Genes regulated in dendritic cell differentiation

Information

  • Patent Application
  • 20030134283
  • Publication Number
    20030134283
  • Date Filed
    October 03, 2001
    22 years ago
  • Date Published
    July 17, 2003
    21 years ago
Abstract
The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed in dendritic cells, 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 cancer, infectious disease, autoimmunity, allergy, and graft versus host disease or to enhance a vaccine.
Description


FIELD OF THE INVENTION

[0001] The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed in dendritic cells and which may be used entirely or in part to diagnose, to stage, to treat, or to monitor the progression or treatment of disorders such as cancer, infectious disease, autoimmunity, allergy, and graft versus host disease.



BACKGROUND OF THE INVENTION

[0002] Array technology can provide a simple way to explore the expression of a single polymorphic gene or the expression profile of a large number of related or unrelated genes. When the expression of a single gene is examined, arrays are employed to detect the expression of that specific gene or its variants. When an expression profile is examined, 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.


[0003] The potential application of gene expression profiling is particularly relevant to characterizing lineage differences during cellular development that will improve diagnosis, prognosis, and treatment of disease. For example, both the levels and sequences expressed in dendritic cells from subjects with autoimmunity may be compared with the levels and sequences expressed in dendritic cells from normal subjects.


[0004] Dendritic cells (DC) are antigen presenting cells (APC) that play a key role in the primary immune response because of their unique ability to present antigens to naive T cells. In addition, DC differentiate into separate subsets that sustain and regulate immune responses following initial contact with antigen. DC subsets include those that preferentially induce particular T helper 1 (Th1) or T helper 2 (Th2) responses and those that regulate B cell responses. Moreover, DC are increasingly being used to manipulate immune responses, either to downregulate an aberrant autoimmune response or to enhance vaccination or a tumor-specific response.


[0005] DC are functionally specialized in correlation with their particular differentiation state. CD34+ myeloid cells found in the bone marrow mature in response to as yet unclear signals into CD14+ CD11c+ monocytes. An innate or antigen non-specific response takes place initially when monocytes circulate to nonlymphoid tissues and respond to lipopolysaccharide (LPS), a bacterially-derived mitogen, and viruses. Such direct encounter with antigen causes secretion of pro-inflammatory cytokines that attract and regulate natural killer cells, macrophages, and eosinophils in the first line of defense against invading pathogens. Monocytes then mature into DC, which capture antigen highly efficiently through endocytosis and antigen receptor uptake. Antigen processing and presentation trigger activation and differentiation into mature DC that express MHC class II molecules on the cell surface and efficiently activate T cells, initiating antigen-specific T cell and B cell responses. In turn, T cells activate DC through CD40 ligand-CD40 interactions, which stimulate expression of the costimulatory molecules CD80 and CD86, the latter most potent in amplifying T cell responses. DC interaction via CD40 with T cells also stimulates the production of inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin (IL)-1. Engagement of RANK, a member of the TNF receptor family by its ligand, TRANCE, which is expressed on activated T cells, enhances the survival of DC through inhibition of apoptosis, thereby enhancing T cell activation. The maturation and differentiation of monocytes into mature DC links the antigen non-specific innate immune response to the antigen-specific adaptive immune response.


[0006] The process by which monocytes differentiate into immature dendritic cells in vivo has not been fully elucidated. Incubation of monocytes with granulocyte-macrophage colony stimulating factor (GM-CSF) and IL4 in vitro yields cells that exhibit functional and morphological characteristics equivalent to immature dendritic cells found in vivo. Moreover, incubation in vitro of immature dendritic cells with TNF-α, CD40 ligand, LPS, or monocyte-conditioned medium yields mature dendritic cells that are potent activators of naive T cells.


[0007] The ability to manipulate DC in vitro and their capacity to mount an effective immune response with small numbers of DC and little antigen has led to: 1) potential immunotherapies for diseases such as cancer, AIDS, and infectious diseases, and 2) enhanced vaccine efficacy. Spontaneous remissions of particular cancers such as renal cell carcinomas and melanomas indicate that the immune system can respond to tumor antigens and eliminate tumors. However, tumors escape immune surveillance through a number of means including secretion of IL-10, macrophage colony stimulating factor, IL-6, and vascular endothelial growth factor, all of which inhibit the activity of DC and promote tolerance of tumor tissue. Delivery of tumor antigen-loaded DC to tumors can induce tumor-specific rejection in animal models. Similarly, pathogens can escape immune surveillance by altering antigen processing and presentation pathways or interfering with maturation of antigen presenting cells. Rather than providing resistance, DC can complicate infection by hosting latent viruses such as Kaposi's virus and cytomegalovirus, and producing HIV-1 and measles virus particles. Vaccines against tumors or infectious pathogens could be improved by systemic or local administration of DC loaded with tumor antigens or attenuated viral particles or components, respectively.


[0008] The expression of killer-inhibitor regulatory molecules, chemokines, chemokine receptors, and proteinases in DC has been verified through sequencing of ESTs. Continuing this search through the use of microarrays may reveal new lymphocyte-binding and antigen-processing molecules, transmembrane and secretory products, and transcription factors that may help to explain the specialized features of DC and allow manipulation of the immune system.


[0009] The present invention provides for a combination comprising a plurality of cDNAs for use in detecting expression of genes encoding proteins in DC. Such a combination satisfies a need in the art in that it can be employed for the improvement of vaccines, in the evaluation of candidates for tissue or organ transplant, and in the diagnosis, prognosis or treatment of cancer, infectious disease, autoimmunity, allergy, and graft versus host disease.



SUMMARY

[0010] The present invention provides a combination comprising a plurality of cDNAs and their complements which are differentially expressed in dendritic cells (DC) and which are selected from SEQ ID NOs:1-260 as presented in the Sequence Listing. In one embodiment, each cDNA is downregulated in DC at least three-fold, SEQ ID NOs:1-29; in another embodiment, each cDNA is upregulated in DC at least three-fold, SEQ ID NOs:30-241; in another embodiment, each cDNA is upregulated in mature DC at least 2.5-fold, SEQ ID NOs:242-257; in yet another embodiment, each cDNA is downregulated in mature DC at least 2.5-fold, SEQ ID NOs:258-260. In one aspect, the combination is useful to characterize monocyte and dendritic cell populations from subjects with disorders such cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. In a second aspect, the combination is useful in evaluation of subjects prior to tissue or organ transplant. In a third aspect, combination is useful in the diagnosis, progression, treatment or monitoring the treatment of cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. In a fourth aspect, the combination is useful to enhance or to measure enhancement of dendritic cell function during immunotherapy of cancer or other disorders. In a fifth aspect, the combination is useful to measure suppression of dendritic cell function during immunotherapy. In a sixth aspect, the combination is useful to enhance the efficacy of vaccines. In a seventh aspect, the combination is immobilized on a substrate.


[0011] The invention provides a high throughput method to detect expression of a nucleic acid which is complementary at least one of the cDNAs of the combination in a sample. The method comprises hybridizing a substrate containing the combination with a sample containing nucleic acids under conditions to form at least one hybridization complex and detecting hybridization complex formation, wherein complex formation indicates expression of at least one complementary nucleic acid in the sample. In one aspect, the sample is from a subject with cancer and differential expression when compared to a standard determines the state of the immune response to the tumor. In another aspect, the sample is from a subject with cancer being treated with dendritic cell immunotherapy, and expression when compared to a standard determines efficacy of the treatment.


[0012] The invention also provides a high throughput method of screening a library or plurality of molecules or compounds to identify a ligand. The method comprises combining 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 aptamers, DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, transcription factors, repressors, and other regulatory proteins. 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.


[0013] The invention further provides an isolated cDNA selected from SEQ ID NOs:24-29, 187-202, 205-213, 216-241, 256, 257, 259, and 260 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 using a host cell to produce a protein comprising culturing the host cell under conditions for the expression of a protein and recovering the protein from the culture.


[0014] 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 aptamers, DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, proteins, agonists, antagonists, antibodies or their fragments, immunoglobulins, inhibitors, drug compounds, and pharmaceutical agents. The invention further provides for using a protein to purify a ligand. The method comprises combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and separating the protein from the ligand, thereby obtaining purified ligand. The invention still further provides a composition comprising the protein and a pharmaceutical carrier.


[0015] The invention also provides methods for using a protein to prepare and purify polyclonal and monoclonal antibodies which specifically bind 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 and purifying 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 from culture monoclonal antibodies which specifically bind the protein.



DESCRIPTION OF THE SEQUENCE LISTING AND TABLES

[0016] 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.


[0017] 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 a template number (TEMPLATE ID).


[0018] Table 1 lists the functional annotation and differential expression of each cDNA that has previously been shown to be upregulated in DC. Columns 1 and 2 show the GenBank identification number (GENBANK ID) and functional annotation (DESCRIPTION), respectively, as determined by BLAST analysis, version 1.4 using default parameters (Altschul (1993) J Mol Evol 36:290-300; Altschul et al. (I 990) J Mol Biol 215:403410) of the cDNA against GenBank (release 117; National Center for Biotechnology Information (NCBI), Bethesda Md.). Column 3 shows the balanced differential expression value(s), BDE, produced by each clone that represented the cDNA in the experiment.


[0019] Tables 2-5 list the functional annotation and differential expression of the cDNAs of the present invention. Table 2 lists those cDNAs that are downregulated in DC; Table 3, cDNAs that are upregulated in DC; Table 4, cDNAs that are upregulated in mature DC; and Table 5, cDNAs that are downregulated in mature DC. In Tables 2-5, Columns 1, 2, and 3 show the SEQ ID NO, CLONE ID, and TEMPLATE ID, respectively. Columns 4 and 5 show the GenBank identification number (GENBANK ID) and functional annotation (DESCRIPTION), respectively, as determined by BLAST analysis of the cDNA against GenBank (release 117; NCBI). Column 6 shows the database from which functional annotation came. Column 7 shows the BDE for each clone that represents that cDNA. Of those sequences that are unique, only the SEQ ID NO, CLONE ID, TEMPLATE ID, and BDE are noted. For SEQ ID NOs:201-215, column 8 of Table 3 shows the E-VALUE for the template, as determined by BLASTx analysis.



DESCRIPTION OF THE INVENTION

[0020] Definitions


[0021] Definitions


[0022] “Array” refers to an ordered arrangement of at least two cDNAs on a substrate. At least one of the cDNAs represents a control or standard sequence, and the other, a cDNA of diagnostic interest. The arrangement of from about two to about 40,000 cDNAs on the substrate assures that the size and signal intensity of each labeled hybridization complex formed between a cDNA and a sample nucleic acid is individually distinguishable.


[0023] The “complement” of a nucleic acid molecule of the Sequence Listing refers to a nucleotide sequence which is completely complementary over the full length of the sequence and which will hybridize to the nucleic acid molecule under conditions of high stringency.


[0024] A “combination” comprises at least two, alternatively about 10, and up to 292 sequences selected from the group consisting of SEQ ID NOs:1-292 as presented in the Sequence Listing.


[0025] “cDNA” refers to a chain of nucleotides, an isolated polynucleotide, a nucleic acid molecule, or any fragment or complement thereof. It may have originated recombinantly or synthetically, be double-stranded or single-stranded, coding and/or noncoding, an exon with, or without, an intron from a genomic DNA molecule, and purified or combined with carbohydrate, lipids, amino acids or inorganic elements or substances. Preferably, the cDNA is from about 400 to about 12000 nucleotides.


[0026] 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 et al. (1993) 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).


[0027] “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 advantages such as longer lifespan or enhanced activity.


[0028] “Differential expression” refers to an increased or upregulated or a decreased or downregulated expression as detected by absence, presence, or at least 2.5-fold change in the amount of transcribed messenger RNA or translated protein in a sample.


[0029] “Disorder” refers to conditions, diseases or syndromes which fall into the categories of cancer, infectious disease, autoimmunity, allergy, and graft versus host disease.


[0030] “Fragment” refers to a chain of consecutive nucleotides from about 200 to about 700 base pairs in length. Fragments may be used in PCR or hybridization technologies to identify identical or related nucleic acid molecules and in binding assays to screen for a ligand. Nucleic acids and ligands identified in this manner are useful as therapeutics to regulate replication, transcription or translation.


[0031] 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.


[0032] “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 22:46734680), or BLAST2 (Altschul et al. (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.


[0033] “Ligand” refers to any agent, molecule, or compound which will bind specifically to a complementary site on a cDNA molecule or fragment thereof or to a protein or an epitope thereof. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic or organic substances including nucleic acids, amino acids, carbohydrates, fats, and lipids.


[0034] “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.


[0035] “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.


[0036] “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.


[0037] “Probe” refers to a cDNA that hybridizes to at least one nucleic acid molecule in a sample. Where targets are single stranded, probes are complementary single strands. Probes can be labeled with reporter molecules for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.


[0038] “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.


[0039] “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.


[0040] “Sample” is used in its broadest sense as containing nucleic acids, proteins, antibodies, and the like. A sample may comprise a bodily fluid; 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; a fingerprint, buccal cells, skin, or hair; and the like.


[0041] “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.


[0042] “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.


[0043] “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.


[0044] The Invention


[0045] The present invention provides for a combination comprising a plurality of cDNAs or their complements, SEQ ID NOs:1-260, which can be used on a substrate to diagnose, to stage, to treat or to monitor the progression or treatment of a disorder such as cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. These cDNAs represent known and novel genes differentially expressed in DC. The combination is used in its entirety or in part, as subsets of cDNAs downregulated in immature DC, SEQ ID NOs:1-29; cDNAs upregulated in immature DC, SEQ ID NOs:30-241; cDNAs upregulated in mature DC, SEQ ID NOs:242-257; or cDNAs downregulated in mature DC, SEQ ID NOs:258-260. SEQ ID NOs:24-29 represent novel cDNAs downregulated in immature DC; SEQ ID NOs:187-202, 205-213 and 216-241 represent novel cDNAs upregulated in immature DC; SEQ ID NOs:256 and 257 represent novel cDNAs upregulated in mature DC; and SEQ ID NOs:259 and 260 represent novel cDNAs downregulated in mature DC. Since the novel cDNAs were identified solely by their differential expression, it is not essential to know a priori the name, structure, or function of the gene or its encoded protein. The usefulness of the cDNAs exists in their immediate value as diagnostics.


[0046] In one embodiment, the combination is useful to characterize monocyte and dendritic cell populations from subjects with disorders such cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. In a second embodiment, the combination is useful in evaluation of subjects prior to tissue or organ transplant. In a third embodiment, the combination is useful in the diagnosis, progression, treatment or monitoring the treatment of cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. In a fourth embodiment, the combination is useful to enhance or to measure enhancement of dendritic cell function during immunotherapy of cancer or other disorders. In a fifth embodiment, the combination is useful to measure suppression of dendritic cell function during immunotherapy. In a sixth embodiment, the combination is useful to enhance the efficacy of vaccines. In a seventh embodiment, the combination is immobilized on a substrate.


[0047] Table 1 shows cDNAs that are upregulated in dendritic cells greater than three-fold and which have been previously shown in the literature to be expressed in dendritic cells. The types of cDNAs that are upregulated in concert during the transition from monocyte to dendritic cell reflect newly acquired DC functions, such as antigen uptake. Both Fc receptors and complement receptors which bind and endocytose antibody-antigen complexes are upregulated on DC. Heat shock proteins, which assist in the capture and transport of antigen, are upregulated approximately five-fold. Molecules involved in the formation of vesicles during endocytosis are upregulated, such as clathrin and clathrin-associated molecules. Fibronectin, which is upregulated up to 80-fold in dendritic cells, can enhance cell adhesion, migration, and proliferation. A large number of cDNAs encoding proteins that are involved in antigen processing and presentation are also upregulated in dendritic cells. These cDNAs include major histocompatibility complex (MHC) class II cDNAs, which encode proteins that present antigen to T cells; cDNAs encoding invariant chain, which is essential for assembly and antigen binding to MHC class II proteins; and cathepsin cDNAs, which encode proteases that reside in lysosomes and degrade antigen into peptides suitable for binding to MHC proteins. In addition, cDNAs encoding chemokines, which attract other immune cells, such as T cells and eosinophils, are upregulated in dendritic cells. Chemokines such as monocyte chemoattractant proteins 1, 2, and 4; eotaxin; thymus- and activation-regulated chemokine; macrophage inflammatory protein-1 alpha; and pulmonary and activation-regulated chemokine that attract both Th1 and Th2 type cells were upregulated, suggesting that immature DC retain their capacity to initiate different types of immune responses and have not yet acquired specialized functionality.


[0048] A number of cDNAs are downregulated three to 10-fold in DC relative to monocytes as shown in Table 2. Since these cDNAs include those that have been shown to be expressed in monocytes, but not in immature DC, the cDNAs shown in Table 2 probably have roles in monocyte function. Previously described monocyte-specific cDNAs include SEQ ID NO:4 and 5. These are cDNA variants that encode CL100 protein tyrosine phosphatase, which can suppress MAP kinase protection from apoptosis. Monocytes also express a number of cDNAs that are upregulated in cells stimulated with mitogen, such as those that regulate entry into the G0/G1 switch in the cell cycle. These cDNAs include SEQ ID NO:8, which encodes the cellular oncogene cfos, SEQ ID NO:11, which encodes GOS2 protein, and SEQ ID NOs:12 and 13, which are two variant cDNAs that encode a helix-loop-helix phosphoprotein (GOS8) involved in regulating G protein signaling.


[0049] Table 2 also shows cDNAs that have not previously been shown to be upregulated in monocytes. For example, two other phosphatases, SEQ ID NOs:9 and 14, are preferentially expressed in monocytes, and together with SEQ ID NOs:4 and 5 are involved in regulating kinases. Several clones representing pre-B cell enhancing factor (PBEF) and its 3′ UTR are upregulated three to six-fold over expression in dendritic cells (SEQ ID NOs:19-25). PBEF is expressed mainly in the bone marrow, liver, and muscle. Expression in bone marrow suggests involvement in the early differentiation stages of hematopoietic cells. The 3′ region of PBEF cDNA contains multiple TATT motifs, which are RNA destabilization sequences found in other upregulated genes including cfos, some of the genes regulating the G0/G1 switch in the cell cycle, and other cytokines. PBEF plays a role in the innate immune response by monocytes, since a PBEF homolog is upregulated in the innate immune response of sponges upon contact with xenogeneic sponge tissue. In the immune system, 29% of PBEF transcripts are found in libraries derived from activated granulocytes, monocytes, macrophages, and dendritic cells. Another indication that monocytes are involved in the innate immune response is the expression of a cDNA encoding a chemokine, SCM-1 beta (SEQ ID NO:7) that is constitutively expressed in natural killer cells.


[0050] SEQ ID NOs:26-29 are unique cDNAs. SEQ ID NO:26 is upregulated greater than three-fold and encodes a protein about 30% identical to CG9591, an uncharacterized protein expressed in Drosophila melanogaster. SEQ ID NO:26 encodes a protein at least 1019 amino acids long and contains potential protein kinase C and casein kinase II phosphorylation sites and myristylation sites. SEQ ID NOs:27-29 are upregulated between four and six-fold. The occurrence of SEQ ID NO:28 primarily in hemic/immune libraries, 33% of which are derived from macrophages or monocytes, suggests that its function is immune-specific, and in particular, specific to the functions of macrophages and monocytes.


[0051] SEQ ID NOs:30-51 are cDNAs that are upregulated in DC and associated with endocytosis, antigen uptake and presentation, and proteolytic processing. SEQ ID NOs:30-32 are cDNAs upregulated about three to five-fold and encode sorting nexins 1-3, which belong to a family of hydrophilic molecules and are partially associated with the cellular membrane. In the immune system, 50% of sequences matching to SEQ ID NO:32 are found in cells of myeloid origin, including monocytes, dendritic cells, and macrophages. These molecules appear to play a role in intracellular trafficking of molecules to lysosomes where antigen is processed for presentation. SEQ ID NO:35 is upregulated six to nine-fold, and 88% of its expression is found in dendritic cells, macrophages, and the promonocytic cell line THP-1. SEQ ID NO:35 encodes a transmembrane receptor (TREM-1) that is similar to the polymeric immunoglobulin receptor which binds and endocytoses immunoglobulins. Like that receptor, TREM-1 is expressed on neutrophils and monocytes, upregulated upon LPS stimulation, and induces secretion of chemokines and cytokines such as MCP-1,TNF-α, and IL-8. Because of these similarities, TREM-1 appears to act as a receptor for antibody bound to antigen. In addition, TREM-1 associates with a transmembrane protein DAP12, SEQ ID NO:64.


[0052] Molecules other than clathrin that are important in endocytosis and vesicle transport are upregulated in dendritic cells, such as cDNAs that encode myoferlin and VAT1, SEQ ID NOs:38 and 39, respectively. Myoferlin is upregulated approximately three-fold, and within the immune system 44% of sequences are found in monocytes, macrophages, granulocytes, and dendritic cells. Myoferlin regulates membrane fusion which is important in endocytosis of antigen. VAT1 is upregulated approximately four-fold, is expressed ubiquitously, and appears to be important in vesicular transport of amines.


[0053] Several proteases other than cathepsins are upregulated in dendritic cells. Several different types of metalloproteinase cDNAs are upregulated; for example, SEQ ID NOs:36, 40, and 45 are upregulated about 14-fold, three-fold, and three-fold, respectively. Metalloproteinases regulate processing of antigen into peptides for presentation on MHC class I molecules, cleave membrane bound proteins, such as TRANCE, and affect migration and differentiation of dendritic cells. Metalloproteinases are sometimes secreted extracellularly for antigen processing. DC secrete proteases which can process antigen extracellularly into peptides which bind to empty MHC class II molecules on the surface of the cell. Metalloproteinases and other proteases are upregulated during the initial innate immune response as well, wherein proteins on surfaces of pathogens are crosslinked and/or degraded. Other proteases upregulated include SEQ ID NOs:41, 43, and 46-49, which encompass aspartic, cysteine, and other peptidases. SEQ ID NOs:50 and 51 encode angiotensin-converting enzyme, which was upregulated three to six-fold. Dendritic cells also express cathepsin inhibitors in the vesicles in which MHC class II proteins are being formed. SEQ ID NO:44 encodes a cysteine proteinase that inhibits cathepsins L, S, and K, and was originally described as a marker of squamous cell carcinoma. In addition, protease inhibitors prevent host cell degradation by proteases produced by pathogens themselves. SEQ ID NOs:42 and 44 inhibit proteases and appear to function in this manner.


[0054] SEQ ID NOs:52-59 are cDNAs that encode growth factors upregulated in dendritic cells. These factors include those that have been described in other immune functions such as SEQ ID NO:52, which encodes an IL-1 receptor antagonist; SEQ ID NO:57, which encodes an IL-1 receptor homolog preferentially expressed on Th2 cells; and SEQ ID NO:58, which encodes the a chain of the IL-13 receptor. Others not previously associated with immune function are epididymal secretory protein, SEQ ID NOs:53-55 and bone-derived growth factor, SEQ ID NO:59. All of these cDNAs are upregulated from three to nine-fold. Although its cDNA encodes an epididymal-specific protein, SEQ ID NO:53 is a splice variant that contains a region that is preferentially expressed in hemic/ immune libraries, 67% of which are of monocytic, dendritic, macrophage, or granulocytic in origin.


[0055] SEQ ID NOs:60-78 are cDNAs that encode proteins that are upregulated in DC and play a role in signal transduction. SEQ ID NO:61 encodes Notch, which is upregulated about 10-fold and is a receptor involved in growth and differentiation in a wide variety of cells including T lymphocytes. Both kinases and phosphatases are upregulated in dendritic cells; for example, B lymphocyte serine/threonine protein kinase, SEQ ID NO:71, which transduces signals from CD40 and is upregulated 30-fold, and a G-protein coupled receptor (GRK5), SEQ ID NO:72, which is upregulated 20-fold. SEQ ID NO:77 encodes doc-1, an S-phase regulator and tumor suppressor in oral cancer that may induce apoptosis. A related sequence, SEQ ID NO:78 encodes doc-2, another tumor suppressor phosphoprotein that contains a potential transmembrane sequence at amino acids 493 to 513. SEQ ID NO:64 is upregulated about four-fold in dendritic cells, and in the immune system 40% of its expression is found in dendritic cells and granulocytes. SEQ ID NO:64 encodes DAP12, a receptor that has homology to the CD3 zeta chain of the T cell receptor complex and may trigger maturation and activation of dendritic cells through antigen uptake by TREM-1.


[0056] SEQ ID NOs:79-98 are cDNAs that are upregulated in DC and encode molecules involved in DNA transcription. In particular, about 40% of upregulated sequences involved in DNA transcription encode zinc finger proteins. SEQ ID NO:90 is upregulated about 30-fold and encodes a zinc finger homeobox protein. SEQ ID NO:94 is upregulated from about three to about six-fold and encodes a breast cancer tumor suppressor gene, bcsc-1, which has two potential transmembrane regions at amino acids 224 to 242 and 370 to 389. Bcsc-1 has three potential glycosylation sites, 7 potential casein kinase II phosphorylation sites, 7 potential protein kinase C phosphorylation sites, and 4 potential myristylation sites.


[0057] SEQ ID NOs:99-143 are sequences upregulated in DC that encode molecules involved in a variety of pathways. SEQ ID NOs:99-101 encode proteins involved in iron metabolism. SEQ ID NOs:102 and 103 encode transmembrane proteins involved in lipid metabolism. SEQ ID NO:104, which encodes apolipoprotein E, is upregulated five to 19-fold. SEQ ID NOs:105 and 106, which encode apolipoprotein C-I, are upregulated three to five-fold and 38% of its immune tissue expression is found in monocytes, dendritic cells, macrophages, and granulocytes. Apolipoprotein E is secreted by macrophages to dispose of cholesterol and other lipids. During the innate immune response, lipoprotein components from pathogens are among the first antigens encountered by dendritic cells and macrophages. Scavenger receptors take up these lipoproteins, which are then processed for antigen presentation. Byproducts such as cholesterol and other lipids are then expelled through transporters, such as the ABCA1 transporter expressed on macrophages. Other cDNAs that encode proteins that export molecules detrimental to the host cell and that are upregulated in dendritic cells include the multidrug resistance associated protein 3, SEQ ID NO:132, which is upregulated four to five-fold. In the immune system, SEQ ID NO:132 has an abundance of 45% in macrophages and LPS-activated monocytes. A number of other cDNAs encoding transporters are upregulated three to five-fold in dendritic cells; these include SEQ ID NOs:133-138, which encode anion transporters, and SEQ ID NOs:140 and 142, which encode chloride channels. Other molecules that protect dendritic cells during immune response are SEQ ID NOs:120-128, which encode the vacuolar H+ATPase subunits which are upregulated three to 12-fold in dendritic cells. Recent studies have shown that ATPase is upregulated during the differentiation of monocytes into macrophages and can serve to downregulate production of inflammatory cytokines such as TNF-α and IL-1 thus preventing deleterious effects of these cytokines on the host. SEQ ID NOs:129 and 130 encode glutathione-S-transferase homolog and glutathione-S-transferase, respectively, proteins which protect cells from oxidative stress and toxic foreign chemicals produced by pathogens. Upregulation of SEQ ID NOs:129 and 130 in DC may prevent damage from production of pro-inflammatory cytokines.


[0058] A number of enzymes and other cytosolic and lysosomal proteins involved in lipid metabolism are upregulated and preferentially expressed in libraries made from monocytes, dendritic cells, macrophages, and granulocytes. These molecules include SEQ ID NOs:107, which encodes a lipoprotein lipase upregulated four to six-fold; and SEQ ID NOs:113-115, which encode fatty acid binding protein and fatty acid binding protein homolog and are upregulated 10 to 30-fold. More than 40% of the immune system expression of SEQ ID NO:116, which encodes lysosomal acid lipase, occurs in monocytes, dendritic cells, macrophages, and granulocytes. SEQ ID NO:118, which encodes palmitoyl-protein thioesterase, is upregulated four to five fold and exhibits 17% abundance in immune cells, 40% of which are monocytes, dendritic cells, macrophages, and granulocytes. These molecules, which metabolize lipids of pathogenic origin, appear to help in the process of remodeling the dendritic plasma membrane and membranes of other lysosomal and other endocytic compartments.


[0059] SEQ ID NOs:144-151 are cDNAs that encode surface or secreted proteins that have a role in adhesion. SEQ ID NOs:145 and 146 encode osteopontin which is upregulated 11 to 29-fold and 56% of immune system expression occurs in activated DC, macrophages, and THP-1 cells. Osteopontin binds to the cell adhesion molecule CD44 and is induced by GM-CSF. Another molecule that may be involved in cell to cell contact is SEQ ID NO:144 which encodes kerato-epithelin and is upregulated about 10-fold. SEQ ID NO:147 is upregulated more than five-fold, expressed most abundantly in dendritic cells, macrophages, and T cells, and encodes a 14 kD lectin which chemoattracts monocytes and macrophages. SEQ ID NO:148 is upregulated 14.4-fold and encodes a member of the claudin protein family which plays a role in adhesion of cells through tight junctions.


[0060] SEQ ID NOs:152-165 are cDNAs upregulated in DC that encode molecules involved in cellular structure. As monocytes mature into DC, profound morphological changes occur that allow increased mobility and the formation of dendritic projections. These changes result in an increased surface area, thereby enhancing uptake of antigen. cDNAs, which are upregulated in dendritic cells and affect structure, encode α tubulin (SEQ ID NOs:152 and 157); vimentin (SEQ ID NO:153); gamma-actin (SEQ ID NOs:155 and 156); and beta-actin (SEQ ID NO:163). EB1 (SEQ ID NO:165) is upregulated almost four-fold and associates with microtubulues and adenomatous polyposis coli (APC) protein, which shuttles beta-catenin from the cytosol to the nucleus. APC is important in signaling through Wnt, a soluble protein that influences cell adhesion, growth, and differentiation.


[0061] SEQ ID NOs:166-176 are cDNAs upregulated in DC that encode molecules which regulate the innate immune response. SEQ ID NO:171 encodes Toll-like receptor 1 (TLR-1), a member of the IL-1 receptor superfamily thought to transduce signals that upregulate costimulatory molecules and cytokines. Immune responses to LPS and bacteria require certain Toll receptors, although the role of TLR-1 has not previously been identified. In Drosophila, the putative Toll receptor ligand is the product of a protease cascade. Other cDNAs that may participate in the innate immune response include SEQ ID NOs:172, 174, 175, and 176, which encode α-2-macroglobulin, factor XIIIa, transglutaminase, and thrombomodulin, respectively. α-2-macroglobulin binds proteases, inhibiting their activity and preventing damage from pathogenic proteases. Transglutaminase and factor XIIIa may participate in the crosslinking of proteins present on pathogens, enhancing phagocytosis. Thrombomodulin is a receptor for thrombin, which has both pro- and anticoagulant activities.


[0062] SEQ ID NOs:177-182 are cDNAs that are upregulated in DC and expressed preferentially in the immune system. SEQ ID NO:177, which encodes a transmembrane protein, DORA, is upregulated 14.5-fold, and 50% of its expression occurs in DC, macrophages, and granulocytes. SEQ ID NO:178, which encodes a membrane-associated protein, is upregulated almost five-fold, and 18% of its expression occurs in the immune system. SEQ ID NO:179, which encodes CD52, a glycosylated protein associated with the membrane, is upregulated from about three to about four-fold, and 35% of its expression occurs in the immune system. SEQ ID NOs:180 and 181, which encode variants of the tetraspanin CD53, are upregulated three to 20-fold, and 30% of their immune system expression occurs in macrophages, activated monocytes, dendritic cells, and granulocytes. SEQ ID NO:182 which encodes PIL protein is upregulated about four-fold; transcript analysis indicates that PIL protein is expressed in both dendritic cells and T cells.


[0063] SEQ ID NOs:183-200 are cDNAs that are upregulated in DC and encode transmembrane proteins. In particular, SEQ ID NOs:186 and 187 are upregulated about six-fold and about three-fold, respectively, and encode proteins that have signal peptides at the N-terminus. SEQ ID NO:189 is upregulated about four-fold and encodes a protein whose expression is preferentially found in dendritic cells and macrophages. SEQ ID NO:196, which is upregulated about four-fold, encodes a protein that has seven predicted transmembrane regions, and 44% of transcripts in the immune system are found in monocytes, macrophages, dendritic cells and granulocytes.


[0064] SEQ ID NOs:201-215 are cDNAs that are upregulated in DC and encode homologs of proteins with known function. SEQ ID NOs:201 and 202 encode variants of HU-K4 which has similarity to a phospholipase-D protein from Vaccinia virus. SEQ ID NO:202 is preferentially expressed in dendritic cells, and SEQ ID NO:201 is preferentially expressed in monocytes, dendritic cells, macrophages, and granulocytes. Several cDNAs encode homologs of proteins involved in immune response. SEQ ID NOs:203 and 204 encode a molecule that has approximately 35% identity to the IL-17 receptor, and 16% of their transcripts are found in the immune system. SEQ ID NO:205 encodes a protein similar to murine ClqC chain, a component of the complement system. SEQ ID NO:211 encodes a protein similar to a murine zinc finger protein and is upregulated 22.7-fold. Some 19% of SEQ ID NO:211 transcripts are found in hemic/immune tissues, preferentially macrophages. SEQ ID NO:212 encodes a novel GATA zinc finger transcription factor, with 65% identity to the murine GATA-5 cardiac transcription factor.


[0065] SEQ ID NOs:215-225 are cDNAs that are upregulated in DC and encode homologs of proteins of unknown function that are upregulated three to five-fold. SEQ ID NO:216 encodes a protein that contains seven potential transmembrane regions. SEQ ID NO:218 encodes a protein with eight potential transmembrane regions, and SEQ ID NO:220 encodes a protein with one predicted transmembrane region. SEQ ID NO:221 encodes a protein that has 12 potential transmembrane regions and is expressed preferentially in monocytes, dendritic cells, macrophages, and granulocytes. In addition, over 14% of transcripts of SEQ ID NOs:219 and 223-225 occurs in immune tissues. Over 30% of transcripts of SEQ ID NOs:219 and 221-225 in the immune system occurs in monocytes, dendritic cells, macrophages, and granulocytes.


[0066] SEQ ID NOs:226-241 are unique cDNAs upregulated in DC from three to 16-fold and contain potential open reading frames. Of these, SEQ ID NOs:228, 229, 233, 237, and 238 have a specific abundance ranging from 14 to 30% in hemic and immune tissues, while SEQ ID NOs:227 and 228 have a specific abundance in the immune system of 36% and 41%, respectively, in monocytes, dendritic cells, macrophages, and granulocytes. SEQ ID NOs:226, 230, and 239 are expressed 15.9-fold, 13.3-fold, and 12.7-fold, respectively, in DC over that of monocytes.


[0067] Table 4 lists cDNAs that are upregulated by mature DC. Survival and function of DC may be enhanced by factors such as SEQ ID NOs:242 and 243, which are upregulated three-fold and encode variants of IL-1 beta, a cytokine involved in regulating Th1 type immune responses; SEQ ID NO:249 which is upregulated between eight and 12-fold and encodes IL-8; SEQ ID NOs:250 and 251, which are upregulated from about three to about five-fold and encode variants of small inducible cytokine A4, a chemokine similar to macrophage inflammatory protein-1 beta. Survival may also be enhanced by upregulation by three-fold of SEQ ID NO:253, which encodes an inhibitor of apoptosis protein, MIHC. SEQ ID NOs:244 and 245 encode CD9, a tetraspanin which is upregulated from about three to about five-fold and interacts with the integrin family and other membrane proteins. Upregulation of CD9 may enhance DC cell migration and adhesion. SEQ ID NO:248 encodes I-TRAF, which interacts with TRAF, a necessary component for transducing signals from the TRANCE receptor. In addition, two cDNAs are unique, SEQ ID NO:256 and SEQ ID NO:257, which are upregulated about four-fold and three-fold, respectively.


[0068] Table 5 lists cDNAs that are downregulated by mature DC. SEQ ID NO:258 encodes neurogranin, which has no known immune function. Although preferentially expressed in the nervous system, neurogranin is expressed in a number of immune tissues, including dendritic cells, eosinophils, B cells, T cells, and the promonocyte line THP-1. SEQ ID NOs:259 and 260 encode two unique proteins, each downregulated about three-fold, which are similar to a zinc finger protein and ribonucleoprotein, respectively.


[0069] The cDNAs of the invention define a differential expression pattern against which to compare the expression pattern of biopsied and/or in vitro treated dendritic cells. 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.


[0070] The combination may be arranged on a substrate and hybridized with dendritic cells from subjects with diagnosed disorders to identify those sequences which are differentially expressed in dendritic cells from normal subjects versus dendritic cells from diseased subjects. 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.


[0071] In another embodiment, the combination can be used for large scale genetic or gene expression analysis of a large number of novel, nucleic acid molecules. 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 acid molecules are hybridized to the combination for the purpose of defining a novel gene profile associated with that developmental stage, treatment, or disorder.


[0072] cDNAs and Their Uses


[0073] 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.


[0074] 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.


[0075] 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 WO95/25 1116). 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.


[0076] 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.


[0077] 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 autoimmune disease or cancer, following a treatment of cancer with in vitro manipulated dendritic cells, following treatment of autoimmune disease with agents that suppress dendritic cell function, or inhibiting or inactivating a therapeutically relevant gene related to the cDNA.


[0078] 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.


[0079] Hybridization


[0080] 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 Pharmacia Biotech (APB), Piscataway N.J.), fluorescent (Operon Technologies, Alameda Calif.), and chemiluminescent labeling (Promega, Madison Wis.) are well known in the art.


[0081] 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.


[0082] 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 acid molecules 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).


[0083] 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.)


[0084] Screening and Purification Assays


[0085] 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.


[0086] 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.


[0087] 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.


[0088] 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.


[0089] Protein Production and Uses


[0090] The full length cDNAs or fragment thereof may be used to produce purified proteins using recombinant DNA technologies described herein and taught in Ausubel et al. (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.


[0091] 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.), MACVECTOR software (Genetics Computer Group, Madison Wis.) and RasMol software (Roger Sayle, 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.


[0092] Expression of Encoded Proteins


[0093] 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 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 residues of linker, and the protein encoded by the cDNA.


[0094] 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.


[0095] 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.


[0096] 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).


[0097] 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 include 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.


[0098] 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 ABI 431A peptide synthesizer (Applied Biosystems, 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.


[0099] Screening and Purification Assays


[0100] A protein or a portion thereof encoded by the cDNA may be used to screen a library or 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 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 DNA, RNA, or PNA molecules, agonists, antagonists, antibodies, immunoglobulins, inhibitors, peptides, pharmaceutical agents, proteins, drugs, 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.


[0101] 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, 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.


[0102] Production of Antibodies


[0103] 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 antigenically-effective portion 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.


[0104] 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.).


[0105] 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.


[0106] Labeling of Molecules for Assay


[0107] 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.).


[0108] 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. No. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.


[0109] Diagnostics


[0110] 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 or differential expression include cancer, infectious disease, autoimmunity, allergy, and graft versus host disease. 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.


[0111] 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.


[0112] 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 substantially 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.


[0113] 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.


[0114] Gene Expression Profiles


[0115] 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 sequences in a sample. The cDNAs of the invention are used as elements on a microarray to analyze gene expression profiles. In one embodiment, the microarray is used to monitor the progression of disease. Researchers can assess and 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 microarray 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.


[0116] 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 microarrays to establish and then follow expression profiles over time. In addition, microarrays 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.


[0117] Assays Using Antibodies


[0118] 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. The antibodies may be used with or without modification, and labeled by joining them, either covalently or noncovalently, with a labeling moiety.


[0119] Protocols for detecting and measuring protein expression using either polyclonal or monoclonal antibodies are well known in the art. Examples include ELISA, RIA, and fluorescent activated cell sorting (FACS). 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). The method may employ a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes, or a competitive binding assay. (See, e.g., Coligan et al. (1997) Current Protocols in Immunology, Wiley-Interscience, New York N.Y.; Pound, supra)


[0120] Therapeutics


[0121] 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(34): 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 Pharmacology, Vol. 40), Academic Press, San Diego Calif.).


[0122] 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 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.


[0123] Molecules which regulate the activity of the cDNA or encoded protein are useful as therapeutics for cancer, infectious disease, autoimmunity, allergy, and graft versus host disease and for enhancing vaccines. 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.


[0124] 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.).


[0125] Model Systems


[0126] 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.


[0127] Transgenic Animal Models


[0128] 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. Nos. 5,175,383 and 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.


[0129] Embryonic Stem Cells


[0130] 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.


[0131] Knockout Analysis


[0132] 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.


[0133] Knockin Analysis


[0134] 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.


[0135] 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. 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


I Construction of cDNA Libraries

[0136] 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 (Life Technologies, Rockville Md.). 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.


[0137] 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.).


[0138] 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 (Life Technologies) 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 (Life Technologies), or pinch plasmid (Incite Genomic, Palo Alto Calif.). 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 (Life Technologies).


[0139] 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, Nucl 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 et al., supra).


[0140] II Isolation and Sequencing of cDNA Clones


[0141] 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.


[0142] 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).


[0143] cDNA sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 thermal cycler (Applied Biosystems) 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 ABI sequencing kits such as the ABI PRISM BIGDYE cycle sequencing kit (Applied Biosystems). Electrophoretic separation of cDNA sequencing reactions and detection of labeled cDNAs were carried out using the MEGABACE 1000 DNA sequencing system (APB); the ABI PRISM 373 or 377 sequencing systems (Applied Biosystems) in conjunction with standard ABI 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).


[0144] III Extension of cDNA Sequences


[0145] 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.


[0146] 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.


[0147] 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 nmol of each primer, reaction buffer containing Mg2+, (NH4)2SO4, and β-mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incite Genomic): 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.


[0148] 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.


[0149] 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.


[0150] 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 ABI PRISM BIGDYE terminator cycle sequencing kit (Applied Biosystems).


[0151] IV Assembly and Analysis of Sequences


[0152] 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 (Incite Genomic), 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 37:417424).


[0153] 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 (supra); Altschul et al. (supra); Karlin et al. (1988) Proc Natl Acad Sci 85:841-845), BLASTn (vers.1.4, WashU), and CROSSMATCH software (Phil 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 (Phil Green, supra). Bins with several overlapping component sequences were assembled using DEEP PHRAP (Phil Green, supra).


[0154] 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 (Incite Genomic).


[0155] 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” are 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 (Incite Genomic).


[0156] 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 (1981) J Mol Biol 147:195-197), 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.


[0157] V Selection of Sequences, Microarray Preparation and Use


[0158] Incite clones represent template sequences derived from the LIFESEQ GOLD assembled human sequence database (Incite Genomic). 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 GENEALBUM GEM series 1-6 microarrays (Incite Genomic) contain 52,616 array elements which represent 17,472 annotated clusters and 35,144 unannotated clusters. The LIFEGEM 1.0 microarray (Incite Genomic) contains 9,203 array elements which represent 4,791 annotated genes and 1,861 unannotated clusters. For the UNIGEM series microarrays (Incite Genomic), Incite 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 microarray (Incite Genomic) contains 9,182 array elements which represent 5,815 annotated genes and 2,699 unannotated clusters. UNIGEM V 2.0 microarray (Incite Genomic) contains 7075 array elements which represent 4610 annotated genes and 2,184 unannotated clusters. The HUMAN GENOME GEM series 1 microarray (Incite Genomic) contains 9,766 array elements which represent 7,612 annotated clusters and 1,382 unannotated clusters. Tables 2-5 show the GenBank annotations for SEQ ID NOs:1-260 of this invention as produced by BLAST analysis. LIFEGEM 1.0, GENEALBUM 1-6, and UNIGEM V microarrays were used to compare cDNA expression between monocytes and immature DC. LIFEGEM 1.0, UNIGEM V 2.0, and HUMAN GENOME GEM 1 were used to compare cDNA expression between mature DC and TRANCE-treated mature DC.


[0159] 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.


[0160] 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.


[0161] VI Preparation of Samples


[0162] Preparation of Monocytes and Immature Dendritic Cells


[0163] Peripheral blood mononuclear cells (PBMCs) were isolated from freshly obtained peripheral blood of four healthy donors by centrifugation of the lymphocyte enriched blood fraction over a HYPAQUE ficoll gradient (Sigma-Aldrich). The PBMCs were allowed to adhere to plastic in Iscove's Modified Dulbecco's Medium supplemented with 10% fetal bovine serum, 200 nM glutamine, and 200 nM each penicillin and streptomycin for 4 hours to separate monocytes from other nonadherent cells. One-half of the monocytes was cultured for seven days in 10 ng/ml GM-CSF (Peprotech, Rocky Hill N.J.), and the other half was cultured for seven days with 10 ng/ml GM-CSF and 10 ng/ml IL-4 (Peprotech) to produce immature dendritic cells.


[0164] Preparation of Mature Dendritic Cells


[0165] Monocytes were isolated as described above and were incubated with 10 ng/ml GM-CSF and 10 ng/ml IL-4 for 13 days to generate immature DC. The DC were activated with anti-CD40 (Biodesign International, Kennebunk Me.) at 10 ng/ml for 24 hours. The DC were divided into half, and each half further divided into thirds. Each third was cultured with soluble human TRANCE protein (Peprotech Inc.) at 10 ng/ml for 2, 8, and 24 hours, respectively, or cultured without TRANCE for 2, 8, and 24 hours, respectively.


[0166] Isolation and Labeling of Sample cDNAs


[0167] Cells were harvested and lysed in 1 ml of TRIZOL reagent (5×106 cells/ml; Life Technologies). The lysates were vortexed thoroughly, 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.


[0168] 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.


[0169] 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 (Incite Genomic). 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 mRNAs (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.


[0170] cDNAs were purified using two successive CHROMA SPIN 30 gel filtration spin columns (Clontech). Cy3- and Cy5-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 and CyS labeled 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 Cy5. 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 CyS. 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 (Incite Genomic). 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 a 2.5-fold change in expression at one or more time points, 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 (Incite Genomic). Differential expression is reported as the balanced differential expression value for each clone representing a gene on the microarray. The cDNAs that are differentially expressed are shown in Tables 2-5. Table 2 identifies downregulated cDNAs in immature DC. Table 3 identifies upregulated cDNAs in immature DC. Tables 4 and 5 identify cDNAs upregulated or downregulated, respectively, in mature DC. The cDNAs are identified by their SEQ ID NO and TEMPLATE ID, and by the description associated with at least a fragment of a polynucleotide found in GenBank. The descriptions were obtained using the sequences of the Sequence Listing and BLAST analysis.


[0180] IX Other Hybridization Technologies and Analyses


[0181] 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.


[0182] 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).


[0183] 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.


[0184] 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.


[0185] 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.


[0186] X Further Characterization of Differentially Expressed cDNAs and Proteins


[0187] Clones were blasted against the LIFESEQ Gold 5.1 database (Incite Genomic) and an Incite template and its sequence variants were chosen for each clone. 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.


[0188] Percent sequence identity can be determined electronically for two or more amino acid or nucleic acid sequences using the MEGALIGN program (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.


[0189] 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 et al.(supra); Attwood et al. (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.


[0190] 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.


[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. 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. Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of transcription.


[0193] 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.).


[0194] XII Production of Specific Antibodies


[0195] 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.


[0196] In another approach, the amino acid sequence translated from a cDNA of the invention is analyzed using PROTEAN software (DNASTAR) to determine regions of high antigenicity, essentially antigenically-effective epitopes 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 ABI 431 peptide synthesizer (Applied Biosystems) 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.


[0197] 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.


[0198] Such clones are expanded and subjected to 2 cycles of cloning at I 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.


[0199] XIII Purification of Naturally Occurring Protein Using Specific Antibodies


[0200] Naturally occurring or recombinant protein is substantially 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.


[0201] XIV Screening Molecules for Specific Binding with the cDNA or Protein


[0202] 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 Burbaum et al. U.S. Pat. No. 5,876,946.


[0203] 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 1GENBANK IDDESCRIPTIONBDEg1006656H. sapiens mRNA for cathepsin C.19.3, 17.0, 16.7, 17.1, 14.3g1262337H. sapiens MB1 gene.3.9, 6.7g1280140Human eotaxin precursor mRNA, complete cds.5.2, 22.6, 6.2, 4.5, 4.4g1373145Human clathrin assembly protein lymphoid myeloid leukemia3.7g1434908Homo sapiens MHC (HLA) DRB intron 1 DNA, partial sequence.3.4g1536878Human mRNA for chemokine, complete cds.5.7, 3.8, 5.3g178535CD13; Human aminopeptidase N3.9g179338Human HLA-B-associated transcript 2 (BAT2) mRNA, complete6.1g179640Human complement C1q B-chain gene, exon A + 1.15.2, 11.1, 8.8, 16.9, 5.3g180055Human thymocyte antigen CD1b mRNA, complete cds.5.3, 3.1, 3.2g180112Human antigen CD36 (clone 21) mRNA3.3g180149Human transmembrane protein (CD59) gene, exon 4.12.9g180154Human lysosomal membrane glycoprotein CD63 mRNA.4.0, 10.4g181179Human cathepsin D mRNA, complete cds.6.7, 4.6, 9.6g182489Human Fc-epsilon-receptor gamma-chain gene, complete cds.4.7, 4.2, 3.6g182598Human IgG low affinity Fc fragment receptor (FcRIIb3) mRNA,4.2g183089Human low-affinity Fc-receptor IIB gene, exons 4-7.3.1g184422Human 90 kD heat shock protein gene, complete cds.5.2g184851Human low affinity IgG receptor CD16 (FcGRIII) mRNA, complete3.5, 4.5g186927Homo sapiens lysosomal membrane glycoprotein-1 (LAMP1) mRNA7.2g187054Human lymphocyte clathrin light-chain A mRNA, complete cds.3.9g187284Human Mac-1 gene encoding complement receptor type 3, CD11b,5.1, 4.6, 3.2, 3.1g187434Human monocyte chemotactic and activating factor (MCAF)11.1g187447Human monocyte chemotactic protein gene, complete cds.15.2, 8.8, 7.3, 12.1, 9.8g188114Human MHC class II HLA-DQ-beta (DQB1, DQw9), complete cds.3.3, 3.3g188134Human MHC class II DQ alpha mRNA, complete cds.6.6, 4.9, 7.1g188136Human MHC class II HLA-DQ-alpha (DR2-DQw1/DR4 DQw3) mRNA,3.4, 3.8g188217Human MHC class II HLA-DR4-beta (DR4,w4) gene, exon 3,4,5,6.4.2, 4.8g188285Human MHC class II HLA-DRB4-null beta chain pseudogene4.0g188352Human MHC class II HLA-DRw53-associated glycoprotein4.0g188427Human HLA-DR alpha-chain (chain-p34) gene, exons 2, 3, 45.5g1905800H. sapiens MCP-2 gene.3.7, 3.3, 7.3g219907Human LD78 beta gene.4.9g264772thymosin beta-10 [human, metastatic melanoma cell line,3.4g288396H. sapiens NC28 mRNA for monocyte chemoattractant protein3.2g291887Homo sapiens cathepsin B mRNA, complete cds.31.5, 44.0, 22.1, 5.3, 10.6g291926Homo sapiens cystatin B mRNA, complete cds.5.2g29571Homo sapiens Hsc 70 pseudogene.6.9, 10.6g29709Human mRNA for cathepsin H (EC 3.4.22.16).3.2, 4.2, 3.2g29714Human mRNA for pro-cathepsin L (major excreted protein MEP).19.9, 34.8, 29.3, 23.5, 4.3g29850Human CDw40 mRNA for nerve growth factor receptor-related4.1, 3.9, 5.0g3002790Homo sapiens macrophage receptor MARCO mRNA, complete cds.4.2, 6.0, 7.7g30257human CST3 gene for cystatin C.3.9, 3.7, 3.5g30263H. sapiens CST4 gene for Cystatin D.3.6, 3.2, 3.9g30884Human mRNA for HLA class II DR-beta 1 (Dw14).3.8g3107914Homo sapiens mRNA for cathepsin V, complete cds.3.8, 4.5, 16.0g312143H. sapiens mRNA for M130 antigen cytoplasmic variant 1.3.4, 3.1g31335Human FcRII mRNA for immunoglobulin G receptor.3.6g31396Human mRNA for fibronectin (FN precursor).10.0, 76.4, 3.2, 15.0, 81.3,9.6, 19.7, 28.9, 5.6, 27.9g32210Human mRNA for HLA class II DR-beta (HLA-DR B).4.3, 5.5g32291Human mRNA fragment for class II histocompatibility antigen4.0g339688Human thymosin beta-4 mRNA, complete cds.3.8, 4.6g34511Membrane cofactor protein part of complement system3.1g34513H. sapiens mRNA for monocyte chemoattractant protein 111.9, 11.3g34648Human gene for class II invariant gamma-chain (exon 2-8).4.4, 4.2, 4.1g35221Human heat-shock protein HSP70B' gene.3.3g3618344Homo sapiens mRNA for 26S proteasome subunit p283.3g37260human tra1 mRNA for human homologue of7.7, 7.0murine tumor rejection proteing3776070histone deacetylase-like protein and complement component C23.6, 3.9, 3.8, 3.5g3869137Homo sapiens gene for CC chemokine PARC precursor26.4g3928171Human mRNA for beta-Fc-gamma receptor II4.1g4038732Homo sapiens mRNA for beta 2-microglobulin, complete cds3.1, 3.4g4097420Human monocyte chemotactic protein-4 precursor (MCP-4)11.2g439838Human CD86 antigen mRNA, complete cds.3.6g487829H. sapiens leukocyte adhesion glycoprotein p150,95 mRNA (CD11c)3.5, 4.1g5478221Homo sapiens MHC class II antigen (HLA-DRB1) mRNA,4.3, 7.9g557701Human HLA-DMB mRNA, complete cds.3.2g565646Human mRNA for proteasome subunit HsC10-II, complete cds.3.7g565648Human mRNA for proteasome subunit HsC7-I, complete cds.3.5g5725511Homo sapiens beta-2 microglobulin gene, complete cds.3.3g6288733Homo sapiens chemokine-like factor 1 (CKLF1) mRNA,3.7g6456875Home sapiens thymic stroma chemokine-1 precursor, mRNA,5.3g6467379Homo sapiens cathepsin Z precursor (CTSZ) mRNA, complete9.0g662840Human heat shock protein 27 (HSPB1) gene exons 1-3, complete3.8g6630853Homo sapiens chemokine-like factor 2 (CKLF2) mRNA,4.3g6996445Homo sapiens p10 gene for chaperonin 10 (Hsp10 protein) and5.0g703088Homo sapiens MHC class II DPw3-alpha-1 chain mRNA, complete6.4, 6.0, 5.3g971269Human mRNA for proteasome subunit p40 / Mov34 protein,3.1g188675Human mannose receptor mRNA, complete cds.47.6, 15.8


[0204]

2











TABLE 2








SEQ








ID NO
CLONE ID
TEMPLATE ID
GENBANK ID
DESCRIPTION
DATABASE
BDE





















1
1728696
992917.38
g762940
apoferritin H subunit
genpept
−4.4


2
3486093
977676.22
g6103358
smoothelin-B
genpept
−8.9


3
1563184
891492.1
g1911529
ficolin [human, uterus, mRNA, 1736 nt].
gbpri
−4.7


3
1716583
891492.1
g1911529
ficolin [human, uterus, mRNA, 1736 nt].
gbpri
−10.1


4
379425
990840.1
g29980


H. sapiens
CL 100 mRNA for protein tyrosine phosphatase

gbpri
−4.4


5
2232685
990840.12
g29980


H. sapiens
CL 100 mRNA for protein tyrosine phosphatase

gbpri
−4.1


4
2232685
990840.1
g29980


H. sapiens
CL 100 mRNA for protein tyrosine phosphatase



6
1900285
021899.7
g401762


Homo sapiens
ataxia-telangiectasia group D-associated protein

gbpri
−3.4


7
292810
203351.6
g1754608


Homo sapiens
DNA for SCM-1 beta precursor, complete cds.

gbpri
−3.2


8
341491
899156.22
g29903
Human cellular oncogene c-fos
gbpri
−5, −5.4, −5


9
1425456
013003.2
g5138994


Homo sapiens
mRNA for dual specificity phosphatase MKP-5

gbpri
−4.1


10
659029
237649.2
g6942243


Homo sapiens
synaptic glycoprotein SC2 (SC2) mRNA

gbpri
−7.8


11
1217764
269725.3
g609453
Human G0S2 protein gene, complete cds.
gbpri
−3.6


12
2850723
989992.6
g292036
Human helix-loop-helix basic phosphoprotein (G0S8) gene,
gbpri
−3.3


13
2850723
989992.1
g292036
Human helix-loop-helix basic phosphoprotein (G0S8) gene,


14
200836
246772.4
g35791
Human HPTP epsilon mRNA for protein tyrosine phosphatase
gbpri
−4


15
2197952
1327183.6
g2828536
Human Hsp27 ERE-TATA-binding protein (HET) mRNA
gbpri
−4.9


16
1958560
127112.53
g292833
Human TR3 orphan receptor mRNA, complete cds.
gbpri
−3.4


17
1958560
127112.19


18
1506061
336965.2
g186410
Human inhibin A-subunit mRNA, complete cds.
gbpri
−5.2


19
2172334
253946.17
g186353
Human membrane glycoprotein gp130 mRNA, complete cds.
gbpri
−5.1


20
1713166
284616.2
g219868
Human mRNA for HM89.
gbpri
−3.3


21
1640312
401269.21
g404012
Human pre-B cell enhancing factor (PBEF)
gbpri
−5.7


22
3238201
401269.14
g404012
Human pre-B cell enhancing factor (PBEF)
gbpri
−3.1, −3.3


21
3238201
401269.21
g404012
Human pre-B cell enhancing factor (PBEF)


23
3280041
401269.5
g404012
Human pre-B cell enhancing factor (PBEF)
gbpri
−5.7


21
3280041
401269.21
g404012
Human pre-B cell enhancing factor (PBEF)


21
2964314
401269.21
g404012
Human pre-B cell enhancing factor (PBEF)


24
2964314
221172.2
g5523832
Incyte Unique
gbpri
−3.8


25
3073089
055554.1
g5523832
Incyte Unique
gbpri
−3.5


26
1360375
331567.1

Incyte Unique
genpept
−3.4


27
702933
010742.1

Incyte Unique
gbpri
−4


28
1562722
202426.1

Incyte Unique
gbpri
−5.7


28
1871721
202426.1

Incyte Unique
gbpri
−5.3


29
3486126
132211.1

Incyte Unique
gbpri
−4.3










[0205]

3












TABLE 3








SEQ









ID

TEMPLATE
GENBANK

DATA-


NO
CLONE ID
ID
ID
DESCRIPTION
BASE
BDE
E-VALUE






















30
2456671
018414.15
g1293679
Human sorting nexin 1 (SNX1) mRNA, complete cds.
gbpri
4.0



31
1901407
480348.17
g3152937


Homo sapiens
sorting nexin 2 (SNX2) mRNA, complete cds.

gbpri
3.7


32
1659647
1135462.6
g3127052


Homo sapiens
sorting nexin 3 (SNX3) mRNA, complete cds.

gbpri
4.5


33
2607957
1094479.5
g1145690
Human E2 ubiquitin conjugating enzyme UbcH5C
gbpri
35.2


34
2762981
155345.4
g4335940


Homo sapiens
leucine aminopeptidase mRNA, complete cds.

gbpri
6.1


35
1661952
198893.4
g7800156
Home sapiens triggering receptor expressed on myeloid cells 2
gbpri
8.8


35
2630138
198893.4
g7800156


Homo sapiens
triggering receptor expressed

gbpri
6.0






on myeloid cells 2


35
2848362
198893.4
g7800156


Homo sapiens
triggering receptor expressed

gbpri
6.2






on myeloid cells 2


36
1452045
200569.1
g435969
Human metalloproteinase (HME) mRNA, complete cds.
gbpri
27.2


36
1553128
200569.1
g435969
Human metalloproteinase (HME) mRNA, complete cds.
gbpri
13.8


36
1563275
200569.1
g435969
Human metalloproteinase (HME) mRNA, complete cds.
gbpri
11.5


37
2240033
233706.3
g2199511


Homo sapiens
beta-3A-adaptin subunit of the AP-3 complex

gbpri
3.6


38
1975430
234729.3
g6731234
Home sapiens myoferlin (MYOF) mRNA
gbpri
3.4


38
2642654
234729.3
g6731234
Home sapiens myoferlin (MYOF) mRNA
gbpri
3.2


39
2060308
235152.9
g1698398
VAT1, an abundant membrane protein of cholinergic
gbpri
4.4, 3.9






synaptic vesicles


40
1528052
238213.8
g177204
Human type IV collagenase mRNA, complete cds.
gbpri
3.4


41
2196988
242275.7
g38068


M.fuscata
mRNA for pepsinogen A-2/3.

gbpri
3.1


42
3257673
257576.34
g546087
cytoplasmic antiproteinase = 38 kda intracellular
gbpri
3.9






serine protease inhibitor (serpin)


43
2992363
343903.1
g35951
Human kidney mRNA fragment for renin (aa 105-401).
gbpri
3.1


44
2799255
348064.3
g1172085
Human squamous cell carcinoma antigen (SCCA1) gene
gbpri
7.5


45
1601009
413969.1
g2228241
Home sapiens clone rasi-3 matrix metalloproteinase RASI-1
gbpri
3.2


46
1747756
416874.3
g340159
Human pro-urokinase mRNA, complete cds.
gbpri
5.3


47
2455295
437025.25
g296739


H. sapiens
mRNA for macropain subunit zeta.

gbpri
3.1


48
1985367
478620.78
g1890049
Home sapiens mRNA for cysteine protease, complete cds.
gbpri
4.7


49
1274803
902393.23
g189841
Human prolidase (imidodipeptidase) mRNA, complete cds.
gbpri
3.1


50
1393032
992309.26
g338668
Human testicular angiotensin converting enzyme mRNA
gbpri
5.2


51
1404616
992309.27
g178285
Human angiotensin I-converting enzyme mRNA, gbpri
3.3






complete cds.


51
1427612
992309.27
g178285
Human angiotensin I-converting enzyme mRNA,
gbpri
4.0, 3.4






complete cds.


52
154946
153659.2
g32576


H. sapiens
mRNA for interleukin-1 receptor antagonist.

gbpri
6.1, 4.6, 4.1


52
519653
153659.2
g32576


H. sapiens
mRNA for interleukin-1 receptor antagonist.

gbpri
6.2


53
1218389
242114.46
g818880
Pan troglodytes epididymal secretory protein precursor
gbpri
6.0, 4.7


53
1875737
242114.46
g818880
Pan troglodytes epididymal secretory protein precursor
gbpri
8.6


54
1446858
242114.47
g818880
Pan troglodytes epididymal secretory protein precursor
gbpri
5.0, 4.8


55
1375759
242114.48
g818880
Pan troglodytes epididymal secretory protein precursor
gbpri
3.4


56
2722128
244973.9
g2978559


Homo sapiens
vascular endothelial cell growth factor 165

gbpri
5.9


57
1273066
290224.2
g1223889
Human putative T1/ST2 receptor binding protein precursor
gbpri
3.1


58
1314322
331428.2
g1885307


H. sapiens
mRNA for IL13 receptor alpha-1 chain.

gbpri
3.5


59
2057240
401593.10
g1203964
Home sapiens bone-derived growth factor (BPGF-1)
gbpri
3.1


60
504786
199772.6
g187297
Human MAL protein gene mRNA, complete cds
gbpri
3.4


61
1505813
330836.1
g189263
Human notch group protein (N) mRNA, partial cds.
gbpri
9.9


62
518094
902142.11
g2653870
Home sapiens leucocyte immunoglobulin-like receptor-5
gbpri
3.1


63
351384
995211.5
g184428
Human heparan sulfate proteoglycan (HSPG) core protein, 3′
gbpri
6.2, 5.4


63
1601035
995211.5
g184428
Human heparan sulfate proteoglycan (HSPG) core protein, 3′
gbpri
6.8


64
1435222
235310.12
g2905995


Homo sapiens
DAP12 gene, complete cds.

gbpri
4.4


65
1402833
900031.4
g2094872


H. sapiens
DAP-kinase mRNA.

gbpri
3.7


66
2493520
013521.17
g38274
human ret proto-oncogene mRNA for tyrosine kinase
gbpri
4.5


67

013521.18


68
1867423
032942.8
g763112


H. sapiens
ERK3 mRNA.

gbpri
3.6


69
157088
149914.21
g1418933


H. sapiens
mRNA for protein-tyrosine-phosphatase

gbpri
3.1


70
2501777
276948.4
g31197
Human c-erb-B-2 mRNA.
gbpri
5.7


71
810002
347283.16
g531819
Human B lymphocyte serine/threonine protein kinase
gbpri
30.4


72
1418741
349727.2
g306804
Human G protein-coupled receptor kinase (GRK5)
gbpri
19.5


73
2662471
350230.2
g189982
Human testis-specific cAMP-dependent protein kinase
gbpri
3.7


74
1524551
386989.40
g1526989
Human cAMP-dependent protein kinase type I-alpha subunit
gbpri
3.2


75
1437116
434608.8
g34275
Human mRNA for T200 leukocyte common
gbpri
3.2






antigen (CD45, LC-A).


76
1725791
891141.5
g3043926
Home sapiens Tat-interacting protein TIP30
gbpri
3.1


77
1820994
233653.13
g2738496


Homo sapiens
putative oral tumor suppressor protein (doc-1)

gbpri
3.5


78
191334
253405.7
g1297329
Human mitogen-responsive phosphoprotein DOC-2 mRNA
gbpri
5.1


79
1509469
197600.12
g3800808


Homo sapiens
methyl-CpG binding

gbpri
3.7






protein MBD4 (MBD4) mRNA,


80
818223
1078199.1
g38031
Human ZNF43 mRNA
gbpri
3.5


81
2208461
025138.1
g2088550
similar to 52kd Ro/SSA protein and RFP
gbpri
9.7


82
33091
1000222.1
g181485
Human DNA-binding protein B (dbpB) gene, 3′end.
gbpri
3.9


83
1818802
1079467.1
g468707


H. sapiens
OZF mRNA.

gbpri
3.3


84
2881835
1082203.1
g6409344
Home sapiens zinc finger protein ZNF180 (ZNF180) mRNA,
gbpri
3.1


85
1434547
1096748.6
g509777


H. sapiens
ERF-2 mRNA.

gbpri
7.7


86
1467384
1098409.1
g181986
Human early growth response 2 protein
gbpri
3.3






(EGR2) mRNA, complete


86
3603037
1098409.1
g181986
Human early growth response 2 protein
gbpri
3.2






(EGR2) mRNA, complete


87
2115028
1098584.1
g6644296
Home sapiens IFI16b (IFI16b) mRNA,
gbpri
3.5






complete cds.


88
2115028
40652.29


89
1685409
1099277.17
g3641537


Homo sapiens
small EDRK-rich factor 2 (SERF2) mRNA

gbpri
3.9


90
2735724
197748.7
g5757883
Home sapiens zinc finger Homeobox protein ZHX1 mRNA,
gbpri
29.3


91
1797396
241930.15
g726512
Human nuclear orphan receptor LXR-alpha
gbpri
3.1






mRNA, complete cds.


92
1274720
246504.1
g287642
H sapiens cDNA for TREB protein.
gbpri
4.2


93
1575463
253341.11
g2947300


Homo sapiens
MMS2 (MMS2) mRNA, complete cds.

gbpri
3.1


94
1292502
334064.3
g2190973


Homo sapiens
breast cancer suppressor candidate 1 (bcsc-1)

gbpri
4.9


94
2378649
334064.3
g2190973
Home sapiens breast cancer suppressor candidate 1 (bcsc-1)
gbpri
5.5


94
2595814
334064.3
g2190973


Homo sapiens
breast cancer suppressor candidate 1 (bcsc-1)

gbpri
3.6


95
1554495
347723.6
g1374697


Homo sapiens
mRNA for nuclear protein,

gbpri
5, 3.4






NP220, complete cds.


96
1628271
902614.15
g1480099


H. sapiens
mRNA for peroxisome proliferactor activated

gbpri
4.3


97
1969563
995119.1
g186624
Human c-jun proto oncogene (JUN),
gbpri
4.0






complete cds, clone hCJ-1.


98
3117417
999437.3
9498722


H. sapiens
HZF2 mRNA for zinc finger protein from U-937

gbpri
3.7


99
27775
1225522.1
g28434
Human mRNA for apoferritin H chain type.
gbpri
3.4, 3.5


100
262983
235447.5
g37432
Human mRNA for transferrin receptor.
gbpri
4.3, 3.3


101
3121380
139899.22
g182513
Human ferritin L chain mRNA, complete cds.
gbpri
23.2


102
2055571
977615.8
g313158


H. sapiens
mRNA for GM2 activator protein.

gbpri
4.1


103
1556827
995174.7
g2276462


Homo sapiens
Niemann-Pick C disease

gbpri
8.0






protein (NPC1) mRNA; similar to patche


104
2515389
346599.14
g178848
Human apolipoprotein E
gbpri
5.7, 22.3,








18.8


104
2516686
346599.14
g178848
Human apolipoprotein E
gbpri
17.2, 16.2


105
2369312
1328674.2
g178830
Human apolipoprotein C-I (VLDL) gene, complete cds.
gbpri
3.9


106
1214221
1099498.9
g178830
Human apolipoprotein C-I (VLDL) gene, complete cds.
gbpri
3.4


106
2604020
1099498.9
g178830
Human apolipoprotein C-I (VLDL) gene, complete cds.
gbpri
4.8, 4.0


107
1694113
049457.6
g187209
Human lipoprotein lipase mRNA, complete cds.
gbpri
6.1


107
2118094
049457.6
g187209
Human lipoprotein lipase mRNA, complete cds.
gbpri
4.5


108
487539
092511.1
g1255603
Human mRNA for cytosolic malate
gbpri
3.4






dehydrogenase, complete cds.


108
491360
092511.1
g1255603
Human mRNA for cytosolic malate
gbpri
7.5






dehydrogenase, complete cds.


109
2446571
092511.6
g1255603
Human mRNA for cytosolic malate
gbpri
19.4






dehydrogenase, complete cds.


110
1488759
238609.2
g1060902
Human mRNA for phosphatidylinositol transfer protein
gbpri
4.5


110
2122844
238609.2
g1060902
Human mRNA for phosphatidylinositol transfer protein
gbpri
4.0


111
2224733
252895.15
g181477
Human diazepam binding inhibitor (DBI) mRNA
gbpri
3.3


112
2805736
252895.16
g181477
Human diazepam binding inhibitor (DBI) mRNA
gbpri
4.8


113
2537805
99588012
g182353
Human fatty acid binding protein homologue
gbpri
10.3, 17.5






(PA-FABP) mRNA,


114
2673842
001726.1
g5901760
fatty acid binding protein homolog
gbpri
28.3


115
4013105
179368.2
g1377853
Human fatty acid binding protein FABP gene, complete cds.
gbpri
3.5


116
1524345
410917.7
g506430


H. sapiens
(HepG2) LAL mRNA for lysosomal acid lipase.

gbpri
11.5


116
1970427
410917.7
g506430


H. sapiens
(HepG2) LAL mRNA for lysosomal acid lipase.

gbpri
6.1


117
1658320
336012.15
g179459
Human alpha-polypeptide of N-acetyl-alpha-glucosaminidase
gbpri
5.8


117
2448129
336012.15
g179459
Human alpha-polypeptide of N-acetyl-alpha-glucosaminidase
gbpri
5.4, 4.6


118
2096385
235128.9
g1160966


Homo sapiens
palmitoyl-protein thioesterase gene, complete

gbpri
4.5


118
1536287
235128.9
g1160966


Homo sapiens
palmitoyl-protein thioesterase gene, complete

gbpri
5.1


119
1968372
215481.15
g34733
Human mRNA for acid phosphatase type 5 (EC 3.1.3.2).
gbpri
4.9


119
2206828
215481.15
g34733
Human mRNA for acid phosphatase type 5 (EC 3.1.3.2).
gbpri
6.0


120
3223710
885713.30
g1008454
Human mitochondrial ATP synthase subunit 9, P3 gene copy,
gbpri
4.6


121
3029307
902700.6
g28939
Human mRNA for F1-ATPase beta subunit (F-1 beta).
gbpri
3.5


122
2510502
401575.10
g3335127


Homo sapiens
F1Fo-ATPase synthase

gbpri
4.1






f subunit mRNA, complete


122
2816429
401575.10
g3335127


Homo sapiens
F1Fo-ATPase synthase

gbpri
4.1, 3.6






f subunit mRNA, complete


123
2487243
232818.15
g3329377


Homo sapiens
vacuolar H(+)-ATPase

gbpri
3.4






subunit mRNA, complete


124
921297
233103.10
g1395161


Homo sapiens
mRNA for vacuolar ATPase, complete cds.

gbpri
3.5


124
2192978
233103.10
g1395161


Homo sapiens
mRNA for vacuolar ATPase, complete cds.

gbpri
3.9


125
2510187
234313.13
g509290


H. sapiens
mRNA for H+-ATP synthase subunit b.

gbpri
3.1


126
2510187
234313.14


126
2683449
234313.14
g509290


H. sapiens
mRNA for H+-ATP synthase subunit b.

gbpri
3.8


127
1648713
246862.17
g559324
Human mRNA for ATP synthase alpha subunit, complete cds.
gbpri
6.2, 6.0


128
859321
253969.2
g522192


Homo sapiens
vacuolar H+-ATPase

gbpri
12.5






Mr 56,000 subunit (HO57)


128
2075603
253969.2
g522192


Homo sapiens
vacuolar H+-ATPase

gbpri
7.5






Mr 56,000 subunit (HO57)


129
1232362
328615.1
g2393721
Human glutathione-S-transferase homolog
gbpri
5.0






mRNA, complete cds.


130
2708221
998594.2
g1747520


Homo sapiens
microsomal glutathione S-transferase 2

gbpri
3.1


131
1923262
220837.3
g183141
Human gamma-glutmyl transpeptidase-related
gbpri
3.2






protein (GGT-Rel)


132
2099268
410296.1
g4106439
multidrug resistance-associated protein 3
gbpri
5.1


132
2642164
410296.1
g3550323


Homo sapiens
canalicular multispecific

gbpri
4.6, 4.3






organic anion transporter; similar to MRP


133
851304
218849.9
g5006263
g5006263 organic anion transporter OATP-B
genpept
4.6


134
851304
218849.15


135
374690
218849.16
g5006263
g5006263 organic anion transporter OATP-B
genpept
4.1


133
374690
218849.9


135
1512278
218849.16
g5006263
g5006263 organic anion transporter OATP-B
genpept
5.0


136
2448113
233319.14
g3329393


Homo sapiens
voltage dependent

gbpri
4.0






anion channel protein mRNA,


137
2021456
434763.22
g340198
Human voltage-dependent anion channel
gbpri
3.3






isoform 1 (VDAC) mRNA,


138
2021456
434763.18


139
1269859
336954.1
g2739502


Homo sapiens
potassium channel mRNA,

gbpri
4.7






complete cds.


140
2742236
337928.1
g2584784


H. sapiens
mRNA homologous to the p64

gbpri
3.2






bovine chloride channel


141
109018
430431.18
g4426566


Homo sapiens
chloride channel ABP mRNA, complete cds.

gbpri
3.4


142
2361578
407376.3
g4581469


Homo sapiens
mRNA for SLC7A8 protein.

gbpri
3.9


143
1749790
439010.7
g6826913


Homo sapiens
calcium transport ATPase ATP2C1

gbpri
4.5


144
2056395
245184.1
g339567
Human transforming growth factor-beta induced gene product
gbpri
10.4, 11.0


145
2965657
352406.29
g3360431


Homo sapiens
clone 23810 osteopontin mRNA, complete cds.

gbpri
28.5


146
229442
352406.28
g3360431


Homo sapiens
clone 23810 osteopontin mRNA, complete cds.

gbpri
11.5


147
2495131
992455.45
g187109
Human 14 kd lectin mRNA, complete cds.
gbpri
5.5


148
2057608
255957.1
g4323580


Homo sapiens
senescence-associated epithelial membrane

gbpri
14.4


149
1310287
235164.25
g28525
Human mRNA for amyloid A4 precursor
gbpri
3.2






of Alzheimer's disease.


150
2645263
997363.3
g4586835


Homo sapiens
mRNA for type II membrane protein

gbpri
3.3


151

997363.1


152
853968
1327354.1
g32014
Human HALPHA44 gene for alpha-tubulin, exons 1-3.
gbpri
3.6


152
1296056
1327354.1
g32014
Human HALPHA44 gene for alpha-tubulin, exons 1-3.
gbpri
4.1


153
1522716
358853.44
g37851


H. sapiens
vimentin gene.

gbpri
10.1


154
2508570
243019.14
g187455


Homo sapiens
macrophage capping protein

gbpri
3.4






mRNA, complete cds.


155
3208407
1000016.22
g36502
Human mRNA for enteric smooth muscle gamma-actin.
gbpri
4.0


156

1000016.44


157
218631
1000084.41
g340020
human alpha-tubulin mRNA, complete cds.
gbpri
6.1


158
1269670
481480.2
g7012928


Homo sapiens
SCG10 like-protein

gbpri
3.5


159
1469939
214181.1
g461396


Homo sapiens
alpha-1 type XV collagen mRNA,

gbpri
3.6






complete cds.


160
2190217
977305.3
g829622
Human myosin regulatory light chain mRNA, complete cds.
gbpri
4.5, 3.3


161
2379695
450223.3
g7288158


Homo sapiens
mRNA for testis specific ankyrin-like protein

gbpri
6.0


162
2665489
375072.25
g2739095


Homo sapiens
protein 4.1-G mRNA, complete cds.

gbpri
3.1


163
2926914
1039889.12
g28251
Human mRNA for beta-actin.
gbpri
4.0


164
3215205
245367.2
g5932031


Homo sapiens
PEA15 protein (PEA15) gene,

4.1






exons 3 and 4 and
gbpri


165
2444446
252747.21
g998356


Homo sapiens
EB1 mRNA, complete cds

gbpri
3.8


166
1648449
229250.3
g3006201


Homo sapiens
prostaglandin transporter

gbpri
14.1






hPGT mRNA, complete


167
1886871
331734.3
g189886
Human prostaglandin endoperoxide
gbpri
3.4






synthase mRNA, complete cds.


167
1595081
331734.3
g249623
prostaglandin G/H synthase {alternative splicing product}
gbpri
4.7


168

331734.8


169
1401683
441261.5
g862374
Human 180 kDa transmembrane PLA2
gbpri
4.6






receptor mRNA, complete cds.


170
638749
411208.4
g2911111


Homo sapiens
mRNA for NRAMP2; innate immunity

gbpri
3.4


170
933140
411208.4
g2911111


Homo sapiens
mRNA for NRAMP2; innate immunity

gbpri
3.1, 3.1


171
1241477
335940.2
g2459617


Homo sapiens
Toll-like receptor 1

gbpri
3.4






(TLR1) mRNA, complete cds.


172
1661537
369213.26
g177869
Human alpha-2-macroglobulin mRNA, complete cds.
gbpri
13.3


173
1605616
222156.2
g1050957
Human chitotriosidase precursor mRNA, complete cds.
gbpri
9.7


174
1000988
234404.4
g182836
Human factor XIII subunit a mRNA, 3′ end.
gbpri
11.0


174
1874280
234404.4
g182836
Human factor XIII subunit a mRNA, 3′ end.
gbpri
11.9, 9.3


175
1436753
474310.15
g339520
Human transglutaminase (TGase) mRNA, complete cds.
gbpri
22.7


175
1672744
474310.15
g339520
Human transglutaminase (TGase) mRNA, complete cds.
gbpri
14.5


176
1557811
254107.1
g37123
Human mRNA for thrombomodulin precursor.
gbpri
4.3


177
1220371
218156.2
g3925598


Homo sapiens
mRNA for DORA protein

gbpri
14.5


178
1533410
350788.2
g407955
Human membrane-associated protein (HEM-1)
gbpri
4.7






mRNA, complete cds.


179
1999422
899239.5
g29645


H. sapiens
mRNA for CAMPATH-1 (CDw52) antigen.

gbpri
4.2, 3.5


180
322866
382906.6
g180142
Human CD53 glycoprotein mRNA, complete cds.
gbpri
3.2


181
1874037
382906.16
g180142
Human CD53 glycoprotein mRNA, complete cds.
gbpri
19.5


182
2419118
269837.15
g6563293


Homo sapiens
PIL protein mRNA, complete cds.

gbpri
3.5


183
1689409
241016.6
g1737489
Human p76 mRNA, complete cds. Membrane protein
genpept
4.6


184
2182130
005486.4
g4336324


Homo sapiens
small membrane protein 1 (SMP1) mRNA,

genpept
3.2


185
2242283
977953.8

Incyte Unique

4.5


186
2041671
404923.1

Incyte Unique

5.5


187
1621280
449028.32

Incyte Unique

3.1


188
1869766
330864.4

Incyte Unique

4.6


189
1427838
39903.4

Incyte Unique

3.7, 3.2


190
1683829
250445.3

Incyte Unique

4.0


191
1996005
400961.22

Incyte Unique

3.3


192
2503016
977835.14

Incyte Unique

3.1


193
1748678
252920.1

Incyte Unique

4.7


194
2716391
343863.10

Incyte Unique

3.1


195
3134070
254547.9

Incyte Unique

10.3


196
1996357
277726.11

Incyte Unique

4.1, 3.8


197
1999313
246177.4

Incyte Unique

4.8


198
2046715
410012.21

Incyte Unique

5.7


199
1886544
410012.24

Incyte Unique

4.4


200

410012.20

Incyte Unique


201
2628690
1099099.16
g2772672
phospholipase D-like protein; g1575347 HU-K4
genpept
4.5
3.00E−99


202

1099099.13


201
642116
1099099.16
g2772672
phospholipase D-like protein; g1575347 HU-K4
genpept
3.7, 3.2


203
663537
400253.14
g2826476
IL-17 receptor
genpept
8.5
9.00E−30


204

400253.17


205
1509631
85803.4
g50229
C1q C chain [Mus musculus]
genpept
9.1, 7.3
3.00E−71


206
2790509
253868.2
g3879017
similar to Ubiquitin-conjugating enzymes; cDNA EST
genpept
3.5
2.00E−42


206
1426987
253868.2
g3879017
similar to Ubiquitin-conjugating enzymes; cDNA EST
genpept
3.6
2.00E−42


207
2674742
158923.9
g607003
beta transducin-like protein
genpept
4.2
2.00E−29


208
3330716
199716.3
g7243708
zinc transporter like 1 [Mus musculus]
genpept
3.2
0


209
1620875
252899.8
g6176319
butyrophilin-like [Homo sapiens ]
genpept
3.2
4.00E−35


210
627910
327544.2
g7406950
N system amino acids transporter NAT-1 [Mus musculus]
genpept
5.0
1.00E−112


210
2007779
327544.2
g7406950
N system amino acids transporter NAT-1 [Mus musculus]
genpept
6.1
1.00E−112


210
2726053
327544.2
g7406950
N system amino acids transporter NAT-1 [Mus musculus]
genpept
4.6, 3.9
1.00E−112


211
868138
334842.5
g6175860
g1-related zinc finger protein [Mus musculus]
genpept
22.7
2.00E−84


212
938514
406770.1
g1841756
GATA-5 cardiac transcription factor [Mus musculus]
genpept
3.2
2.00E−84


213
518335
996806.15
g19867
extensin (AA 1-620)
genpept
3.1
0.018


213
2727735
996806.15
g19867
extensin (AA 1-620)
genpept
4.1
0.018


214
2495063
332263.1
g5817244
dJ20N2.1 similar to yeast and bacterial cytosine deaminase
genpept
4.1
1.00E−89


215
2659055
255824.34
g6572231
dJ402G11.6 similar to Xenopus gamma-tubulin
genpept
4.0
0


216
1963927
1095059.19

Incyte Unique

3.5


217
1662589
902753.4

Incyte Unique

4.6


218
2674037
232259.1

Incyte Unique

4.0


219
1220608
330879.1

Incyte Unique

3.2


220
1868520
370489.34

Incyte Unique

3.5


221
1447681
233007.5

Incyte Unique

3.3


222
2236427
422389.9

Incyte Unique

3.1


223
1632936
228814.8

Incyte Unique

3.4


223
1519414
228814.8

Incyte Unique

3.1


224

228814.10

Incyte Unique


225
519879
234169.3

Incyte Unique

3.4


226
1656490
403676.1

Incyte Unique

15.9


227
1536275
231546.1

Incyte Unique

3.3


228
2212027
247544.1

Incyte Unique

7.2


229
782704
237320.1

Incyte Unique

3.2


230
1848664
983197.1

Incyte Unique

13.3


231
777478
481455.4

Incyte Unique

3.6


231
2444774
481455.4

Incyte Unique

4.2


232

481455.6


233
2718363
237733.2

Incyte Unique

3.3


234
1907948
040151.2

Incyte Unique

3.4


235
1864720
425448.2

Incyte Unique

3.6


236
1857223
012712.1

Incyte Unique

4.2


237
104201
009707.3

Incyte Unique

4.7


238
1871285
019751.1

Incyte Unique

3.9


239
1931160
065689.1

Incyte Unique

12.7


240
1611844
279164.6

Incyte Unique

3.1


241
2041593
428329.1

Incyte Unique

3.2










[0206]

4











TABLE 4








SEQ








ID


NO
CLONE ID
TEMPLATE ID
GENBANK ID
DESCRIPTION
DATABASE
BDE





















242
154371
988891.1
g35662
Human mRNA for prointerleukin 1 beta.
gbpri
−3.1


243

988891.13


244
3661567
1327819.1
g306500
Human CD9 antigen mRNA, complete cds; splice variant?
gbpri
−3.1


245

1328161.1


246
1904149
1099039.5
g307390
Human ribosomal protein S13 (RPS13) mRNA, complete cds.
gbpri
−4.2


247
2527879
1328423.2
g682747
Human mRNA for Apo1_Human (MER5(Aop1-Mouse)-like protein),
gbpri
−2.8


248
3168108
232691.21
g1518017
Human TRAF-interacting protein I-TRAF mRNA, complete cds.
gbpri
−3.2


249
2785701
241888.40
g186367
Human interleukin 8 (IL8) gene, complete cds.
gbpri
−8.4


249
3732536
241888.40
g186367
Human interleukin 8 (IL8) gene, complete cds.
gbpri
−12.5


250
1972768
256005.49
g178017
Human activation (Act-2) mRNA, complete cds.
gbpri
−3.3


251
80154
256005.51
g178017
Human activation (Act-2) mRNA, complete cds.
gbpri
−4.6


252
1618459
331851.5
g2276318


Homo sapiens
mRNA for dynein heavy chain.

gbpri
−2.8


253
1603857
341546.3
g3978243


Homo sapiens
inhibitor of apoptosis protein-1 (MIHC) mRNA,

gbpri
−3


254
3126072
400834.2
g5042402
UCRI_HUMAN; RIESKE IRON-SULFUR PROTEIN; RISP
genpept
−2.6


255
4718119
407740.1
g5305442


Homo sapiens
carnitine octanoyltransferase mRNA, complete

gbpri
−5


256
1543740
1082684.1

Incyte Unique

−4.5


257
3122044
318000.8

Incyte Unique

−3










[0207]

5











TABLE 5








SEQ ID NO
CLONE ID
TEMPLATE ID
GENBANK ID
DESCRIPTION
DATABASE
BDE





















258
1943863
206382.35
g2558583
neurogranin
genpept
4


259
790977
1090400.1

Incyte Unique

2.9


260
1444550
018342.1

Incyte Unique

2.6










[0208]


Claims
  • 1. A combination comprising a plurality of cDNAs that are differentially expressed in DC, and selected from SEQ ID NOs:1-260 or their complements.
  • 2. The combination of claim 1, wherein each of the cDNAs is downregulated in DC and is selected from SEQ ID NOs:1-29.
  • 3. The combination of claim 1, wherein each of the cDNAs is upregulated in DC and is selected from SEQ ID NOs:30-241.
  • 4. The combination of claim 1, wherein each of the cDNAs is upregulated in mature DC and is selected from SEQ ID NOs:242-257.
  • 5. The combination of claim 1, wherein each of the cDNAs is downregulated in mature DC and is selected from SEQ ID NOs:258-260.
  • 6. The combination of claim 1, wherein the cDNAs are immobilized on a substrate.
  • 7. A high throughput 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, wherein 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 cancer, infectious disease, autoimmunity, allergy, or graft versus host disease and comparison with a standard identifies or stages the disorder.
  • 10. A high throughput method of screening a plurality of molecules or compounds to identify a ligand 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 ligand that specifically binds to each cDNA.
  • 11. The method of claim 10 wherein the plurality of molecules or compounds are selected from aptamers, DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, transcription factors, repressors, and regulatory proteins.
  • 12. An isolated cDNA selected from the group consisting of SEQ ID NOs:24-29, 187-202, 205-213, 216-241, 256, 257, 259, and 260.
  • 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 high-throughput 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 DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, proteins, agonists, antagonists, antibodies or their fragments, immunoglobulins, inhibitors, drug compounds, and pharmaceutical agents.
  • 19. A method of using a protein to produce an antibody, 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. A antibody produced by the method of claim 19.
Provisional Applications (1)
Number Date Country
60237652 Oct 2000 US