injected into pristane-treated mice to produce ascites, and monoclonal antibody is purified from the ascitic fluid by affinity chromatography on protein A (APB). Monoclonal antibodies with affinities of at least 108 M−1, preferably 109 to 1010 M−1 or stronger, are made by procedures well known in the art.


[0200] XIII Purification of Naturally Occurring Protein Using Specific Antibodies


[0201] Naturally occurring or recombinant protein is purified by immunoaffinity chromatography using antibodies specific for the protein. An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected.


[0202] XIV Screening Molecules for Specific Binding With the cDNA or Protein


[0203] The cDNA or fragments thereof and the protein or portions thereof are labeled with 32P-dCTP, Cy3-dCTP, Cy5-dCTP (APB), or BIODIPY or FITC (Molecular Probes), respectively. Candidate molecules or compounds previously arranged on a substrate are incubated in the presence of labeled nucleic or amino acid. After incubation under conditions for either a cDNA or a protein, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed. The binding molecule is identified by its arrayed position on the substrate. Data obtained using different concentrations of the nucleic acid or protein are used to calculate affinity between the labeled nucleic acid or protein and the bound molecule. High throughput screening using very small assay volumes and very small amounts of test compound is fully described in U.S. Pat. No. 5,876,946.


[0204] All patents and publications mentioned in the specification are incorporated herein by reference. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the field of molecular biology or related fields are intended to be within the scope of the following claims.
1TABLE 1136814971569CloneIDHAECHCAECHPAECHIAECUtMVECHMVECDHUAECHUVECHUVECHUVEC311197ND  0.28ND  0.12  0.15−1.43  0.18−0.11  0.04  0.162586533ND  0.03NDND−0.17−0.66−0.05NDNDND2539263−0.35−0.25−0.49−0.10−0.48−0.65−0.54ND−0.48−0.142228990ND  0.16ND−0.04−0.50−0.61−0.12−0.25  0.09  0.093126622ND−0.50ND  0.23  0.26−0.64−0.17−0.16  0.19  0.141612384ND−0.38ND−0.36ND−0.60NDNDND−0.38924936ND  0.03ND−0.28  0.17−0.62  0.36−0.26ND−0.061655416NDNDNDNDND−1.25NDNDNDND1746640ND−0.13ND−0.18−0.18−0.79−0.27−0.12−0.14−0.192325661−0.07−0.24−0.08  0.05−0.16−0.59−0.08ND  0.15−0.062373263ND−0.33ND−0.26ND−0.59NDNDND−0.172510835ND−0.28ND−0.26−0.35−0.78−0.19−0.12−0.18−0.053206154ND−0.49ND  0.10−0.19−0.59−0.33−0.31−0.32  0.103249851ND  0.02NDNDND−0.63NDNDNDND3600367NDNDNDNDND−0.61NDNDNDND3891867ND−0.01ND−0.23  0.03−0.59−0.06ND−0.22−0.263972234−0.21−0.22−0.24−0.08−0.14−0.61−0.33ND−0.35−0.253973887ND−0.09ND−0.01−0.02−0.61−0.06−0.08−0.17  0.034514944ND  0.38ND  0.40−0.42−0.89  0.02  0.07  0.06−0.02797406ND−0.23ND−0.10−0.08−0.59−0.40  0.00  0.02−0.131655734NDNDNDNDND−0.82NDNDNDND1817646ND−0.16ND  0.14  0.19−0.71  0.09  0.25  0.34  0.192278772ND−0.29ND−0.46−0.13−0.70−0.30ND−0.41−0.122739076ND−0.44ND−0.30−0.61−0.71−0.52−0.35ND−0.462828159ND  0.01ND−0.02−0.04−0.65−0.07  0.02−0.16  0.053068978ND−0.15ND−0.49−0.16−0.69−0.17ND−0.03  0.203172265ND  0.57ND−0.12  0.29−0.72  0.77ND  0.29  0.533639751−0.02−0.17  0.21  0.22  0.29−0.600.31ND  0.45  0.313734094ND−0.38ND−0.24−0.49−0.60−0.46−0.07ND−0.473804843ND−0.36ND−0.19−0.11−0.58−0.34−0.28−0.24−0.094092750ND  0.04NDND  0.09−0.65  0.08−0.05  0.02ND4175323ND−0.21ND  0.11−0.42−0.66−0.21−0.30ND−0.254380064ND−0.33ND−0.09−0.05−0.68−0.22ND−0.18−0.064413637ND  0.23ND  0.33  0.44−0.69  0.19−0.08  0.31  0.494543123ND−0.43ND−0.01−0.30−0.64−0.16ND−0.42−0.044571104ND−0.31ND  0.10−0.45−0.58−0.39−0.27−0.39−0.114853673ND−0.11ND  0.15−0.03−0.66−0.12  0.07−0.02ND5271674ND−0.19ND−0.14−0.07−0.68−0.08ND−0.13−0.035293028ND−0.31ND  0.22  0.07−0.69  0.32ND−0.21  0.475509328−0.17  0.48  0.09  0.32−0.54−0.81−0.22ND−0.21−0.375682290−0.03  0.12  0.32  0.05−0.03−0.97  0.11ND  0.30  0.195903108−0.25−0.30−0.43−0.11−0.26−0.62−0.29ND−0.30−0.202717435ND−0.46ND−0.25−0.60  0.29−0.10  0.29−0.05−0.095013541ND−0.06ND  0.01−0.67−0.01−0.01ND−0.03−0.041405844ND  0.64ND−0.31−0.62−0.44−0.40−0.58−0.41−0.381624102−0.05−0.18−0.25−0.01−0.63  0.22−0.29ND−0.15−0.171889028ND−0.40ND−0.34−0.74−0.04−0.27−0.24−0.29−0.222175008ND−0.47ND−0.50−0.94  0.13−0.28−0.08ND−0.454252862ND−0.11ND−0.21−0.75−0.08−0.21ND−0.06−0.084521868ND−0.18ND−0.20−0.69−0.06−0.32ND−0.26−0.16682741ND  0.25ND  0.01−0.77−0.35−0.26−0.30ND−0.402042056ND  0.07ND−0.04−0.58−0.20−0.31−0.42−0.42−0.382416447ND−0.16ND−0.24−0.76−0.53  0.00−0.26  0.18−0.062462908ND−0.26ND−0.22−0.58  0.28−0.23−0.08−0.35−0.112727104  0.09  0.11  0.36  0.01−0.85  0.06−0.07ND  0.14  0.112739076ND−0.44ND−0.30−0.61−0.71−0.52−0.35ND−0.463426776NDNDNDND−0.67−0.43NDNDNDND3961930ND  0.04ND−0.07−0.96−0.02  0.05ND  0.00  0.053970808ND−0.06ND−0.04−0.86  0.10  0.03ND  0.10  0.023982273ND  0.06ND  0.05−0.67  0.05  0.17ND  0.11  0.144090358ND  0.10ND  0.11−0.68  0.05  0.16ND  0.07  0.004146967−0.04  0.06  0.14  0.12−0.78  0.07−0.14NDND  0.104185527ND−0.09ND  0.02−1.39  0.03−0.05ND−0.10−0.064219295ND−0.22ND−0.27−1.35−0.12−0.28ND−0.09−0.164306221ND−0.04ND−0.20−0.59−0.02−0.09ND−0.06−0.054334474ND  0.14ND  0.27−1.63−0.18  0.15ND  0.06  0.224383888ND−0.06ND−0.12−1.27  0.23−0.01ND  0.06−0.044420064ND−0.04ND−0.03−0.60  0.18  0.05ND  0.02  0.074422135ND−0.01ND  0.01−0.60  0.17−0.02ND−0.01  0.074508211ND−0.05ND−0.07−0.97  0.14−0.10ND  0.09  0.004533406ND−0.14ND  0.00−0.73  0.05−0.13ND  0.07  0.124561939ND−0.10ND  0.06−1.49  0.21−0.07ND−0.08  0.064606503ND  0.00ND−0.01−1.48  0.07  0.04ND  0.05  0.064633555ND  0.17ND  0.15−1.42  0.01  0.09ND  0.08  0.034636285ND−0.06ND  0.03−0.97  0.12  0.08ND  0.08  0.094639612ND  0.08ND  0.02−1.25  0.06  0.13ND  0.12  0.134704961ND  0.02ND  0.11−1.14  0.07−0.04ND  0.02  0.014710591ND−0.03ND  0.05−1.53  0.13−0.05ND−0.17  0.004718317ND  0.08ND  0.06−1.04−0.08  0.12ND  0.05−0.024766304ND−0.04ND−0.03−0.62  0.14  0.11ND−0.02−0.044771265ND−0.11ND  0.07−0.69  0.07  0.03ND−0.03−0.024860482ND−0.02ND  0.02−0.73  0.07−0.06ND−0.03  0.004875773ND−0.03ND  0.14−0.92  0.04  0.02ND  0.09−0.014886749ND  0.09ND  0.04−1.02  0.07  0.15ND  0.07  0.085024011ND−0.10ND  0.02−0.79  0.05−0.18ND−0.09−0.065069809ND−0.01ND−0.03−1.10−0.01  0.00ND  0.12  0.085094869ND−0.12ND  0.03−0.73  0.18−0.04ND  0.11−0.015099959ND−0.04ND−0.21−0.73−0.03−0.17ND−0.04−0.135219277ND  0.17ND  0.06−1.10  0.18  0.07ND  0.13  0.165262750ND  0.13ND  0.12−0.83−0.09  0.08ND  0.06−0.035265974ND−0.12ND−0.07−1.67  0.04−0.03ND  0.01  0.065407110NDNDNDND−1.24ND  0.12NDND  0.205508135ND  0.13ND  0.05−1.15−0.17  0.20ND−0.09−0.025639879−0.42−0.47−0.28−0.43−0.64  0.07−0.18ND−0.30−0.235680271ND−0.11ND−0.10−1.78  0.04−0.16ND−0.11−0.08


[0205]

2










TABLE 2












avg log2 ratio
avg log2 ratio
Δ MV and



Clone ID
MV
non-MV
non-MV





















1624543
−1.36
0.88
−2.25



2527665
−1.17
0.29
−1.46



2069806
0.22
1.63
−1.41



5407110
−1.24
0.16
−1.40



3180888
−0.74
0.55
−1.29



2862539
−0.84
0.39
−1.22



1558081
−0.96
0.24
−1.20



3428945
−0.77
0.37
−1.14



4175376
−0.51
0.57
−1.08



4334474
−0.90
0.14
−1.05



1555545
−0.50
0.46
−0.96



392380
−0.10
0.80
−0.90



302934
−0.16
0.73
−0.89



1921574
−0.54
0.35
−0.88



1963963
−0.48
0.39
−0.87



4737158
−0.36
0.51
−0.86



2656707
−0.76
0.08
−0.85



211389
−0.06
0.77
−0.82



2009822
0.03
0.85
−0.81



3328081
−0.34
0.47
−0.81



4633555
−0.71
0.09
−0.80



1938951
−0.10
0.70
−0.80



2955269
−0.68
0.11
−0.80



5265974
−0.81
−0.02
−0.79



3172586
−0.69
0.08
−0.78



275587
0.00
0.77
−0.78



3205003
−0.48
0.28
−0.76



148825
0.00
0.76
−0.76



1959179
−0.52
0.24
−0.76



3735389
−0.69
0.06
−0.76



4514944
−0.65
0.10
−0.75



3172265
−0.21
0.54
−0.75



5680271
−0.87
−0.12
−0.75



311197
−0.64
0.11
−0.75



4606503
−0.70
0.04
−0.74



263340
0.00
0.74
−0.74



1887959
−0.88
−0.15
−0.73



289407
0.06
0.79
−0.73



318182
−0.12
0.61
−0.73



1909038
−0.44
0.29
−0.73



2005968
−0.04
0.69
−0.73



5026408
−0.65
0.08
−0.72



341884
−0.08
0.65
−0.72



351475
0.02
0.74
−0.72



680
0.01
0.73
−0.72



2007151
0.04
0.76
−0.72



1938947
0.01
0.73
−0.72



82954
−0.03
0.69
−0.72



58000
−0.05
0.67
−0.72



5508135
−0.66
0.06
−0.71



44877
−0.10
0.61
−0.71



4639612
−0.60
0.11
−0.71



3182642
−0.35
0.35
−0.70



508113
0.03
0.72
−0.69



387305
−0.15
0.54
−0.69



1447055
−0.34
0.34
−0.69



3519816
−0.31
0.38
−0.68



419316
0.08
0.75
−0.67



5047895
−0.83
−0.16
−0.67



320332
−0.21
0.46
−0.67



499848
−0.04
0.63
−0.67



5501816
−0.58
0.10
−0.67



5682290
−0.50
0.17
−0.67



1358185
−0.35
0.32
−0.67



219488
−0.04
0.63
−0.67



2137464
−0.36
0.31
−0.67



2137041
−0.37
0.29
−0.66



96425
−0.60
0.06
−0.66



3249851
−0.63
0.02
−0.65



345378
0.06
0.71
−0.65



385550
−0.04
0.61
−0.65



349854
0.07
0.71
−0.65



42920
−0.20
0.44
−0.65



2776
−0.02
0.63
−0.64



238793
−0.07
0.57
−0.64



1599036
−0.33
0.31
−0.64



2924347
−0.39
0.24
−0.64



465496
0.07
0.71
−0.64



284124
−0.02
0.62
−0.64



464323
0.01
0.65
−0.64



4710591
−0.70
−0.06
−0.63



388931
0.09
0.72
−0.63



3086929
−0.50
0.13
−0.63



2011839
0.04
0.67
−0.63



1987738
−0.43
0.19
−0.62



75549
0.02
0.65
−0.62



4718317
−0.56
0.06
−0.62



1995380
−0.26
0.36
−0.62



4386789
−0.11
0.50
−0.61



408522
0.04
0.65
−0.61



5509328
−0.67
−0.07
−0.61



274627
0.03
0.64
−0.61



4185527
−0.68
−0.07
−0.61



414891
0.00
0.61
−0.61



5069809
−0.55
0.05
−0.60



278282
−0.02
0.58
−0.60



9079
0.00
0.61
−0.60



241643
−0.02
0.58
−0.60



44525
0.01
0.60
−0.60



4561939
−0.64
−0.05
−0.60











[0206]

3















TABLE 3
















1368
1497
1569


CloneID
HAEC
HCAEC
HPAEC
HIAEC
UtMVEC
HMVECD
HUAEC
HUVEC
HUVEC
HUVEC

























4398915
−0.82
−0.51
−0.23
−0.52
0.09
−0.42
0.23
ND
−0.02
0.38


2947513
−0.58
−0.43
−0.30
−0.13
−0.15
−0.29
−0.20
ND
−0.46
−0.15


2152363
−0.71
−0.42
−0.21
−0.33
−0.13
−0.44
−0.43
ND
−0.27
−0.23


5680843
−0.65
−0.35
−0.24
−0.26
0.05
−0.17
−0.23
ND
−0.32
0.01


3099768
−0.74
−0.30
0.14
−0.08
−0.11
−0.29
−0.19
ND
−0.21
−0.16


5911570
−0.63
−0.24
−0.23
−0.29
0.15
−0.01
−0.11
ND
−0.20
0.07


5902267
−1.00
−0.22
−0.37
−0.12
−0.15
−0.51
−0.65
ND
−0.41
−0.44


4557571
−0.73
−0.22
−0.38
−0.29
−0.17
−0.44
−0.32
ND
−0.27
−0.52


5911592
−0.87
−0.16
−0.47
−0.17
−0.36
−0.45
−0.86
ND
−0.32
−0.30


5519430
−0.62
−0.03
−0.29
−0.21
0.01
0.07
−0.02
ND
−0.22
−0.07


1969756
−0.60
−0.02
0.51
0.16
0.04
−0.18
0.10
ND
0.04
0.14


1512990
−0.60
0.02
0.72
0.15
−0.01
−0.01
0.16
ND
−0.04
−0.03


4739765
−0.71
0.07
−0.22
0.15
0.03
−0.33
−0.23
ND
−0.24
−0.14


5046165
−0.70
0.14
−0.10
0.16
0.20
−0.08
0.31
ND
−0.30
−0.04


2580580
−0.62
0.20
0.66
0.15
0.12
−0.02
0.10
ND
0.08
0.13


760553
−0.90
0.23
−0.55
0.34
−0.56
−0.20
−0.35
ND
−0.43
−0.20


4295711
−0.68
0.28
1.07
0.11
0.12
0.02
0.11
ND
−0.08
0.15


431455
−0.58
0.42
1.01
0.29
0.07
0.18
0.27
ND
0.35
0.37


3142396
−0.85
0.55
−0.13
0.64
−0.21
−0.11
−0.07
ND
−0.04
−0.21


3129691
−0.96
0.66
−0.24
0.74
−0.33
0.10
0.04
ND
0.10
−0.26


2962167
ND
−0.83
ND
−0.41
−0.21
0.04
−0.30
0.36
ND
0.56


5587406
ND
−0.79
ND
−0.10
0.35
0.10
−0.72
0.10
−0.03
0.07


2966620
ND
−0.76
ND
−0.40
−0.17
−0.35
0.60
−0.26
ND
−0.19


2895245
ND
−0.70
ND
−0.43
−0.41
−0.13
−0.54
0.28
ND
0.15


1648251
ND
−0.70
ND
−0.58
−0.11
−0.30
−0.67
−0.43
ND
−0.54


1601323
ND
−0.70
ND
−0.41
−0.45
−0.04
−0.55
−0.55
ND
−0.48


2047549
ND
−0.68
ND
−0.54
0.07
−0.32
−0.38
0.00
−0.01
0.08


1624459
ND
−0.68
ND
−0.36
−0.49
−0.17
−0.53
0.22
0.00
0.29


2235515
ND
−0.68
ND
−0.42
0.27
0.13
−0.16
0.00
0.27
0.29


2771046
ND
−0.68
ND
−0.24
0.18
0.01
−0.05
0.89
ND
0.32


3658034
ND
−0.67
ND
−0.51
−0.06
0.01
0.13
0.15
0.38
0.71


2722128
ND
−0.66
ND
−0.50
−0.17
−0.45
−0.14
0.34
ND
0.08


2845102
ND
−0.66
ND
0.01
−0.21
−0.28
0.06
0.27
ND
0.57


1271606
ND
−0.66
ND
−0.42
−0.19
−0.45
−0.53
−0.13
ND
−0.56


2151054
ND
−0.65
ND
ND
ND
ND
ND
ND
ND
ND


1600586
ND
−0.63
ND
−0.51
−0.27
−0.25
−0.42
−0.23
ND
−0.28


2740187
ND
−0.61
ND
−0.04
0.05
−0.21
−0.17
0.25
ND
−0.02


4462162
ND
−0.61
ND
−0.56
0.18
−0.02
−0.78
−0.26
ND
−0.52


1510714
ND
−0.61
ND
0.08
0.21
−0.09
−0.30
−0.54
ND
−0.48


2134968
ND
−0.60
ND
−0.42
−0.42
−0.04
−0.34
−0.17
ND
−0.20


2200604
ND
−0.60
ND
−0.44
−0.25
−0.10
−0.31
0.02
ND
−0.02


2657738
ND
−0.60
ND
−0.31
−0.38
−0.20
−0.34
−0.26
ND
−0.30


1559731
ND
−0.60
ND
−0.48
0.06
−0.22
−0.31
0.18
ND
0.09


3136864
ND
−0.59
ND
−0.45
−0.32
−0.39
−0.51
−0.19
ND
−0.15


5198974
ND
−0.59
ND
0.07
−0.10
−0.06
−0.10
−0.11
−0.19
0.03


2765165
ND
−0.59
ND
−0.44
−0.25
−0.15
−0.49
ND
−0.27
−0.33


1481225
ND
−0.59
ND
−0.25
−0.29
0.10
−0.17
0.08
ND
0.27


3088261
ND
−0.59
ND
−0.47
0.02
−0.18
−0.19
−0.04
−0.26
−0.16


1518328
ND
−0.59
ND
−0.50
0.68
0.06
−0.45
0.31
ND
0.23


1619865
ND
−0.58
ND
−0.18
0.02
−0.57
0.81
−0.21
0.21
0.11


1551767
ND
−0.58
ND
−0.43
−0.28
0.47
−0.58
−0.05
ND
−0.43


1498947
ND
−1.11
ND
−0.63
0.24
0.16
0.03
0.27
0.44
0.35


3316684
ND
−0.95
ND
−0.71
0.33
−0.25
0.67
−0.24
−0.15
−0.28


1617218
ND
−0.91
ND
−0.87
1.22
−0.21
0.15
ND
0.35
0.25


4144001
ND
−0.83
ND
−0.83
0.95
−0.19
−0.01
0.13
0.23
0.14


2657149
ND
−0.75
ND
−0.60
0.28
0.13
−0.03
0.24
ND
0.49


2455118
ND
−0.72
ND
−0.59
−0.29
−0.44
−0.31
−0.11
ND
−0.38


2174441
ND
−0.69
ND
−0.69
0.70
−0.10
−0.61
−0.47
ND
−0.23


1997915
ND
−0.68
ND
−0.65
−0.18
−0.48
−0.31
0.27
−0.02
−0.07


1357231
ND
−0.65
ND
−0.70
−0.04
−0.07
−0.35
ND
0.41
0.15


698665
ND
−0.65
ND
−0.76
−0.52
0.53
−0.87
−0.51
ND
−0.42


4295364
ND
−0.62
ND
−0.63
−0.29
−0.29
−0.49
0.04
ND
−0.28


1909266
ND
−0.59
ND
−0.62
−0.12
−0.28
−0.44
−0.23
ND
−0.32


3132987
ND
−0.56
ND
−0.70
0.18
−0.31
−0.25
0.30
ND
−0.23


1886842
ND
−0.55
ND
−0.65
−0.33
−0.47
−0.55
−0.48
ND
−0.36


4144001
ND
−0.54
ND
−0.61
0.97
−0.01
0.05
0.14
ND
0.28


3596853
ND
−0.53
ND
−0.60
−0.01
−0.21
−0.29
0.30
ND
0.24


3393606
ND
−0.53
ND
−0.67
0.22
0.36
0.08
0.67
0.63
0.44


693783
ND
−0.52
ND
−0.58
0.64
−0.52
−0.04
ND
−0.40
−0.56


2470485
ND
−0.46
ND
−0.77
0.09
−0.01
−0.10
0.06
ND
0.22


3081067
ND
−0.45
ND
−0.62
0.21
0.18
−0.11
0.55
ND
0.34


1313183
ND
−0.43
ND
−0.63
0.48
−0.20
0.82
−0.17
ND
−0.29


1979792
ND
−0.37
ND
−0.66
−0.28
−0.50
−0.56
−0.46
ND
−0.33


494970
ND
−0.33
ND
−0.60
0.03
0.16
−0.21
ND
0.06
0.07


3030988
ND
−0.28
ND
−0.62
−0.19
−0.22
−0.47
−0.28
ND
−0.26


2453436
ND
−0.06
ND
−0.60
−0.29
0.07
−0.58
−0.25
ND
−0.12


129009
ND
0.05
ND
−0.60
−0.05
−0.13
−0.34
0.64
ND
0.12


2220338
ND
0.05
ND
−0.75
−0.20
−0.12
−0.52
−0.26
ND
0.05


1581451
−0.51
−0.29
−0.79
0.05
0.03
−0.05
0.41
ND
0.38
0.21


3598343
0.04
−0.06
−0.68
−0.14
−0.34
−0.14
−0.18
ND
−0.20
−0.26










[0207]

4









TABLE 4








SEQ ID NO
Template ID
Clone ID
Start
Stop



















1
1383156.23
311197
36
607


2
1097334.8
2586533
533
940


3
1330210.32
2539263
1
521


4
1330210.33
2539263
1043
1568


5
978840.1
2228990
15
1740


6
1850670CB1
3126622
115
2046


7
427830.26
1612384
371
802


8
002838.45
924936
113
534


8
002838.45
3891867
74
536


9
002838.44
924936
226
924


10
1451265CB1
1655416
250
1459


11
347284.6
1746640
2375
2776


12
255966.9
2325661
1472
1734


13
406387.7
2373263
5565
6689


14
062199.7
2510835
630
3421


15
1383059.61
3206154
737
1305


16
332595.5
3249851
160
733


17
332595.12
3249851
4134
4685


18
5401223CB1
3600367
4817
5226


19
002838.40
3891867
346
659


20
378186.30
3972234
109
638


20
378186.30
5903108
231
506


21
348196.83
3973887
1
321


22
348196.89
3973887
5446
5720


23
1136856.23
4514944
564
1065


24
1136856.25
4514944
1198
1615


25
101575.1
797406
853
1279


26
276310.1
1655734
1
275


27
 014284CB1
1817646
1360
1854


28
272599.1
2278772
1
278


29
1383712.24
2739076
183
657


30
1820861CB1
2828159
18
439


31
330902.2
2828159
723
1137


32
211881.1
3068978
1
548


33
399161.1
3172265
473
1102


34
138206.1
3639751
1
806


35
1241528.18
3734094
228
801


36
1091353.11
3804843
−132
287


37
405058.1
4092750
2916
3391


38
406573.9
4175323
60
247


39
3365683CB1
4380064
132
462


40
406992.1
4413637
875
1787


41
399626.1
4543123
−1
813


42
474725.3
4571104
1
257


43
402460.13
4853673
1164
1687


44
038033.1
5271674
28
619


45
208075.1
5293028
417
737


46
3479061CB1
5509328
395
1113


47
1454465.1
5682290
88
600


48
378186.29
5903108
51
592


49
330836.4
2717435
2752
3160


50
330836.5
2717435
1929
2391


51
410882.6
5013541
1082
1848


52
1405844CB1
1405844
72
424


53
330933.15
1624102
4491
4895


54
979182.3
1889028
1751
2236


55
1382838.129
2175008
4210
4704


56
898864.15
4252862
1
300


57
033527.1
4521868
896
1523


58
351122.2
682741
879
1348


59
 522433CB1
2042056
4
384


60
081187.1
2416447
1
64


61
2462908CB1
2462908
0
1028


62
350416.3
2462908
1453
1650


63
1061366.15
2727104
29
462


64
026200.1
3426776
1
388


65
214744.1
3961930
1
291


66
214811.1
3970808
297
516


67
107572.1
3982273
1
614


68
215003.1
4090358
−82
473


69
106250.1
4146967
1
544


70
403668.1
4185527
144
694


71
011664.1
4219295
54
201


72
059849.1
4306221
858
1191


73
1197030.23
4334474
766
1514


74
042195.1
4383888
1
256


75
983907.1
4420064
1420
1861


76
210826.1
4422135
1778
2267


77
343356.1
4508211
97
461


78
1439948.1
4533406
1
546


79
984996.3
4561939
13
552


80
102564.1
4606503
121
520


81
146768.1
4633555
241
747


82
340291.1
4636285
1
405


83
134460.1
4639612
499
924


84
053411.1
4704961
80
571


85
1448208.1
4710591
256
742


86
070655.1
4718317
97
490


87
406389.1
4766304
697
1495


88
167601.1
4771265
172
449


89
341084.1
4860482
1
231


90
404224.1
4875773
807
1490


91
026596.1
4886749
1
499


92
1041038.1
5024011
1
601


93
080588.1
5069809
1
206


94
014408.2
5094869
633
989


95
351752.1
5099959
853
1138


96
002661.1
5219277
38
833


97
399162.11
5262750
502
746


98
174086.1
5265974
219
542


99
116731.1
5407110
96
365


100
1453971.4
5508135
1819
2037


101
347635.1
5639879
258
785


102
1401326.2
5680271
−2
1079


103
1330204.51
1624543
750
1807


104
1327351.31
2527665
1
345


105
1327351.35
2527665
943
1530


106
332723.6
2069806
1883
2175


107
1184503.1
3180888
45
567


108
410610.1
2862539
502
771


109
986073.1
1558081
1571
3089


110
1102234.29
3428945
1
1199


111
1102234.28
3428945
698
1194


112
029061.1
4175376
176
1158


113
233180.39
1555545
889
1940


114
010317.1
392380
252
635


115
2007614CB1
302934
2163
2527


116
065582.1
1921574
1
492


117
1188409.1
1963963
5567
6043


117
1188409.15
1963963
1936
2307


119
1102234.75
4737158
1
237


120
403534.6
2656707
3273
4071


121
238349.4
211389
3334
3488


122
017058.1
2009822
216
477


123
 190188CB1
3328081
6981
7353


124
1383106.7
1938951
2878
3117


125
1510895.1
2955269
843
1348


126
1454873.5
3172586
683
1217


127
222836.2
275587
21
410


128
3098081CB1
3205003
687
1188


129
1383398.6
148825
67
612


130
1383398.7
148825
173
979


131
198862.10
1959179
669
1046


132
2515360CB1
3735389
26
474


132
2515360CB1
1887959
4110
4667


133
334180.6
263340
1556
2800


134
982535.2
289407
748
1100


135
1501794.11
318182
3742
4129


136
 983910CB1
1909038
399
849


137
999097.1
2005968
902
1069


138
293858.32
5026408
1
561


139
1269631CB1
341884
5433
6307


140
337208.11
351475
1327
2388


141
337800.3
680
1875
2134


142
1361343CB1
2007151
3511
3706


143
1504867.1
1938947
290
440


144
410917.13
82954
1480
1625


145
2080904CB1
58000
676
927


146
2520680CB1
44877
1312
1482


147
1274560.12
3182642
20
1177


148
313149.1
508113
1567
1968


149
268514.25
387305
1522
1901


150
2100016CB1
1447055
17
735


151
2906265CB1
3519816
1
464


152
189155.2
419316
1042
1239


153
336733.9
5047895
1
419


154
1273136.1
320332
1083
1471


155
253888.1
499848
1444
1851


156
407570.3
5501816
1094
1274


157
977136.8
1358185
1
425


158
977136.1
1358185
2274
2803


159
1500085.3
219488
203
564


160
1439712.1
2137464
1
630


161
015508.1
2137041
23
414


162
 111552CB1
96425
1391
1645


163
1641902CB1
345378
1011
1676


164
252999.1
385550
874
1379


165
977391.15
349854
1740
2103


166
1363208CB1
42920
2573
2827


167
 002776CB1
2776
1604
1896


168
140557.23
238793
2888
3029


169
140557.22
238793
4820
4961


170
1286746CB1
1599036
391
1072


171
1102234.13
2924347
286
808


172
1401226.4
465496
321
558


173
331677.2
284124
4201
4705


174
234870.4
464323
2255
2659


175
346051.5
3086929
474
954


176
232055.1
2011839
720
859


177
070248.106
1987738
38
901


178
028253.6
75549
329
508


179
989381.1
1995380
86
951


180
984009.1
4386789
130
1807


181
404758.7
408522
469
653


182
208350.1
274627
2871
3129


183
334193.1
414891
1242
1602


184
335046.8
278282
63
429


185
198947.2
9079
1153
1406


186
014669.1
241643
994
1143


187
2951807CB1
44525
451
683


188
482452.13
388931
1
434


189
2513610CB1
4398915
39
675


190
1376812.4
2947513
7
338


191
1446355.3
2152363
345
805


192
1446355.2
2152363
757
1138


193
365121.1
5680843
708
841


194
1399068.1
3099768
1
367


195
474395.1
5911570
0
588


196
001300.10
5902267
19
429


197
482193.2
4557571
1
294


198
450887.1
5911592
−30
569


199
1281959CB1
5519430
31
316


200
197856.4
1969756
1514
1888


201
197856.9
1969756
756
966


202
1088680.3
1512990
2186
2790


203
246020.6
4739765
529
882


204
1446842.1
5046165
1
502


205
2580580CB1
2580580
520
1281


206
1503093.1
760553
503
965


207
338662.1
4295711
1
1043


208
324037.1
431455
610
1245


209
989010.3
3142396
1
780


210
 470784CB1
3142396
16
704


211
989010.5
3129691
279
1000


212
473040.1
2962167
953
4477


213
400203.1
5587406
1234
1598


214
474437.17
2966620
744
934


215
1345561CB1
2895245
486
1988


216
1513913CB1
1648251
761
1296


217
1453827.5
1601323
1028
1456


218
199471.2
2047549
149
1452


219
 000278CB1
1624459
39
1688


220
233942.1
2235515
128
1984


221
1136923.19
2771046
1585
2068


222
1136923.18
2771046
3424
3644


223
3658034CB1
3658034
33
1112


224
1450005.5
2722128
708
1186


225
1450005.6
2722128
1771
2258


226
464689.64
2845102
2180
4636


227
334388.7
1271606
227
714


228
994660.1
2151054
1090
1518


229
3147649CB1
1600586
3059
4037


230
2652949CB1
2740187
1
1692


231
350300.25
4462162
6221
6833


232
1273213CB1
1510714
416
1968


233
222242.35
2134968
1
1553


234
 621792CB1
2200604
379
1464


235
 148820CB1
2657738
1534
1980


236
1812221CB1
1559731
2850
4201


237
1504168.4
3136864
338
408


238
109925.11
5198974
171
442


239
1073168.8
2765165
786
4417


240
2239072CB1
1481225
730
2171


241
4767318CB1
3088261
1282
1562


242
1339068CB1
1518328
1597
1933


243
2966620CB1
1619865
84
275


244
1347119.1
1551767
771
3446


245
 816792CB1
1498947
136
514


245
 816792CB1
2657149
1379
1843


246
1092427.6
3316684
388
642


246
1092427.6
1313183
4362
5823


247
251793.1
1617218
16
651


248
1622869CB1
4144001
722
1109


249
2581158CB1
2455118
144
2070


250
2174441CB1
2174441
471
946


251
235191.3
1997915
12
473


252
235191.4
1997915
213
782


253
1867417CB1
1357231
3075
3752


254
1285632CB1
698665
1377
4119


255
336992.5
4295364
1431
3446


256
1909266CB1
1909266
23
556


257
1214953.16
3132987
−20
1965


258
1850092CB1
1886842
1589
2576


259
1555752CB1
3596853
27
3636


260
441283.5
3393606
5296
8423


261
4349106CB1
693783
344
1030


262
2700132CB1
2470485
412
985


263
441283.4
3081067
713
1208


264
2613976CB1
1979792
961
1906


265
337894.8
494970
1
458


266
1452351.17
3030988
586
1971


267
1754695CB1
2453436
10
1489


268
232888.4
129009
3844
5648


269
247817.4
2220338
3585
5794


270
365801.2
1581451
210
1205


271
1449816.11
3598343
1
535










[0208]

5









TABLE 5








SEQ ID






NO
Template ID
GenBank ID
E-value
Annotation



















1
1383156.23
g262770
9.00E−47
type II surfactant protein C, type II SP-C [Oryctolagus cuniculus]


2
1097334.8
g4321787
1.00E−12
6-pyruvoyl-tetrahydropterin synthase


3
1330210.32
g10439446
0
FLJ22911 fis, clone KAT05860, highly similar to stimulatory regulatory component Gs of adenylyl






cyclase


4
1330210.33
g31914
0
Human mRNA for coupling protein G(s) alpha subunit (alpha-S2)


5
978840.1
g3882333
5.00E−82
KIAA0806 protein


6
1850670CB1
g182606
0
Human factor I (C3b/C4b inactivator) mRNA, complete cds.


7
427830.26
g182967
0
Human GTP-binding protein superfamily, G-protein alpha-olf subunit (olfactory) mRNA, complete






cds.


8
002838.45
g2326174
0
Human selenoprotein W (selW) mRNA, complete cds.


9
002838.44
g2326174
0
Human selenoprotein W (selW) mRNA, complete cds.


10
1451265CB1
g3025445
0
R32184_1


11
347284.6
g1575562
0
Human hematopoietic progenitor kinase (HPK1) mRNA, complete cds.


12
255966.9
g7020703
0
Human cDNA FLJ20533 fis, clone KAT10931.


13
406387.7
g5689439
0
KIAA1051 protein [Homo sapiens]


14
062199.7
g7320864
0
Human mRNA for putative integral membrane transporter protein (LC27 gene).


15
1383059.61
g186840
0
Human colin carcinoma laminin-binding protein mRNA, complete cds.


16
332595.5
g2979417
0
Human mRNA for PCDH7 (BH-Pcdh)a, complete cds.


17
332595.12
g2979419
0
Human mRNA for PCDH7 (BH-Pcdh)b, complete cds.


18
5401223CB1
g3021700
0
Human BAI3 mRNA, complete cds.


19
002838.40
g2384720
0
Human selenoprotein W (hSelW) mRNA, complete cds.


20
378186.30
g338446
0
Human ribosomal protein S16 mRNA, complete cds.


21
348196.83
g177208
0
Human 4F2 glycosylated heavy chain (4F2HC) antigen gene, exon 1 and 2.


22
348196.89
g1200185
0
Human U22 snoRNA host gene (UHG) gene, complete sequence.


23
1136856.23
g183994
0
Human heregulin-beta1 gene, complete cds.


24
1136856.25
g183998
0
Human heregulin-beta3 gene, complete cds.


25
101575.1
g34328
0
Human mRNA for lactate dehydrogenase B (LDH-B).


26
276310.1
g488074
4.00E−57
Human insulinoma-associated (IA-1) gene, partial cds.


27
014284CB1
g1006656
0
Human mRNA for cathepsin C.


28
272599.1


Incyte Unique


29
1383712.24
g292442
0
Human ribosomal protein S20 (RPS2O) mRNA, complete cds.


30
1820861CB1
g2580587
0
Human platelet activating receptor homolog (H963) mRNA, complete cds.


31
330902.2
g2580588
0
platelet activating receptor homolog [Homo sapiens]


32
211881.1
g340088
7.00E−15
Human small nuclear rna pseudogene (clone pul-1) and flanks.


33
399161.1
g36076
2.00E−37
Human gene possibly encoding U1 RNA.


34
138206.1


Incyte Unique


35
1241528.18
g388031
0
Human ribosomal protein L11 mRNA, complete cds.


36
1091353.11
g506338
5.00E−75
flavoprotein subunit of complex II [Homo sapiens]


37
405058.1
g30398
0
Human mRNA for D-1 dopamine receptor.


38
406573.9
g10433269
3.00E−60
Human cDNA FLJ11898 fis, clone HEMBA1007322.


39
3365683CB1
g34328
0
Human mRNA for lactate dehydrogenase B (LDH-B).


40
406992.1
g11493651
0
Human calcium channel blocker resistance protein CCBR1 mRNA, complete cds.


41
399626.1
g5668857
2.00E−53


Homo sapiens
glutathione transferase A4 gene, exon 1.



42
474725.3
g10440217
1.00E−16
Human cDNA: FLJ23506 fis, clone LNG03055.


43
402460.13
g10438153
0
Human cDNA: FLJ21937 fis, clone HEP04432.


44
038033.1
g6807879
0
Human mRNA; cDNA DKFZp434C0326 (from clone DKFZp434C0326).


45
208075.1
g23914
1.00E−117
Human 7SK45 DNA.


46
3479061CB1
g1103903
0
Human spermidine/spermine N1-acetyltransferase (SSAT) gene, complete cds.


47
1454465.1
g5419854
0
Human mRNA; cDNA DKFZp566N043 (from clone DKFZp566N043).


48
378186.29
g338446
0
Human ribosomal protein S16 mRNA, complete cds.


49
330836.4
g11275977
0
Human NOTCH 2 (N2) mRNA, complete cds.


50
330836.5
g11527996
0
Human NOTCH2 protein (NOTCH2) mRNA, complete cds.


51
410882.6
g1674386
0
diabetes mellitus type I autoantigen


52
1405844CB1
g180925
0
Human CO-029.


53
330933.15
g6330032
0
Human mRNA for KIAA1147 protein, partial cds.


54
979182.3
g424133
0
Human connexin45 gene, complete cds.


55
1382838.129
g189208
0
Human nidogen mRNA, complete cds.


56
898864.15
g11691653
7.00E−10
Human PDX1 gene for lipoyl-containing component X, exons 1-11.


57
033527.1
g10434733
3.00E−86
FLJ12953 fis, clone NT2RP2005490, moderately similar to Mouse D3Mm3e mRNA.


58
351122.2
g183581
0
Human glycoprotein IIIa (ITGB3) gene, exon 15 and partial cds.


59
 522433CB1
g1813326
0
Human mRNA for TGF-beta superfamily protein, complete cds.


60
081187.1
g219928
1.00E−19
Human midkine gene, complete cds.


61
2462908CB1
g7861556
0
Human mRNA for SIRP-b2, complete cds.


62
350416.3
g7861556
0
Human mRNA for SIRP-b2, complete cds.


63
1061366.15


Incyte Unique


64
026200.1


Incyte Unique


65
214744.1
g3978547
0
Human chromosome 3 subtelomeric region.


66
214811.1
g10433865
0
Human cDNA FLJ12399 fis, clone MAMMA1002780.


67
107572.1


Incyte Unique


68
215003.1


Incyte Unique


69
106250.1
g7768719
4.00E−10


Homo sapiens
genomic DNA, chromosome 21q, section 63/105.



70
403668.1


Incyte Unique


71
011664.1
g35504
0
Human M gene for M1-type and M2-type pyruvate kinase.


72
059849.1


Incyte Unique


73
1197030.23
g36139
0
Human mRNA for ribosomal protein L7.


74
042195.1


Incyte Unique


75
983907.1


Incyte Unique


76
210826.1
g7768770
2.00E−12


Homo sapiens
genomic DNA, chromosome 21q, section 97/105.



77
343356.1


Incyte Unique


78
1439948.1


Incyte Unique


79
984996.3


Incyte Unique


80
102564.1


Incyte Unique


81
146768.1


Incyte Unique


82
340291.1
g3387951
3.00E−10


Homo sapiens
clone 24487 mRNA sequence.



83
134460.1
g5823146
3.00E−80
testis specific protein


84
053411.1


Incyte Unique


85
1448208.1


Incyte Unique


86
070655.1
g5926700
0
Human genomic DNA, chromosome 6p21.3, HLA Class I region, section 12/20.


87
406389.1
g4150939
2.00E−33
GSG1 [Mus musculus]


88
167601.1


Incyte Unique


89
341084.1


Incyte Unique


90
404224.1


Incyte Unique


91
026596.1


Incyte Unique


92
1041038.1
g5042384
0
Human KVLQT1 gene.


93
080588.1


Incyte Unique


94
014408.2


Incyte Unique


95
351752.1
g10432680
0


Homo sapiens
cDNA FLJ11425 fis, clone HEMBA1001051.



96
002661.1


Incyte Unique


97
399162.11
g7770237
5.00E−11
PRO2822 [Homo sapiens]


98
174086.1


Incyte Unique


99
116731.1


Incyte Unique


100
1453971.4
g5106994
9.00E−38
HSPC038 protein [Homo sapiens]


101
347635.1
g11275977
0
Human NOTCH 2 (N2) mRNA, complete cds.


102
1401326.2
g5926687
9.00E−09
Human genomic DNA, chromosome 3p21.3, clone: 603 to 320, anti-oncogene region, section 3/3.


103
1330204.51
g2554613
0
Human mRNA for RGS5, complete cds.


104
1327351.31
g34613
0
Human mRNA for matrix Gla protein.


105
1327351.35
g187590
0
Human matrix Gla protein (MGP) gene, complete cds.


106
332723.6
g10434712
0
Human cDNA FLJ12937 fis, clone NT2RP2005020.


107
1184503.1
g3002924
0
Human T-cell receptor beta chain (TCRBV13S1-TCRBJ2S1) mRNA, complete cds.


108
410610.1
g187590
0
Human matrix Gla protein (MGP) gene, complete cds.


109
986073.1
g180670
0
Human collagenase type IV mRNA, 3’ end.


110
1102234.29
g36760
0
Human mRNA fragment for T-cell receptor beta chain D-J-C (JUR-beta 2).


111
1102234.28
g36760
0
Human mRNA fragment for T-cell receptor beta chain D-J-C (JUR-beta 2).


112
029061.1


Incyte Unique


113
233180.39
g7770154
0
Human PRO1873 mRNA, complete cds.


114
010317.1


Incyte Unique


115
2007614CB1
g4151118
0
Human fertilin beta mRNA, complete cds.


116
065582.1


Incyte Unique


117
1188409.1
g818
0
protein-glutamine gamma-glutamyltransferase [Bos taurus]


118
1188409.15
g53667
0
biglycan (PGI) [Mus musculus]


119
1102234.75
g33543
0
Human mRNA for T-cell receptor V beta gene segment V-beta-6, clone IGRb11.


120
403534.6
g7243180
0
Human mRNA for KIAA1400 protein, partial cds.


121
238349.4
g6624095
1.00E−07
similar to Coch-5B2


122
017058.1
g8515813
0.0004
RSD-6


123
 190188CB1
g29469
0
Human mRNA for basement membrane heparan sulfate proteoglycan.


124
1383106.7
g10437335
0
Human cDNA: FLJ21267 fis, clone COL01717.


125
1510895.1
g189519
5.00E−66
Human pro-alpha-1 (V) collagen mRNA, complete cds.


126
1454873.5
g339561
0
Human transforming growth factor-beta (tgf-beta) mRNA, complete cds.


127
222836.2
g35685
0
Human testis specific mRNA for protamine 1 (P1).


128
3098081CB1
g3970845
0
Human mRNA for RALDH2-T, complete cds.


129
1383398.6
g1165211
0
Human microfibril-associated glycoprotein-2 MAGP-2 mRNA, complete cds.


130
1383398.7
g1165211
0
Human microfibril-associated glycoprotein-2 MAGP-2 mRNA, complete cds.


131
198862.10
g1907077
0
Human mRNA for Pirin, isolate 17.


132
2515360CB1
g2370201
0
Human mRNA for procollagen alpha 2(V).


133
334180.6
g2224696
0
Human mRNA for KIAA0378 gene, partial cds.


134
982535.2
g2612787
0
Human P2X7 gene, exon 9-11.


135
1501794.11
g7672348
0
Human unknown mRNA.


136
 983910CB1
g36760
0
Human mRNA fragment for T-cell receptor beta chain D-J-C (JUR-beta 2).


137
999097.1
g2781413
0
Human clone DT2P1G7 mRNA, CAG repeat region.


138
293858.32
g10439538
0
Human cDNA: FLJ22988 fis, clone KAT11812.


139
1269631CB1
g2654025
0
gp250 precursor [Mus musculus]


140
337208.11
g10439667
0
FLJ23092 fis, clone LNG07245


141
337800.3
g7959737
1.00E−177
PRO1037 [Homo sapiens]


142
1361343CB1
g3882220
0
Human mRNA for KIAA0750 protein, complete cds.


143
1504867.1
g10436845
0
Human cDNA: FLJ20887 fis, clone ADKA03276.


144
410917.13
g506431
0
lysosomal acid lipase [Homo sapiens]


145
2080904CB1
g10437138
0
Human cDNA: FLJ21110 fis, clone CAS05377.


146
2520680CB1
g2058395
0
Human adenovirus protein E3-14.7 k interacting protein 1 (FIP-1) mRNA, complete cds.


147
1274560.12
g36748
0
Human mRNA for T-cell specific protein.


148
313149.1
g10435386
0
FLJ13386 fis, clone PLACE1001104, weakly similar to MYOSIN HEAVY CHAIN,






NON-MUSCLE.


149
268514.25
g1552523
0
Human ionotropic ATP receptor P2X5b mRNA, complete cds.


150
2100016CB1
g2565275
4.00E−48
Dim1p homolog [Homo sapiens]


151
2906265CB1
g338928
0
Human T-cell receptor active beta-chain V-D-J-beta-1.2-C-beta-1 (TCRB) mRNA, partial cds.


152
189155.2
g5931525
3.00E−12
Human genomic DNA, chromosome 22q11.2, Cat Eye Syndrome region


153
336733.9
g6049603
0
Human dickkopf-1 (DKK-1) mRNA, complete cds.


154
1273136.1
g9956031
0
Human clone CDABP0045 mRNA sequence.


155
253888.1
g6682959
0
nuclear receptor binding factor-2


156
407570.3
g11907922
0
Human enhancer of polycomb mRNA, complete cds.


157
977136.8
g1314305
0
Human channel-like integral membrane protein (AQP-1) mRNA, clone AQP-1-2344, partial cds.


158
977136.1
g1314305
0
Human channel-like integral membrane protein (AQP-1) mRNA, clone AQP-1-2344, partial cds.


159
1500085.3
g5410279
0
Human HSPCO34 protein mRNA, complete cds.


160
1439712.1
g10434784
0
FLJ12987 fis, clone NT2RP3000068, weakly similar to SON OF SEVENLESS PROTEIN






HOMOLOG 1.


161
015508.1
g7243180
0
Human mRNA for KIAA1400 protein, partial cds.


162
 111552CB1
g10439538
0
Human cDNA: FLJ22988 fis, clone KAT11812.


163
1641902CB1
g6093424
0
Human connexin 37 (GJA4) mRNA, complete cds.


164
252999.1
g10047170
0
Human mRNA for KIAA1553 protein, partial cds.


165
977391.15
g7019914
0
Human cDNA FLJ20061 fis, clone COL01383.


166
1363208CB1
g3283351
0
Human lens epithelium-derived growth factor mRNA, complete cds.


167
 002776CB1
g5868895
0
Human BAG-family molecular chaperone regulator-4 mRNA, complete cds.


168
140557.23
g7020279
0
Human cDNA FLJ20287 fis, clone HEP04390.


169
140557.22
g3925386
0
Human inversin protein mRNA, complete cds.


170
1286746CB1
g35439
0
Human mRNA for protein gene product (PGP) 9.5.


171
1102234.13
g36172
0
Human rearranged T-cell receptor beta chain mRNA.


172
1401226.4
g7768753
3.00E−12


Homo sapiens
genomic DNA, chromosome 21q, section 95/105.



173
331677.2
g190220
0
protein phosphatase 2A 130 kDa regulatory subunit [Homo sapiens]


174
234870.4
g11320875
3.00E−79
cdk-binding protein


175
346051.5
g532677
0
Human mRNA for ribonuclease A (RNase A), complete cds.


176
232055.1


Incyte Unique


177
070248.106
g7959920
0
Human PRO2646 mRNA, complete cds.


178
028253.6
g4096056
8.00E−84
R28379_1


179
989381.1
g183655
0
Human glutathione S-transferase mRNA, complete cds.


180
984009.1
g863029
0
Human high-mobility group phosphoprotein isoform I-C (HMGIC) gene, exon 5.


181
404758.7
g7684246
0
Human mRNA for organic anion transporter OATP-D, complete cds.


182
208350.1
g10434511
0


Homo sapiens
cDNA FLJ12807 fis, clone NT2RP2002316.



183
334193.1


Incyte Unique


184
335046.8
g940201
0
Human clone LS637A M73 hypoxanthine phosphoribosyltransferase (hprt) deletion mutant mRNA,






partial cd


185
198947.2
g10435130
0
Human cDNA FLJ13209 fis, clone NT2RP4000424.


186
014669.1


Incyte Unique


187
2951807CB1
g4589683
0
Human mRNA for KIAA1017 protein, complete cds.


188
482452.13
g180055
0
Human thymocyte antigen CD1b mRNA, complete cds.


189
2513610CB1
g2088550
0
Human hereditary haemochromatosis region


190
1376812.4
g339840
0
Human triosephosphate isomerase mRNA, complete cds.


191
1446355.3
g8919392
0
Human mRNA full length insert cDNA clone EUROIMAGE 781354.


192
1446355.2
g8919392
0
Human mRNA full length insert cDNA clone EUROIMAGE 781354.


193
365121.1
g10439528
0
FLJ22979 fis, clone KAT11379, highly similar to HUMTUBAK Human alpha-tubulin mRNA.


194
1399068.1
g23925
2.00E−41
Human mRNA for 7SL RNA pseudogene.


195
474395.1
g496886
8.00E−60
Human mRNA for beta tubulin, clone nuk_278.


196
001300.10
g6457274
7.00E−12
putative E1-E2 ATPase


197
482193.2
g5926699
2.00E−86
Human genomic DNA, chromosome 6p21.3, HLA Class I region, section 11/20.


198
450887.1
g7629994
4.00E−33
60S RIBOSOMAL PROTEIN L36 homolog


199
1281959CB1
g1805279
0
Human alpha II spectrin mRNA, complete cds.


200
197856.4
g3947687
0
Human mRNA for Sec24 protein (Sec24A isoform), partial.


201
197856.9
g6807899
0
Human mRNA; cDNA DKFZp434C1526 (from clone DKFZp434C1526).


202
1088680.3
g10434855
0
Human cDNA FLJ13032 fis, clone NT2RP3001120, moderately similar to ZINC FINGER






PROTEIN 136.


203
246020.6
g5689566
0
Human mRNA for KIAA1115 protein, complete cds.


204
1446842.1
g7293054
3.00E−26
Top1 gene product


205
2580580CB1
g9957753
0
Human kidney-specific membrane protein NX-17 mRNA, complete cds.


206
1503093.1
g207500
4.00E−07
alpha-tropomyosin 2 [Rattus norvegicus]


207
338662.1
g804810
1.00E−08
unknown protein [Rattus norvegicus]


208
324037.1
g5689412
0
Human mRNA for KIAA1038 protein, partial cds.


209
989010.3
g34513
0
Human mRNA for monocyte chemoattractant protein 1 (MCP-1).


210
 470784CB1
g240867
0
monocyte chemoattractant protein-1 [Human, mRNA, 739 nt].


211
989010.5
g240867
0
monocyte chemoattractant protein-1 [Human, mRNA, 739 nt].


212
473040.1
g10442025
0
seladin-1 [Homo sapiens]


213
400203.1
g4836418
0
Human endothelial lipase mRNA, complete cds.


214
474437.17
g339972
0
Human TRPM-2 mRNA, complete cds.


215
1345561CB1
g1698395
0
Human lanosterol 14-demethylase cytochrome P450 (CYP51) mRNA, complete cds.


216
1513913CB1
g1665722
0
Human mRNA for RPD3 protein, complete cds.


217
1453827.5
g1350551
0
Human mRNA for ceramide glucosyltransferase, complete cds.


218
199471.2
g2463195
0
Human mRNA for MAD2 protein.


219
 000278CB1
g435676
0
Human mRNA for squalene synthase.


220
233942.1
g11934947
0
Human neuropilin 2 (NRP2) gene, complete cds, alternatively spliced.


221
1136923.19
g178535
0
Human aminopeptidase N/CD13 mRNA encoding aminopeptidase N, complete cds.


222
1136923.18
g178535
0
Human aminopeptidase N/CD13 mRNA encoding aminopeptidase N, complete cds.


223
3658034CB1
g3881654
2.00E−50
similarity to Pfam domain: PF00207 (Alpha-2-macroglobulin family), Score = 377.3,






E-value = 1.1e−113, N = 1


224
1450005.5
g7271464
0
Human soluble neuropilin-1 mRNA,






complete cds.


225
1450005.6
g2978559
0
Human vascular endothelial cell growth factor 165 receptor/neuropilin (VEGF165) mRNA,






complete cds.


226
464689.64
g7415720
0
Human Scd mRNA for stearoyl-CoA desaturase, complete cds.


227
334388.7
g285944
0
Human mRNA for KIAA0104 gene, complete cds.


228
994660.1
g2326205
0
Human Rac3 (RAC3) mRNA, complete cds.


229
3147649CB1
g6006514
0
Human mRNA for SAP 130 spliceosomal protein.


230
2652949CB1
g1827451
0
Human mRNA for VRK2, complete cds.


231
350300.25
g3600027
0
Human dysferlin mRNA, complete cds.


232
1273213CB1
g34458
0
Human DNA for monoamine oxidase type A (14) (partial).


233
222242.35
g2228774
0
Human paraoxonase (PON2) mRNA, complete cds.


234
 621792CB1
g10439430
0
FLJ22896 fis, clone KAT04996


235
 148820CB1
g3417295
0
KIAA0220 [Homo sapiens]


236
1812221CB1
g307382
0
Human RNA helicase A mRNA, complete cds.


237
1504168.4
g403455
0
Human 26S protease (S4) regulatory subunit mRNA, complete cds.


238
109925.11
g188542
0
Human MIC2 mRNA, complete cds.


239
1073168.8
g5689450
0
Human mRNA for KIAA1057 protein, partial cds.


240
2239072CB1
g2696053
0
Human ADDL mRNA for adducin-like protein, complete cds.


241
4767318CB1
g7299015
0
CG9615 gene product


242
1339068CB1
g33943
0
Human mRNA for integrin alpha 6.


243
2966620CB1
g339972
0
Human TRPM-2 mRNA, complete cds.


244
1347119.1
g2370125
0
Human mRNA for ZNF185 gene.


245
 816792CB1
g11934948
0
neuropilin-2a(17) [Homo sapiens]


246
1092427.6
g339547
0
Human transforming growth factor-beta 1 binding protein mRNA, complete cds.


247
251793.1
g5852061
0
Human partial mRNA for Claudin-11 protein (CLDN11 gene).


248
1622869CB1
g3283414
0
Human oligodendrocyte-specific protein (OSP) mRNA, complete cds.


249
2581158CB1
g35070
0
Human mRNA for NAD-dependent methylene tetrahydrofolate dehydrogenase cyclohydrolase






(EC 1.5.1.15).


250
2174441CB1
g2257932
0
Human angiopoietin-2 mRNA, complete cds.


251
235191.3
g6706799
6.00E−46
dJ447F3.2 (ubiquitin-conjugating enzyme E2 H10) [Homo sapiens]


252
235191.4
g2062372
0
Human cyclin-selective ubiquitin carrier protein mRNA, complete cds.


253
1867417CB1
g292829
0
Human DNA topoisomerase II (top2) mRNA, complete cds.


254
1285632CB1
g3342264
0
Na-K-Cl cotransporter [Rattus norvegicus]


255
336992.5
g339805
0
Human topoisomerase I mRNA, complete cds.


256
1909266CB1
g184564
0
Human transformation-sensitive protein (IEF SSP 3521) mRNA, complete cds.


257
1214953.16
g190126
0
Human mitochondrial matrix protein P1 (nuclear encoded) mRNA, complete cds.


258
1850092CB1
g339679
0
Human threonyl-tRNA synthetase mRNA, complete cds.


259
1555752CB1
g2992633
0
Human protein kinase mRNA, complete cds.


260
441283.5
g29864
0
Human CENP-E mRNA.


261
4349106CB1
g10439047
0
FLJ22600 fis, clone HSI04447, highly similar to zinc finger transcription factor GKLF mRNA.


262
2700132CB1
g415818
0
Human mki67a mRNA (long type) for antigen of monoclonal antibody Ki-67.


263
441283.4
g29864
0
Human CENP-E mRNA.


264
2613976CB1
g603954
0
Human mRNA for KIAA0098 protein, partial cds.


265
337894.8
g8489880
0
Human actin binding protein anillin mRNA, complete cds.


266
1452351.17
g5809677
0
Human sperm membrane protein BS-63 mRNA, complete cds.


267
1754695CB1
g35564
0
Human mRNA for purine nucleoside phosphorylase (PNP; EC 2.4.2.1).


268
232888.4
g292829
0
Human DNA topoisomerase II (top2) mRNA, complete cds.


269
247817.4
g2655411
0
Human KDR/flk-1 protein mRNA, complete cds.


270
365801.2
g32368
0
Human Hox1.8 gene for homeobox protein.


271
1449816.11
g1203968
0
Human chromosome X region from filamin (FLN) gene to glucose-6-phosphate dehydrogenase






(G6PD) gene










[0209]


Claims
  • 1. A combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs:1-292 that are differentially expressed in vascular endothelium and the complements of SEQ ID NOs:1-292.
  • 2. The combination of claim 1 wherein the cDNAs are SEQ ID NOs:1-209 that are expressed at a higher level in vascular endothelium from microvasculature and the complements of SEQ ID NOs:1-209.
  • 3. The combination of claim 1, wherein the cDNAs are SEQ ID NOs:210-292 that are expressed at a higher level in vascular endothelium from non-microvasculature and the complements of SEQ ID NOs:210-292.
  • 4. The combination of claim 1, wherein the vascular endothelium is from a subject with a vascular disorder.
  • 5. The combination of claim 4, wherein the vascular disorder is selected from atherosclerosis, hemangioma, hemangioendothelioma, diabetic retinopathy, warts, pyogenic granulomas, Kaposi's sarcoma, scar keloids, allergic oedema, neoplasms, psoriasis, ulcers, follicular cysts, endometriosis, peritoneal sclerosis, and obesity.
  • 6. The combination of claim 1, wherein the cDNAs are immobilized on a substrate.
  • 7. A method for detecting differential expression of one or more cDNAs in a sample containing nucleic acids, the method comprising: a) hybridizing the substrate of claim 6 with nucleic acids of the sample, thereby forming one or more hybridization complexes; b) detecting the hybridization complexes; and c) comparing the hybridization complexes with those of a standard, wherein differences between the standard and sample hybridization complexes indicate differential expression of cDNAs in the sample.
  • 8. The method of claim 7, where in the nucleic acids of the sample are amplified prior to hybridization.
  • 9. The method of claim 7, wherein the sample is from a subject with a vascular disorder and comparison with a standard defines an early, mid, or late stage of that disease or monitors efficacy of a therapeutic regime.
  • 10. A method of screening a plurality of molecules or compounds to identify a molecule or compound which specifically binds a cDNA, the method comprising: a) combining the combination of claim 1 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between each cDNA and at least one molecule or compound, thereby identifying a molecule or compound that specifically binds to each cDNA.
  • 11. The method of claim 10 wherein the plurality of molecules or compounds are selected from DNA molecules, enhancers, mimetics, peptide nucleic acids, proteins, repressors, regulatory proteins, RNA molecules, and transcription factors.
  • 12. An isolated cDNA selected from SEQ ID NOs:36, 51, 61, 83, 87, 110, 111, 136, 159, 198, and 244.
  • 13. A vector containing the cDNA of claim 12.
  • 14. A host cell containing the vector of claim 13.
  • 15. A method for producing a protein, the method comprising the steps of: a) culturing the host cell of claim 14 under conditions for expression of protein; and b) recovering the protein from the host cell culture.
  • 16. A protein or a portion thereof produced by the method of claim 15.
  • 17. A method for using a protein to screen a plurality of molecules or compounds to identify at least one ligand which specifically binds the protein, the method comprising: a) combining the protein of claim 16 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between the protein and a molecule or compound, thereby identifying a ligand which specifically binds the protein.
  • 18. The method of claim 17 wherein the plurality of molecules or compounds is selected from agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA molecules, and ribozymes.
  • 19. A method of using a protein to produce an antibody which specifically binds the protein, the method comprising: a) immunizing an animal with the protein of claim 16 under conditions to elicit an antibody response; b) isolating animal antibodies; and c) screening the isolated antibodies with the protein, thereby identifying an antibody which specifically binds the protein.
  • 20. An antibody produced by the method of claim 19.
Parent Case Info

[0001] This application claims benefit of provisional application Serial No. 60/287,067, filed Apr. 27, 2001.

Provisional Applications (1)
Number Date Country
60287067 Apr 2001 US