Genes regulated by MYCN activation

Information

  • Patent Application
  • 20030119009
  • Publication Number
    20030119009
  • Date Filed
    February 25, 2002
    22 years ago
  • Date Published
    June 26, 2003
    21 years ago
Abstract
The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed by MYCN activation and which may be used in their entirety or in part to diagnose, to stage, to treat, or to monitor the treatment of a subject with neuroblastoma.
Description


FIELD OF THE INVENTION

[0002] The present invention relates to a combination comprising a plurality of cDNAs which are differentially expressed in response to MYCN activation and which may be used entirely or in part to diagnose, to stage, to treat, or to monitor the progression or treatment of disorders associated with MYCN activation such as neuroblastoma and other cancers.



BACKGROUND OF THE INVENTION

[0003] MYCN functions as a transactivator by forming a heterodimer with a helix-loop-helix (HLH)-leucine zipper protein, MAX, and binding to a core E-box promoter element, CAT/cGTG. Although MYCC, MYCN and MYCL can all bind to the canonical core element, promoter-flanking sequences can markedly affect binding affinities of the different MYC isoforms to individual target genes (Prochownik et al. (1993). Proc Natl Acad Sci 90:960-964). In addition, other regulatory molecules that influence both the activity of MYC and the interaction of MYC with MAX contribute to the tissue specificity of MYCN and MYCC oncogene transactivation (O'Hagan et al. (2000) Nat Genet 24:113-9; and Hurlin et al. (1996) Embo J 15:2030).


[0004] The MYCN proto-oncogene is amplified in approximately 30% of neuroblastoma and 20-25% of small cell lung cancers. Amplification of MYCN is the most reliable negative prognostic factor in neuroblastoma (Bordow et al. (1998) J Clin Oncol 16:3286-94). Three-year survival for patients with metastatic neuroblastoma decreases from approximately 40% to less than 10% with MYCN amplification (Matthay et al. (1999) N Engl J Med 341:1165-73). Despite intensive efforts, MYCN transcriptional targets responsible for the particularly malignant phenotype of these tumors have remained elusive (Nesbit et al. (1999) Oncogene 18:3004-16).


[0005] Minichromosome maintenance (MCM) molecules are a highly conserved group of DNA binding proteins with a vital function of “licensing” DNA synthesis during the transition from G1 to S phase of the cell cycle (Takisawa et al. (2000) Curr Opin Cell Biol 12:690-696). Originally characterized in yeast, the MCM group of proteins is required for activation of autonomous replicating sequences and progression through the cell division cycle. In eukaryotes, MCM2 to MCM7 are sequentially assembled into a heteromeric hexamer, the replication licensing factor (RLF), that binds to DNA replication origins after the origin recognition complex (ORC) has assembled at the end of G1 (Maiorano et al. (2000) J Biol Chem 275:8426-31; for review, see Kearsey and Labib (1998) Biochim Biophys Acta 1398:113-36; Tye (1999) Annu Rev Biochem 68:649-86; and Thommes and Blow (1997) Cancer Surv 29:75-90). MCM-mediated regulation of DNA synthesis ensures that DNA replicates only once during each cell cycle and is essential for maintaining euploidy.


[0006] Immunohistochemical studies in a variety of tissues demonstrate increased expression of MCM2, 5, and 7 in solid tumors and pre-malignant proliferative states (Todorov et al. (1998) Lab Invest 78:73-8; Freeman et al. (1999) Clin Cancer Res 5:2121-32; and Hiraiwa et al. (1998) J Cutan Pathol 25:285-90). Their vital role in the maintenance of chromosomal integrity in normal cells makes the MCM molecules potential targets of the transforming effects of cellular oncogenes. The promoter regions of MCM5, 6 and 7 each contain numerous E2F transactivation sites, suggesting that the E2F transcription factor may be primarily responsible for the coordinated increase in MCM mRNA noted at the G1/S boundary (Ohtani et al. (1999) Oncogene 18, 2299-309; Suzuki et al. (1998) Gene 216:85-91). However, in addition to the E2F sites, the MCM7 promoter has an E-box binding site for the MYC oncogene (Suzuki, supra).


[0007] Array technology can provide a simple way to explore the expression profile of a large number of related or unrelated genes. The potential application of gene expression profiling is particularly relevant to improving diagnosis, prognosis, and treatment of disease. For example, both the levels and sequences expressed in tissues from subjects with neuroblastoma associated with MYCN amplification may be compared with the levels and sequences expressed in neuroblastoma without MYCN amplification.


[0008] The present invention provides a combination comprising a plurality of cDNAs for use in detecting changes in expression of genes encoding proteins that are associated with MYCN activation. Such a combination satisfies a need in the art by providing a combination of cDNAs that represent a set of differentially expressed genes which may be used entirely or in part to diagnose, to stage, to treat, or to monitor the progression or treatment of disorders such as neuroblastoma and other cancers.



SUMMARY

[0009] The present invention provides a combination comprising a plurality of cDNAs and their complements which are differentially expressed by MYCN activation and which are selected from SEQ ID NOs:1-365 as presented in the Sequence Listing. In one embodiment, each cDNA is downregulated at least two-fold, SEQ ID NOs:1-280; in another embodiment, each cDNA is upregulated at least two-fold, SEQ ID NOs:281-365. In one aspect, the combination is useful to diagnose, to stage, to treat, or to monitor the progression or treatment of disorders associated with MYCN activation such as neuroblastoma and other cancers. In another aspect, the combination is immobilized on a substrate.


[0010] The invention also provides a high throughput method to detect differential expression of one or more of the cDNAs of the combination. The method comprises hybridizing the substrate containing the combination with nucleic acids of a sample, thereby forming one or more hybridization complexes, detecting complex formation, and comparing complexes with those of a standard, wherein differences in the size and signal intensity of each complex indicates differential expression of nucleic acids in the sample. In one aspect, the sample is from a subject with neuroblastoma and differential expression determines an early, mid, and late stage of that disorder.


[0011] The invention further provides a high throughput method for screening a library or a plurality of molecules or compounds to identify a ligand. The method comprises combining the substrate comprising the combination of cDNAs with a library or a plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds at least one cDNA of the combination. The library or plurality of molecules or compounds are selected from 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.


[0012] The invention provides an isolated cDNA selected from SEQ ID NOs: 1-365. The invention also provides a vector comprising the cDNA, a host cell comprising the vector, and a method for producing a protein comprising culturing the host cell under conditions for the expression of a protein and recovering the protein from the host cell culture. The invention further provides a method to detect differential expression of a cDNAs of the combination. The method comprises using a cDNA of the invention to detect expression of nucleic acids in a sample comprising contacting the cDNA with the sample, thereby forming a hybridization complex, detecting complex formation wherein complex formation indicates expression of the nucleic acid in the sample. In one aspect, the sample is from a subject with neuroblastoma and differential expression of the cDNA determines the stage of the disorder.


[0013] The invention provides a method for using a cDNA to screen a library or a plurality of molecules or compounds to identify a ligand. The method comprises combining the cDNA with a library or a plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds the cDNA. The library or plurality of molecules or compounds are selected from 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 such as a transcription factor, the method comprising combining the cDNA with a sample under conditions which allow specific binding, recovering the bound cDNA, and separating the cDNA from the ligand, thereby obtaining purified ligand.


[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 DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, proteins, agonists, antagonists, antibodies or their fragments, immunoglobulins, inhibitors, drug compounds, and pharmaceutical agents.


[0015] The invention provides a method for using a protein to produce and purify an antibody, the method comprising immunizing an animal with the protein under conditions to elicit an antibody response; isolating animal antibodies; contacting the protein with the isolated antibodies under conditions to allow specific binding; recovering the bound protein; and separating the protein from the antibody, thereby obtaining purified antibody. The invention also provides a method for using an antibody to detect expression of a protein in a sample, the method comprising contacting the antibody with a sample under condition for the formation of an antibody:protein complex; and detecting the antibody:protein complex wherein complex formation indicates the expression of the protein in the sample. In one aspect, complex formation is compared to standards and is diagnostic of cancer, particularly a neuroblastoma. The invention further provides a method of using an antibody to immunopurify a protein comprising combining the antibody with a sample under conditions to allow formation of an antibody:protein complex, and separating the antibody from the protein, thereby obtaining purified protein.


[0016] The invention further provides a composition comprising a cDNA, a protein, an antibody, or a ligand which has agonistic or antagonistic activity.



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

[0017] CD-R 1 is labeled: “PA-0046 US, Copy 1,” was created on Feb. 25, 2002 and contains: the Sequence Listing formatted in plain ASCII text. The file for the Sequence Listing is entitled pa46sqls.txt, created on Feb. 25, 2002 and is 1360 KB in size.


[0018] CD-R 2 is an exact copy of CD-R 1. CD-R 2 is labeled: “PA-0046 US, Copy 2,” and was created on Feb. 25, 2002.


[0019] The CD-R labeled as: “PA-0046 US, CRF,” contains the Sequence Listing formatted in plain ASCII text. The file for the Sequence Listing is entitled pa46sqsl.txt, was created on Feb. 25, 2002 and is 1360 KB in size.


[0020] The content of the Sequence Listing named above and as described below, submitted in duplicate on two (2) CD-Rs (labeled “PA-0046 US, Copy 1” and “PA-0046 US, Copy 2”), and the CRF (labeled “PA-0046 US, CRF”) containing the Sequence Listing, are incorporated by reference herein, in their entirety.



DESCRIPTION OF THE SEQUENCE LISTING AND TABLES

[0021] 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.


[0022] The Sequence Listing is a compilation of cDNAs obtained by sequencing and extending clone inserts. Each sequence is identified by a sequence identification number (SEQ ID NO) and by the template number (INCYTE ID) from which it was obtained.


[0023] Table 1 lists the functional annotation and differential expression of the cDNAs of the present invention. Columns 1, 2 and 3 show the SEQ ID NO, TEMPLATE ID, and CLONE ID, respectively. Columns 4, 5 and 6 show the GenBank hit (GENBANK HIT), probability score (E-VALUE), and functional annotation (ANNOTATION), respectively, as determined by BLAST analysis (version 1.4 using default parameters; Altschul (1993) J Mol Evol 36: 290-300; Altschul et al. (1990) J Mol Biol 215:403-410) of the cDNA against GenBank (release 121; National Center for Biotechnology Information (NCBI; Bethesda, Md.). Column 7 shows the balanced differential expression (BAL DE) of each cDNA. Downregulation is represented by positive values and is calculated as the ratio of expression in non-activated cells relative to MYCN activated cells. Upregulation is represented by negative values and is calculated as the ratio of expression in MYCN activated cells relative to non-activated cells.


[0024] Table 2 shows the region of each cDNA encompassed by the clone present on a microarray and identified as differentially expressed. Columns 1 and 2 show the SEQ ID NO and TEMPLATE ID, respectively. Column 3 shows the CLONE ID and columns 4 and 5 show the first residue (START) and last residue (STOP) encompassed by the clone on the template.



DESCRIPTION OF THE INVENTION

[0025] Definitions


[0026] “Antibody” refers to intact immunoglobulin molecule, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab′)2 fragment, an Fv fragment; and an antibody-peptide fusion protein.


[0027] “Antigenic determinant” refers to an immunogenic epitope, structural feature, or region of an oligopeptide, peptide, or protein which is capable of inducing formation of an antibody which specifically binds the protein. Biological activity is not a prerequisite for immunogenicity.


[0028] “Array” refers to an ordered arrangement of at least two cDNAs, proteins, or antibodies on a substrate. At least one of the cDNAs, proteins, or antibodies represents a control or standard, and the other cDNA, protein, or antibody is of diagnostic or therapeutic interest. The arrangement of two to about 40,000 cDNAs, proteins, or antibodies on the substrate assures that the size and signal intensity of each labeled complex, formed between each cDNA and at least one nucleic acid, each protein and at least one ligand or antibody, or each antibody and at least one protein to which the antibody specifically binds, is individually distinguishable.


[0029] “Cancer” refers to adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers or tumors of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, colon, esophagus, gall bladder, ganglia, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, small intestine, spleen, stomach, testis, thymus, thyroid, and uterus.


[0030] A“combination” comprises at least two and up to about 730 sequences selected from SEQ ID NOs:1-365 as presented in the Sequence Listing and the complements thereof.


[0031] The “complement” of a nucleic acid 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 under conditions of high stringency.


[0032] “cDNA” refers to a chain of from about 400 to about 12000 nucleotides, an isolated polynucleotide, nucleic acid, or any fragment 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, and purified or combined with carbohydrate, lipids, protein or inorganic elements or substances.


[0033] The “complement” of a nucleic acid 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 under conditions of high stringency.


[0034] The phrase “cDNA encoding a protein” refers to a nucleic acid sequence that closely aligns with sequences which encode conserved regions, motifs or domains that were identified by employing analyses well known in the art. These analyses include BLAST (Basic Local Alignment Search Tool; Altschul (1993) supra; Altschul (1990) supra) which provides identity within the conserved region. Brenner et al. (1998; Proc Natl Acad Sci 95:6073-6078) who analyzed BLAST for its ability to identify structural homologs by sequence identity found 30% identity is a reliable threshold for sequence alignments of at least 150 residues and 40% is a reasonable threshold for alignments of at least 70 residues (Brenner, page 6076, column 2).


[0035] “Derivative” refers to a cDNA or a protein that has been subjected to a chemical modification. Derivatization of a cDNA can involve substitution of a nontraditional base such as queosine or of an analog such as hypoxanthine. These substitutions are well known in the art. Derivatization of a protein involves the replacement of a hydrogen by an acetyl, acyl, alkyl, amino, formyl, or morpholino group. Derivative molecules retain the biological activities of the naturally occurring molecules but may confer longer lifespan or enhanced activity.


[0036] “Differential expression” refers to an increased or upregulated or a decreased or downregulated expression as detected by presence, absence or at least two-fold change in the amount or abundance of a transcribed messenger RNA or translated protein in a sample.


[0037] “Disorder” refers to conditions, diseases or syndromes associated with MYCN activation, defined by amplification of MYCN gene expression, and includes neuroblastoma and other cancers.


[0038] “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 related nucleic acids and in binding assays to screen for a ligand. Nucleic acids and their ligands identified in this manner are useful as therapeutics to regulate replication, transcription or translation.


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


[0040] 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.


[0041] “Identity” as applied to nucleic acid or protein sequences, refers to the quantification (usually percentage) of nucleotide or residue matches between at least two sequences aligned using a standardized algorithm such as Smith-Waterman alignment (Smith and Waterman (1981) J Mol Biol 147:195-197), CLUSTALW (Thompson et al. (1994) Nucleic Acids Res 22:4673-4680), or BLAST2 (Altschul et al. (1997) Nucleic Acids Res 25:3389-3402). 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 is an analogous score, but it is calculated with conservative substitutions taken into account; for example, substitution of a valine for a isoleucine or leucine.


[0042] “Isolated or purified” refers to a cDNA, protein, or antibody that is removed from its natural environment or from cell culture and that is separated from other components with which it is associated.


[0043] “Labeling moiety” refers to any reporter molecule whether a visible or radioactive label, stain or dye that can be attached to or incorporated into a cDNA or protein. Visible labels and dyes include but are not limited to anthocyanins, β glucuronidase, BIODIPY, Coomassie blue, Cy3 and Cy5, digoxigenin, FITC, green fluorescent protein (GFP), luciferase, spyro red, silver, and the like. Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like.


[0044] “Ligand” refers to any agent, molecule, or compound which will bind specifically to a complementary site on a polynucleotide, protein, or antibody of the invention. Such ligands stabilize, modulate, or disrupt the activity of polynucleotides, proteins, or antibodies and may be composed of inorganic and/or organic substances including minerals, cofactors, nucleic acids, proteins, carbohydrates, fats, and lipids.


[0045] “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 are amplimer, primer, and oligomer.


[0046] “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.


[0047] “Probe” refers to a cDNA that hybridizes to at least one nucleic acid in a sample. Where targets are single stranded, probes are complementary single strands. Probes can be labeled for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies.


[0048] “Protein” refers to a polypeptide or any portion thereof. A “portion” of a protein refers to that length of amino acid sequence which would retain at least one biological activity, a domain identified by PFAM or PRINTS analysis or an antigenic determinant of the protein identified using Kyte-Doolittle algorithms of the PROTEAN program (DNASTAR, Madison, Wis.). 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.


[0049] “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; buccal cells, skin, a hair or hair follicle; and the like.


[0050] “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 antigenic determinant of a protein and an agonist, antagonist, or antibody.


[0051] “Substrate” refers to any rigid or semi-rigid support to which cDNAs, proteins, or antibodies 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.


[0052] A “transcript image” (TI) is a profile of gene transcription activity in a particular tissue at a particular time. TI provides assessment of the relative abundance of expressed polynucleotides in the cDNA libraries of an EST database as described in U.S. Pat. No. 5,840,484, incorporated herein by reference.


[0053] “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.


[0054] The Invention


[0055] The present invention provides for a combination comprising a plurality of cDNAs or their complements, SEQ ID NOs:1-365 which may be used to diagnose, to stage, to treat or to monitor the progression or treatment of a disorder associated with MYCN activation. MYCN activation occurs upon amplification of MYCN gene expression. The cDNAs of the invention represent known and novel genes differentially expressed between a tumor explant from an INSS stage 4 neuroblastoma patient showing amplified MYCN (P4) and a tumor explant from an INSS stage 4 neuroblastoma patient showing non-amplified MYCN (P67). The combination may be used in its entirety or in part, as subsets of downregulated cDNAs, SEQ ID NOs: 1-280, or of upregulated cDNAs, SEQ ID NOs:280-365. Since the 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 it's encoded protein. The usefulness of the cDNAs exists in their immediate value as diagnostics for disorders associated with MYCN activation such as neuroblastoma.


[0056] Table 1 shows those genes on the array having differential expression (4-fold or greater increase or decrease) in the MYCN amplified neuroblastoma. Columns 1, 2, and 3 show the SEQ ID NO, Template ID, and Clone ID, respectively. Columns 4, 5, and 6 show the GenBank Hit ID, the probability score (E-value) for the hit relative to the template or its encoded protein, and a relevant description (annotation). Column 7 shows the balanced differential expression (BAL DE) of each cDNA. Positive values represent a comparison of expression level in non-activated cells relative to MYCN activated cells (downregulation). Negative values represent a comparison of expression level in MYCN activated cells relative to non-activated cells (upregulation). Table 2 shows the region of each cDNA encompassed by the clone present on a microarray and identified as differentially expressed. Columns 1 and 2 show the SEQ ID NO and TEMPLATE ID, respectively. Column 3 shows the CLONE ID and columns 4 and 5 show the first residue (START) and last residue (STOP) encompassed by the clone on the template.


[0057] Regulation of the cDNAs of the invention by MYCN amplification were verified using SEQ ID NO:354, which is upregulated 9.9-fold in the MYCN amplified (P4) tumor relative to non-amplified (P67) tumor (Table 1). Immunohistochemistry on tissue sections from several MYCN amplified and non-amplified INSS stage 4 neuroblastoma tumors with an anti-MCM7 mAb showed the majority of tumor cells in the MYCN amplified samples stained positive with a well-defined nuclear staining pattern. By contrast, only scattered foci of positive staining were demonstrated in the MYCN non-amplified samples despite the fact that the overall proliferative fractions of the tumor samples were very similar. Hematoxylin and eosin staining and neuron-specific enolase staining demonstrated no gross histological differences between the amplified and non-amplified specimens tested. Western blot analysis demonstrated a three fold increase in MCM7 protein upon induction of MYCN in a MYCN conditional neuroblastoma cell line. MCM7 upregulation was also confirmed by RT-PCR in a MYCN non-amplified neuroblastoma cell line, SH-EP, stably transfected with the MYCN gene under the control of the rTet inducible expression system wherein MYCN expression is induced upon removal of tetracycline. Phenotypic changes observed upon MYCN induction in this cell line, TET-21, have been well characterized and include cell cycle alterations, increased proliferation rate and increased metastatic potential (Lutz et al. (1996) Oncogene 13:803-12). Up-regulation of MCM7 transcription was maximal in the TET21 cell line 6 h after removal of tetracycline. The protein level of MCM7 also increased 3-fold as determined by quantitative western blots.


[0058] The cDNAs of the invention define a differential expression pattern against which to compare the expression pattern of biopsied and/or in vitro treated neuroblastoma tissues. Experimentally, differential expression of the cDNAs can be evaluated by other 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 other array technologies. These methods may be used alone or in combination to verify the differential expression patterns that characterize a particular tissue, disorder, or therapy.


[0059] The combination may be arranged on a substrate and hybridized with tissues from subjects with diagnosed neuroblastoma to identify those sequences which are differentially expressed in both neuroblastoma and other phenotypically similar disorders. This allows identification of those sequences of highest diagnostic and potential therapeutic value. In one embodiment, an additional set of cDNAs, such as cDNAs encoding signaling molecules, are arranged on the substrate with the combination. Such combinations may be useful in the elucidation of pathways which are affected in a particular disorder or to identify new, coexpressed, candidate, therapeutic molecules.


[0060] In another embodiment, the combination can be used for large scale genetic or gene expression analysis of a large number of novel, nucleic acids. These samples are prepared by methods well known in the art and are from mammalian cells or tissues which are in a certain stage of development; have been treated with a known molecule or compound, such as a cytokine, growth factor, a drug, and the like; or have been extracted or biopsied from a mammal with a known or unknown condition, disorder, or disease before or after treatment. The sample nucleic acids are hybridized to the combination for the purpose of defining a novel gene profile associated with that developmental stage, treatment, or disorder.


[0061] cDNAs and Their Uses


[0062] cDNAs can be prepared by a variety of synthetic or enzymatic methods well known in the art. cDNAs can be synthesized, entirely 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.


[0063] 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.


[0064] cDNAs can be synthesized on a substrate. Synthesis on the surface of a substrate may be accomplished using a chemical coupling procedure and a piezoelectric printing apparatus as described by Baldeschweiler et al. (PCT publication WO95/251116). Alternatively, the cDNAs can be synthesized on a substrate surface using a self-addressable electronic device that controls when reagents are added as described in 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.


[0065] 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 (U.S. Pat. No. 5,605,662). 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.


[0066] 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 neuroblastoma, or inhibiting or inactivating a therapeutically relevant gene related to the cDNA.


[0067] 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. Hybridization


[0068] 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 (Qiagen-Operon, Alameda Calif.), and chemiluminescent labeling (Promega, Madison, Wis.) are well known in the art.


[0069] 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.


[0070] 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 60°-68° C. (high stringency). At high stringency, hybridization complexes will remain stable only where the nucleic acids are completely complementary. In some membrane-based hybridizations, preferably 35% or most preferably 50%, formamide may be added to the hybridization solution to reduce the temperature at which hybridization is performed. Background signals may be reduced by the use of detergents such as Sarkosyl or TRITON X-100 (Sigma-Aldrich, St. Louis, Mo.) and a blocking agent such as denatured salmon sperm DNA. Selection of components and conditions for hybridization are well known to those skilled in the art and are reviewed in Ausubel et al. (1997, Short Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., Units 2.8-2.11, 3.18-3.19 and 4-6-4.9).


[0071] 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.)


[0072] Screening and Purification Assays


[0073] 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.


[0074] 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.


[0075] 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.


[0076] 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.


[0077] Protein Production and Uses


[0078] The full length cDNAs or fragments thereof may be used to produce purified proteins using recombinant DNA technologies described herein and taught in Ausubel (supra; Units 16.1-16.62). One of the advantages of producing proteins by these procedures is the ability to obtain highly-enriched sources of the proteins thereby simplifying purification procedures.


[0079] 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 algorithms included in RasMol software (University of Massachusetts, Amherst, Mass.) may be used to help determine which and how many amino acid residues in a particular portion of the protein may be substituted, inserted, or deleted without abolishing biological or immunological activity.


[0080] Expression of Encoded Proteins


[0081] Expression of a particular cDNA may be accomplished by cloning the cDNA into a vector and transforming this vector into a host cell. The cloning vector used for the construction of cDNA libraries in the LIFESEQ databases (Incyte Genomics, Palo Alto, Calif.) may also be used for expression. Such vectors usually contain a promoter and a polylinker useful for cloning, priming, and transcription. An exemplary vector may also contain the promoter for β-galactosidase, an amino-terminal methionine and the subsequent seven amino acid residues of β-galactosidase. The vector may be transformed into competent E. coli cells. Induction of the isolated bacterial strain with isopropylthiogalactoside (IPTG) using standard methods will produce a fusion protein that contains an N terminal methionine, the first seven residues of β-galactosidase, about 15 residues of linker, and the protein encoded by the cDNA.


[0082] 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.


[0083] 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.


[0084] 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 Saccharomvces 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).


[0085] Additionally, the vector may contain promoters or enhancers that increase gene expression. Many promoters are known and used in the art. Most promoters are host specific and exemplary promoters includes SV40 promoters for CHO cells; T7 promoters for bacterial hosts; viral promoters and enhancers for plant cells; and PGH promoters for yeast. Adenoviral vectors with the rous sarcoma virus enhancer or retroviral vectors with long terminal repeat promoters may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of secreted soluble protein may be recovered from the conditioned medium and analyzed using chromatographic methods well known in the art. An alternative method for the production of large amounts of secreted protein involves the transformation of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, and the like.


[0086] In addition to recombinant production, proteins or portions thereof may be produced manually, using solid-phase techniques (Stewart et al. (1969) Solid-Phase Peptide Synthesis, W H Freeman, San Francisco, Calif.; Merrifield (1963) J Am Chem Soc 5:2149-2154), or using machines such as the 431A peptide synthesizer (Applied Biosystems (ABI), Foster City, Calif.). Proteins produced by any of the above methods may be used as pharmaceutical compositions to treat disorders associated with null or inadequate expression of the genomic sequence.


[0087] Screening and Purification Assays


[0088] A protein or a portion thereof encoded by the cDNA may be used to screen a library or a plurality of molecules or compounds for a ligand with specific binding affinity or to purify a molecule or compound from a sample. The protein or portion thereof employed in such screening may be free in solution, affixed to an abiotic or biotic substrate, or located intracellularly. For example, viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a protein on their cell surface can be used in screening assays. The cells are screened against a library or a plurality of ligands and the specificity of binding or formation of complexes between the expressed protein and the ligand may be measured. The ligands may be agonists, antagonists, antibodies, DNA molecules, enhancers, small drug molecules, immunoglobulins, inhibitors, mimetics, peptide nucleic acid molecules, peptides, pharmaceutical agents, proteins, and regulatory proteins, repressors, RNA molecules, ribozymes, transcription factors, 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.


[0089] This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding the protein specifically compete with a test compound capable of binding to the protein or oligopeptide or fragment thereof. One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in U.S. Pat. No. 5,876,946. Molecules or compounds identified by screening may be used in a model system to evaluate their toxicity, diagnostic, or therapeutic potential.


[0090] The protein may be used to purify a ligand from a sample. A method for using a protein to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified ligand.


[0091] Production of Antibodies


[0092] 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.


[0093] 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.).


[0094] 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.


[0095] Antibody Arrays


[0096] In an alternative to yeast two hybrid system analysis of proteins, an antibody array can be used to study protein-protein interactions and phosphorylation. A variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until protein:antibody complexes are formed. Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest. In the alternative, a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti-DIG antibody which reveals where the protein of interest forms a complex. The identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.


[0097] Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt et al. (2000) Nat Biotechnol 18:989-94).


[0098] Assays Using Antibodies


[0099] Antibodies directed against antigenic determinant 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.


[0100] Protocols for detecting and measuring protein expression using either polyclonal or monoclonal antibodies are well known in the art. Examples include ELISA, RIA, fluorescent activated cell sorting (FACS) and arrays. Such immunoassays typically involve the formation of complexes between the protein and its specific antibody and the measurement of such complexes.


[0101] Labeling of Molecules for Assay


[0102] 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.).


[0103] The proteins and antibodies may be labeled for purposes of assay by joining them, either covalently or noncovalently, with a reporter molecule that provides for a detectable signal. A wide variety of labels and conjugation techniques are known and have been reported in the scientific and patent literature including, but not limited to U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.



DIAGNOSTICS

[0104] The cDNAs, or fragments thereof, may be used to detect and quantify differential gene expression; absence, presence, or excess expression of mRNAs; or to monitor mRNA levels during therapeutic intervention. Disorders associated with altered expression include cancers, particularly neuroblastoma.


[0105] Expression Profiles


[0106] A gene expression profile comprises the expression of a plurality of cDNAs or proteins as measured using assay well known in the art. The cDNAs of the invention may be used as elements on a substrate to produce an expression profile. In one embodiment, the profile is used to diagnose or monitor the progression of disease. The differences between gene expression in healthy and diseased tissues or cells can be assessed and cataloged.


[0107] For example, the cDNA or protein, may be labeled by standard methods and added to a biological sample from a patient under conditions for complex formation (specific binding). After an incubation period, the sample is washed and the amount of label (or signal) associated with complex formation, is quantified and compared with a standard value. If complex formation in the patient sample is significantly altered (higher or lower) in comparison to either a normal or disease standard, then differential expression indicates the presence of a disorder.


[0108] In order to provide standards for establishing differential expression, normal and disease expression profiles are established. This is accomplished by combining a sample taken from normal subjects, either animal or human, with a cDNA or protein under conditions for complex formation to occur. Standards may be obtained by comparing the expression levels from normal subject tissues with those from an experiment in which a known amount of a purified sequence is used. Standard expression levels obtained in this manner may be compared with those obtained from samples from patients who were diagnosed with a particular condition, disease, or disorder. Deviation from the standard toward those associated with a particular disorder is used to diagnose that disorder.


[0109] By analyzing changes in patterns of gene expression, disease can be diagnosed at earlier stages before the patient is symptomatic. The invention can be used to formulate a prognosis and to design a treatment regimen. The invention can also be used to monitor the efficacy of treatment. For treatments with known side effects, the array is employed to improve the treatment regimen. A dosage is established that causes a change in genetic expression patterns indicative of successful treatment. Expression patterns associated with the onset of undesirable side effects are avoided. This approach may be more sensitive and rapid than waiting for the patient to show inadequate improvement, or to manifest side effects, before altering the course of treatment.


[0110] In another embodiment, animal models which mimic a human disease can be used to characterize expression profiles associated with a particular condition, disease, or disorder; or treatment of the condition, disease, or disorder. Novel treatment regimens may be tested in these animal models using arrays to establish and then follow expression profiles over time. In addition, arrays may be used with cell cultures or tissues removed from animal models to rapidly screen large numbers of candidate drug molecules, looking for ones that produce an expression profile similar to those of known therapeutic drugs, with the expectation that molecules with the same expression profile will likely have similar therapeutic effects. Thus, the invention provides the means to rapidly determine the molecular mode of action of a drug.


[0111] 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.


[0112] Gene Expression Profiles


[0113] A gene expression profile comprises a plurality of cDNAs and a plurality of detectable hybridization complexes, wherein each complex is formed by hybridization of one or more probes to one or more complementary nucleic acids in a sample. The cDNAs of the invention are used as elements on a array to analyze gene expression profiles. In one embodiment, the array is used to monitor the progression of disease. Researchers 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 array is employed to improve the treatment regimen. A dosage is established that causes a change in genetic expression patterns indicative of successful treatment. Expression patterns associated with the onset of undesirable side effects are avoided. This approach may be more sensitive and rapid than waiting for the patient to show inadequate improvement, or to manifest side effects, before altering the course of treatment.


[0114] Experimentally, expression profiles can also be evaluated by methods including, but not limited to, differential display by spatial immobilization or by gel electrophoresis, genome mismatch scanning, representational discriminant analysis, transcript imaging, and by protein or antibody arrays. Expression profiles produced by these methods may be used alone or in combination. The correspondence between mRNA and protein expression has been discussed by Zweiger (2001, Transducing the Genome. McGraw-Hill, San Francisco, Calif.) and Glavas et al. (2001; T cell activation upregulates cyclic nucleotide phosphodiesterases 8A1 and 7A3, Proc Natl Acad Sci 98:6319-6342) among others.


[0115] In another embodiment, animal models which mimic a human disease can be used to characterize expression profiles associated with a particular condition, disorder or disease; or treatment of the condition, disorder or disease. Novel treatment regimens may be tested in these animal models using arrays to establish and then follow expression profiles over time. In addition, arrays may be used with cell cultures or tissues removed from animal models to rapidly screen large numbers of candidate drug molecules, looking for ones that produce an expression profile similar to those of known therapeutic drugs, with the expectation that molecules with the same expression profile will likely have similar therapeutic effects. Thus, the invention provides the means to rapidly determine the molecular mode of action of a drug.



THERAPEUTICS

[0116] The cDNAs and fragments thereof can be used in gene therapy. cDNAs can be delivered ex vivo to target cells, such as cells of bone marrow. Once stable integration and transcription and or translation are confirmed, the bone marrow may be reintroduced into the subject. Expression of the protein encoded by the cDNA may correct a disorder associated with mutation of a normal sequence, reduction or loss of an endogenous target protein, or overepression of an endogenous or mutant protein. Alternatively, cDNAs may be delivered in vivo using vectors such as retrovirus, adenovirus, adeno-associated virus, herpes simplex virus, and bacterial plasmids. Non-viral methods of gene delivery include cationic liposomes, polylysine conjugates, artificial viral envelopes, and direct injection of DNA (Anderson (1998) Nature 392:25-30; Dachs et al. (1997) Oncol Res 9:313-325; Chu et al. (1998) J Mol Med 76(3-4):184-192; Weiss et al. (1999) Cell Mol Life Sci 55(3):334-358; Agrawal (1996) Antisense Therapeutics, Humana Press, Totowa, N.J.; and August et al. (1997) Gene Therapy (Advances in Pharmacology, Vol. 40), Academic Press, San Diego, Calif.).


[0117] 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.


[0118] Molecules which regulate the activity of the cDNA or encoded protein are useful as therapeutics for neuroblastoma. 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.


[0119] 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.).


[0120] Model Systems


[0121] 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.


[0122] Transgenic Animal Models


[0123] Transgenic rodents that overexpress or underexpress a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents. (See, e.g., U.S. Pat. No. 5,175,383 and U.S. Pat. No. 5,767,337.) In some cases, the introduced gene may be activated at a specific time in a specific tissue type during fetal or postnatal development. Expression of the transgene is monitored by analysis of phenotype, of tissue-specific mRNA expression, or of serum and tissue protein levels in transgenic animals before, during, and after challenge with experimental drug therapies.


[0124] Embryonic Stem Cells


[0125] 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.


[0126] Knockout Analysis


[0127] 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.


[0128] Knockin Analysis


[0129] 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.


[0130] 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

[0131] I Construction of cDNA Libraries


[0132] RNA was purchased from Clontech Laboratories (Palo Alto, Calif.) or isolated from various tissues. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL reagent (Invitrogen). The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated with either isopropanol or ethanol and sodium acetate, or by other routine methods.


[0133] 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.).


[0134] In some cases, Stratagene (La Jolla, Calif.) was provided with RNA and constructed the corresponding cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen) using the recommended procedures or similar methods known in the art. (See Ausubel, supra, Units 5.1 through 6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (APB) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of the pBLUESCRIPT phagemid (Stratagene), pSPORT1 plasmid (Invitrogen), or pINCY plasmid (Incyte Genomics). Recombinant plasmids were transformed into XL1-BLUE, XL1-BLUEMRF, or SOLR competent E. coli cells (Stratagene) or DH5α, DH10B, or ELECTROMAX DH10B competent E. coli cells (Invitrogen).


[0135] In some cases, libraries were superinfected with a 5× excess of the helper phage, M13K07, according to the method of Vieira et al. (1987, Methods Enzymol 153:3-11) and normalized or subtracted using a methodology adapted from Soares (1994, Proc Natl Acad Sci 91:9228-9232), Swaroop et al. (1991, Nucleic Acids Res 19:1954), and Bonaldo et al. (1996, Genome Research 6:791-806). The modified Soares normalization procedure was utilized to reduce the repetitive cloning of highly expressed high abundance cDNAs while maintaining the overall sequence complexity of the library. Modification included significantly longer hybridization times which allowed for increased gene discovery rates by biasing the normalized libraries toward those infrequently expressed low-abundance cDNAs which are poorly represented in a standard transcript image (Soares, supra).


[0136] II Isolation and Sequencing of cDNA Clones


[0137] 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.


[0138] 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).


[0139] cDNA sequencing reactions were processed using standard methods or high-throughput instrumentation such as the CATALYST 800 thermal cycler (ABI) or the DNA ENGINE thermal cycler (MJ Research, Watertown, Mass.) in conjunction with the HYDRA microdispenser (Robbins Scientific, Sunnyvale, Calif.) or the MICROLAB 2200 system (Hamilton, Reno, Nev.). cDNA sequencing reactions were prepared using reagents provided by APB or supplied in sequencing kits such as the PRISM BIGDYE cycle sequencing kit (ABI). Electrophoretic separation of cDNA sequencing reactions and detection of labeled cDNAs were carried out using the MEGABACE 1000 DNA sequencing system (APB); the PRISM 373 or 377 sequencing systems (ABI) in conjunction with standard protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, supra, Unit 7.7).


[0140] III Extension of cDNA Sequences


[0141] 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.


[0142] 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.


[0143] 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 (Invitrogen), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 60° C., 1 min; Step 4: 68° C., 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68° C., 5 min; Step 7: storage at 4° C. In the alternative, the parameters for primer pair T7 and SK+ (Stratagene) were as follows: Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 57° C., 1 min; Step 4: 68° C., 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68° C., 5 min; Step 7: storage at 4° C.


[0144] 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.


[0145] 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.


[0146] The cells were lysed, and DNA was amplified by PCR using Taq DNA polymerase (APB) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94° C., 3 min; Step 2: 94° C., 15 sec; Step 3: 60° C., 1 min; Step 4: 72° C., 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72° C., 5 min; Step 7: storage at 4° C. DNA was quantified using PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions described above. Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the PRISM BIGDYE terminator cycle sequencing kit (ABI).


[0147] IV Assembly and Analysis of Sequences


[0148] Component nucleotide sequences from chromatograms were subjected to PHRED analysis (Phil Green, University of Washington, Seattle, Wash.) and assigned a quality score. The sequences having at least a required quality score were subject to various pre-processing algorithms to eliminate low quality 3′ ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, bacterial contamination sequences, and sequences smaller than 50 base pairs. Sequences were screened using the BLOCK 2 program (Incyte Genomics), a motif analysis program based on sequence information contained in the SWISS-PROT and PROSITE databases (Bairoch et al. (1997) Nucleic Acids Res 25:217-221; Attwood et al. (1997) J Chem Inf Comput Sci 37:417-424).


[0149] Processed sequences were subjected to assembly procedures in which the sequences were assigned to bins, one sequence per bin. Sequences in each bin were assembled to produce consensus sequences, templates. Subsequent new sequences were added to existing bins using BLAST (Altschul, 1990 (supra); Altschul,1993 (supra); Karlin et al. (1988) Proc Natl Acad Sci 85:841-845), BLASTn (vers.1.4, WashU), and CROSSMATCH software (Green, supra). Candidate pairs were identified as all BLAST hits having a quality score greater than or equal to 150. Alignments of at least 82% local identity were accepted into the bin. The component sequences from each bin were assembled using PHRAP (Green, supra). Bins with several overlapping component sequences were assembled using DEEP PHRAP (Green, supra).


[0150] Bins were compared against each other, and those having local similarity of at least 82% were combined and reassembled. Reassembled bins having templates of insufficient overlap (less than 95% local identity) were re-split. Assembled templates were also subjected to analysis by STITCHER/EXON MAPPER algorithms which analyzed the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced genes across tissue types, disease states, and the like. These resulting bins were subjected to several rounds of the above assembly procedures to generate the template sequences found in the LIFESEQ GOLD database (Incyte Genomics).


[0151] The assembled templates were annotated using the following procedure. Template sequences were analyzed using BLASTn (vers. 2.0, NCBI) versus GBpri (GenBank vers. 116). “Hits” were defined as an exact match having from 95% local identity over 200 base pairs through 100% local identity over 100 base pairs, or a homolog match having an E-value equal to or greater than 1×10−8. (The “E-value” quantifies the statistical probability that a match between two sequences occurred by chance). The hits were subjected to frameshift FASTx versus GENPEPT (GenBank version 109). In this analysis, a homolog match was defined as having an E-value of 1×10−8. The assembly method used above was described in U.S. Ser. No. 09/276,534, filed Mar.25, 1999, and the LIFESEQ GOLD user manual (Incyte Genomics).


[0152] Following assembly, template sequences were subjected to motif, BLAST, Hidden Markov Model (HMM; Pearson and Lipman (1988) Proc Natl Acad Sci 85:2444-2448; Smith and Waterman (supra), and functional analyses, and categorized in protein hierarchies using methods described in U.S. Ser. No. 08/812,290, filed Mar. 6, 1997; U.S. Ser. No. 08/947,845, filed Oct. 9, 1997; U.S. Pat. No. 5,953,727; and U.S. Ser. No. 09/034,807, filed Mar. 4, 1998. Template sequences may be further queried against public databases such as the GenBank rodent, mammalian, vertebrate, eukaryote, prokaryote, and human EST databases.


[0153] V Selection of Sequences, Microarray Preparation and Use


[0154] Incyte clones represent template sequences derived from the LIFESEQ GOLD assembled human sequence database (Incyte Genomics). In cases where more than one clone was available for a particular template, the 5′-most clone in the template was used on the microarray. For the UNIGEM series microarrays (Incyte Genomics), Incyte clones were mapped to non-redundant Unigene clusters (Unigene database (build 46), NCBI; Shuler (1997) J Mol Med 75:694-698), and the 5′ clone with the strongest BLAST alignment (at least 90% identity and 100 bp overlap) was chosen, verified, and used in the construction of the microarray. The UNIGEM V microarray (Incyte Genomics) contains 7075 array elements which represent 4610 annotated genes and 2,184 unannotated clusters. Table 1 shows the GenBank annotations for SEQ ID NOs: 1-365 of this invention as produced by BLAST analysis.


[0155] 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.


[0156] 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.


[0157] VI Preparation of Samples


[0158] Tissues and cell lines:


[0159] The neuroblastoma primary tumor cultures (P4 and P67) were established as described by Sarkar and Nuchtern (2000; Cancer Res 60:1908-1913) and cultured in MEM with 10% Fetal Calf Serum and antibiotics. The MYCN inducible neuroblastoma cell line TET21 derived from the SH-EP line was obtained from Dr. Manfred Schwab. The TET21 cells were grown in 10% serum/RPMI with penicillin and streptomycin as described by Lutz, supra). Tetracycline was used at a concentration of 1 μM.


[0160] cDNA Northerns:


[0161] Total RNA from induced and non-induced TET21 cells was isolated using the RNEASY kit (Qiagen) and reverse-transcribed using SUPERSCRIPT-II reverse transcriptase (Invitrogen) with the CDS and SMART-II primer oligonucleotides in the SMART cDNA synthesis kit (Clontech). The resulting cDNA was amplified for 15 cycles. This method of Franz et al. (1999; Nucleic Acid Res 27:e3) was used to maximize detection sensitivity for MCM7 and other messages upon MYCN induction and to efficiently use tumor mRNA available in limiting quantity. The number of amplification cycles was optimized with electrophoretic analysis of the cDNA on 1.4% agarose to prevent over-cycling.


[0162] Equal amounts of MYCN-induced and non-induced cDNA was electrophoresed and electroblotted onto nylon membranes. Probes for MYCN and MCM7 were generated from PCR amplified fragments (MCYN: 5′-CCTGCCCGCCGAGCTCG-3′ and reverse 5′-CTCGCTGGACTGAGCCCA-3′, MCM7: 5′-AGCAGAACATACAGCTACCTG-3′ and reverse 5′-CCCTTGTCTCCTAGAAGAGAG-3′) and either random hexamer labeled with α-P32-CTP or labeled with alkaline phosphatase using ALKPHOSDIRECT (APB). Probes were hybridized at 42° C. in ULTRAHYB hybridization buffer (Ambion) or alkaline phosphatase hybridization buffer overnight and washed. Probe signals were detected using a PHOSPHOIMAGER cassette (APB). Expression levels were normalized against the signal from β-actin.


[0163] Probe Preparation


[0164] Total RNA was extracted from exponentially growing cultures using an RNEASY kit (Qiagen) according to the instructions of the manufacturer. Poly(A) RNA was purified using the OLIGOTEX mRNA kit (Qiagen). Each poly(A) RNA sample was reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/μl oligo-d(T) primer (21 mer), 1× first strand buffer, 0.03 units/ul RNAse inhibitor, 500 uM dATP, 500 uM dGTP, 500 uM dTTP, 40 uM dCTP, and 40 uM either dCTP-Cy3 or dCTP-Cy5 (APB). The reverse transcription reaction was performed in a 25 ml volume containing 200 ng poly(A) RNA using the GEMBRIGHT kit (Incyte Genomics). Specific control poly(A) RNAs (YCFRO6, YCFR45, YCFR67, YCFR85, YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were synthesized by in vitro transcription from non-coding yeast genomic DNA (W. Lei, unpublished). As quantitative controls, control mRNAs (YCFR06, 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.


[0165] cDNAs were purified using two successive CHROMA SPIN 30 gel filtration spin columns (Clontech). Cy3- and CyS-labeled reaction samples were combined as described below and ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The cDNAs were then dried to completion using a SpeedVAC system (Savant Instruments, Holbrook, N.Y.) and resuspended in 14 μl 5X SSC, 0.2% SDS.


[0166] VII Hybridization and Detection


[0167] Hybridization reactions contained 9 μl of sample mixture containing 0.2 μg each of Cy3 and Cy5 labeled cDNA synthesis products in 5X 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.


[0168] 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 20X 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.


[0169] In two separate scans, the mixed gas multiline laser excited the two fluorophores sequentially. Emitted light was split, based on wavelength, into two photomultiplier tube detectors (PMT R1477; Hamamatsu Photonics Systems, Bridgewater, N.J.) corresponding to the two fluorophores. Appropriate filters positioned between the microarray and the photomultiplier tubes were used to filter the signals. The emission maxima of the fluorophores used were 565 nm for Cy3 and 650 nm for Cy5. Each microarray was typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus was capable of recording the spectra from both fluorophores simultaneously.


[0170] 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.


[0171] 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.


[0172] A grid was superimposed over the fluorescence signal image such that the signal from each spot was centered in each element of the grid. The fluorescence signal within each element was then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis was the GEMTOOLS gene expression analysis program (Incyte Genomics). Significance was defined as signal to background ratio exceeding 2× and area hybridization exceeding 40%.


[0173] VIII Data Analysis and Results


[0174] Array elements that exhibited at least 4-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 (Incyte Genomics). Positive differential expression values (downregulation) represent non-amplified tumor (P4) relative to MYCN amplified tumor (P67). Negative differential expression values (upregulation) represent MYCN amplified tumor (P67) relative to non-amplified tumor (P4). Table 1 identifies upregulated and downregulated cDNAs. 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.


[0175] IX Further Characterization of Differentially Expressed cDNAs and Proteins


[0176] Clones were blasted against the LIFESEQ Gold 5.1 database (Incyte Genomics) and an Incyte template and its sequence variants were chosen for each clone. The template and variant sequences were blasted against GenBank database to acquire annotation. The nucleotide sequences were translated into amino acid sequences which were blasted against the GenPept and other protein databases to acquire annotation and characterization, i.e., structural motifs. Different templates identified in Table 1 may share an identical GenBank annotation and single clones may be mapped to more than one template. These templates represent related homologs or splice variants. Templates with no match to a sequence in the GenBank database are identified in Table 1 as “Incyte Unique”.


[0177] Percent sequence identity can be determined electronically for two or more nucleic acid or amino acid sequences using the MEGALIGN program, a component of LASERGENE software (DNASTAR). The percent identity between two amino acid sequences is calculated by dividing the length of sequence A, minus the number of gap residues in sequence A, minus the number of gap residues in sequence B, into the sum of the residue matches between sequence A and sequence B, times one hundred. Gaps of low or of no homology between the two amino acid sequences are not included in determining percentage identity.


[0178] Sequences with conserved protein motifs may be searched using the BLOCKS search program. This program analyses sequence information contained in the Swiss-Prot and PROSITE databases and is useful for determining the classification of uncharacterized proteins translated from genomic or cDNA sequences (Bairoch, supra; Attwood, supra). PROSITE database is a useful source for identifying functional or structural domains that are not detected using motifs due to extreme sequence divergence. Using weight matrices, these domains are calibrated against the SWISS-PROT database to obtain a measure of the chance distribution of the matches.


[0179] 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. Pfam is a large collection of multiple sequence alignments and hidden Markov models covering many common protein domains. Version 5.5 of Pfam (Sept 2000) contains alignments and models for 2478 protein families, based on the Swissprot 38 and SP-TrEMBL 11 protein sequence databases.


[0180] X 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] XI Expression of the Encoded Protein


[0187] 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.


[0188] 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.).


[0189] XII Production of Specific Antibodies


[0190] 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.


[0191] 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 antigenic determinants of the protein. The optimal sequences for immunization are usually at the C-terminus, the N-terminus, and those intervening, hydrophilic regions of the protein that are likely to be exposed to the external environment when the protein is in its natural conformation. Typically, oligopeptides about 15 residues in length are synthesized using an 431 peptide synthesizer (ABI) using Fmoc-chemistry and then coupled to keyhole limpet hemocyanin (KLH; Sigma-Aldrich) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester. If necessary, a cysteine may be introduced at the N-terminus of the peptide to permit coupling to KLH. Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated goat anti-rabbit IgG.


[0192] 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.


[0193] Such clones are expanded and subjected to 2 cycles of cloning at 1 cell/3 wells. Cloned hybridomas are injected into pristane-treated mice to produce ascites, and monoclonal antibody is purified from the ascitic fluid by affinity chromatography on protein A (APB). Monoclonal antibodies with affinities of at least 108 M−1, preferably 109 to 1010M−1 or stronger, are made by procedures well known in the art.


[0194] XIII Purification of Protein Using Specific Antibodies


[0195] Naturally occurring or recombinant protein is purified by immunoaffinity chromatography using antibodies specific for the protein. An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected.


[0196] XIV Screening Molecules for Specific Binding with the cDNA or Protein


[0197] The cDNA or fragments thereof and the protein or portions thereof are labeled with 32P-dCTP, Cy3-dCTP, Cy5-dCTP (APB), or BIODIPY or FITC (Molecular Probes), respectively. Candidate molecules or compounds previously arranged on a substrate are incubated in the presence of labeled nucleic or amino acid. After incubation under conditions for either a cDNA or a protein, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed. The binding molecule is identified by its arrayed position on the substrate. Data obtained using different concentrations of the nucleic acid or protein are used to calculate affinity between the labeled nucleic acid or protein and the bound molecule. High throughput screening using very small assay volumes and very small amounts of test compound is fully described in U.S. Pat. No. 5,876,946.


[0198] 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 1SEQ IDNOTemplate IDClone IDGenBank HitE-valueAnnotationBal DE11497123CB11497123g12947821.00E−107phosphomevalonate kinase [Homo sapiens]89.322985802CB13553729g34208460fibronectin [Danio rerio]70.23475532.42859033g4398583.00E−35Human MHC Class I HLA heavy chain (HLA-B-7301) mRNA,58.2complete cds.43138290CB11870965g23885550alpha2(I) collagen [Homo sapiens]51.15474310.401672744g8180protein-glutamine gamma-glutamyltransferase [Bos taurus]49.16410580.161445767g3869970prebeta-migrating plasminogen activator inhibitor41.4[Homo sapiens]7337518.251674454g1809240connective tissue growth factor [Homo sapiens]40.581303785CB1079576g343881.00E−145lipocortin (AA 1-346) [Homo sapiens]36.591044033.41514989g21818710Gig 1 protein [Homo sapiens]35.5101000222.31690313g8615210prostacyclin-stimulating factor (PSF) [Homo sapeins]33.411403873.42329216g1799671.00E−160carbonic anhydrase VII [Homo sapiens]33121383105.124049957g23408331.00E−114SM22 alpha [Homo sapiens]29.6131383354.131572533g69837291.00E−86dJ977B1.5 (myosin regulatory light chain29.62, smooth muscle isoform) [Homo sapiens]14697785CB12495131g3071228.00E−77lectin precursor [Homo sapiens]28.815420115CB11904751g1796460complement component Cls [Homo sapiens]28.8161101453.22949427g233983.00E−641-8U [Homo sapiens]25.2171399366.202055534g5118690thrombospondin [Mus musculus]24.6183072333CB11447903g3981641.00E−145insulin-like growth factor binding protein 3 [Homo sapiens]24.4191270681.11804548g25887891.00E−22p21/WAF1 [Felis catus]23.9201505038CB11987358g5368981.00E−166follistatin-related protein precursor [Homo sapiens]23.2211035602.51854220g30045020quiescin [Homo sapiens]22.1221330167.31001730g321311.00E−121putative p33 [Homo sapiens]21.1231003386CB11664320g295390precursor of Clr (AA −17 to 688) [Homo sapiens]21.1241097334.12483605g19249820integral membrane serine protease Seprase [Homo sapiens]20.125959142CB12804667g29951380thrombospondin 2 [Bos taurus]20.1261359783CB11798209g4582285.00E−75extracellular protein [Homo sapiens]19.927063646CB1557012g5127780protein with miniactivin activity [synthetic construct]19.5281519595CB12056395g15181070transforming growth factor induced protein19.4[Oryctolagus cuniculus]292054176CB13142736g60702533.00E−41Dickkopf-3 [Homo sapiens]17.8301312325CB11319608g2198986.00E−961-caldesmon II [Homo sapiens]17.731022404.251319608g1801949.00E−96caldesmon [Homo sapiens]17.7321787335CB11958902g58154610insulin-like growth factor binding protein 5 protease17.5[Rattus norvegicus]331193648.71851696g32734058.00E−79laminin alpha 4 [Mus musculus]17.1341193648.11851696g31689930dJ142L7.1 (Laminin Alpha 4 LIKE isoform 1) [Homo sapiens]17.1351867861CB11448051g7263240fibrillin-1 [Mus musculus]16.3365511889CB11650238g26128682.00E−94down syndrome candidate region 1; one of four16.3alternatively spliced exon 1 [Homo sapiens]373094768CB12902903g11774764.00E−67interferon-inducible protein [Homo sapiens]16.2381256895CB11720056g69961558.00E−81prion protein (p27-30) [Homo sapiens]15.5392019981CB11733490g1886260moesin B [Homo sapiens]15.3402708240CB13084122g26541980deleted in liver cancer-1 [Homo sapiens]15.2411092427.11313183g3395480transforming growth factor-beta 1 binding protein precursor14.9[Homo sapiens]42351841.71852047g1871890lysyl oxidase [Homo sapiens]14.743022221.431314882g41042320collagen alpha3(VI) [Mus musculus]14.5442190217CB1078783g347561.00E−84myosin regulatory light chain [Homo sapiens]13.845410910.32518178g300971.00E−152pro-alpha I (II) collagen (313 AA; AA 975-271c)13.7[Homo sapiens]461966280CB11700077g3399920tumor necrosis factor [Homo sapiens]13.647430669.391572555g2075082.00E−95alpha-tropomyosin 5b [Rattus norvegicus]13.548430669.231572555g48843936.00E−83hypothetical protein [Homo sapiens]13.5491870753CB1782235g14189281.00E−154prepro-alphal(I) collagen [Homo sapiens]13.1503173735CB1521139g1934400guanylate binding protein isoform I [Mus musculus]12.8511330185.142868138g1825142.00E−90ferritin light chain [Homo sapiens]12.3522314132CB13118643g300821.00E−89alpha 1(VIII) collagen [Homo sapiens]12.1532508205CB12057296g57485810dJ65P5.1 (reticulocalbin 1, EF-hand calcium binding domain)12[Homo sapiens]543326672CB11995380g11954831.00E−73microsomal glutathione transferase [Homo sapiens]11.955234202.341995380g3068083.00E−78glutathione S-transferase [Homo sapiens]11.956078242CB11319020g6939330carbonate dehydratase [Homo sapiens]11.857220943.20417451g314410Human mRNA for integrin beta 1 subunit.11.8581383320.131558081g1984660type IV collagenase [Mus musculus]11.5593526170CB12242648g1819470erythroid differentiation protein precursor [Homo sapiens]11.560184081.24027775g5072526.00E−35ferritin heavy chain [Homo sapiens]11.2611821331CB11394401g8958400lrp [Homo sapiens]10.9623660006CB11720114g314381.00E−153integrin alpha 5 subunit precursor [Homo sapiens]10.963089172.133693273g1626940aspartyl (asparaginyl) beta hydroxylase [Bos taurus]10.8643084563CB12852042g1810712.00E−89cysteine-rich protein [Homo sapiens]10.665241227.171402715g7044411.00E−146unknown [Homo sapiens]10.466348151.21618422g384161.00E−143cyclin D2 [Homo sapiens]10.4671720808CB12606307g1808251.00E−165collagen type IV alpha 5 chain [Homo sapiens]10.368998552.6459651g28221691.00E−132homeodomain protein HOXA9 [Homo sapiens]10.369040652.352508261g81765251.00E−119interferon-inducible myeloid differentiation transcriptional10.2activator [Homo sapiens]70040652.362508261g1845690interferon-gamma induced protein [Homo sapiens]10.271082155CB1522294g8993000gpStaf50 [Homo sapiens]10.172190144CB11668794g4878094.00E−78cell surface protein [Homo sapiens]9.973234537.31718651g30468750ecto-5'-nucleotidase [Mus musculus]9.6741088425.1162769g1882561.00E−134cell surface glycoprotein [Homo sapiens]9.675254547.13134070g76347798.00E−58HDCMA39P [Homo sapiens]9.5762676170CB13820761g55324110decorin variant A [Homo sapiens]9.4771092181.12105963g3072691.00E−120HLA-DRB1 [Homo sapiens]9.478471362.331720149g72396981.00E−138myosin light chain kinase isoform 2 [Homo sapiens]9.379471362.271720149g72396965.00E−86myosin light chain kinase [Homo sapiens]9.3801162416.12102320g2112053.00E−37asma gene product [Gallus gallus]9.381252151.121599344g67063350laminin gamma 1 precursor [Anopheles gambiae]9.282252151.71599344g1869644.00E−99laminin B2 chain [Homo sapiens]9.283358892.12057260g35502830XRP2 protein [Homo sapiens]9.2841296867CB13506985g1801170antigen CD36 [Homo sapiens]9.185337518.73506985g1801110antigen CD36 [Homo sapiens]9.1861344279CB12771046g5447550aminopeptidase N, APN {type II membrane protein} {EC9.13.4.11.2} [Oryctolagus cuniculus]872731776CB12057601g72943190CG6778 gene product [Drosophila melanogaster]9.1881090035.11994472g6734175.00E−66class II antigen [Homo sapiens]9891089929.92683564g54782221.00E−143MHC class II antigen [Homo sapiens]9902723092CB12633207g18052700endothelial PAS domain protein 1 [Mus musculus]8.9912174489CB12173208g1808030alpha-1 type IV collagen [Homo sapiens]8.9921253978CB11867652g54412460branching enzyme 1 [Phaseolus vulgaris]8.8932274011CB11909488g72922135.00E−44CG1275 gene product [Drosophila melanogaster]8.8943119737CB12733928g360341.00E−110rhoC coding region (AA 1-193) [Homo sapiens]8.8951384695.1021514318g379870Human XIST, coding sequence ‘a’ mRNA (locus DXS399E).8.796257332CB11402228g573810T-plastin [Rattus norvegicus]8.7972972880CB11636171g64750310sushi-repeat-containing protein [Mus musculus]8.598550425CB1550425g13734270FLT4 ligand DHM [Homo sapiens]8.599014284CB11822716g41061260dipeptidyl peptidase 1 [Cams familiaris]8.51001091854.71708528g12039690filamin [Homo sapiens]8.4101138709.52844989g60312120heat shock protein hsp40 homolog [Homo sapiens]8.4102375954.11404153g13817920H-cadherin [Homo sapiens]8.41031262781CB13511216g32015890endoglin [Homo sapiens]8.3104282761.163511216g32015890endoglin [Homo sapiens]8.31053090708CB11283532g380594705T4 oncofetal trophoblast glycoprotein [Homo sapiens]8.3106230062.41711206g1830846.00E−8821 kd basic fibroblast growth factor (ctg start codon; put.);8.2putative [Homo sapiens]107483043CB11854277g14058930MHC class 1 chain-related protein A [Homo sapiens]8.1108348205.91854277g14058931.00E−139MHC class 1 chain-related protein A [Homo sapiens]8.11091256295.181702350g4031281.00E−91[Human gadd45 gene, complete cds.], gene product8[Homo sapiens]110875668CB11418741g3068050G protein-coupled receptor kinase [Homo sapiens]81111180189.12664388g40636300Homo sapiens clone IMAGE 286356.81123109992CB12483173g376371.00E−178VAC protein (AA 1-320) [Homo sapiens]81131250434CB11711151g66363170hypoxia-inducible factor 1 alpha [Homo sapiens]7.81141327838.13721987g29470541.00E−18Gene product with similarity to Rat P8 [Homo sapiens]7.71152021477CB12190284g16695472.00E−91RBP-MS/type 1 [Homo sapiens]7.7116235171.201940994g16637040KIAA0242 protein [Homo sapiens]7.7117149832CB1604856g4949891.00E−152nicotinamide N-methyltransferase [Homo sapiens]7.61181759127CB11759127g4090590lysyl hydroxylase [Rattus norvegicus]7.61192048551CB12048551g14954631.00E−68H. sapiens mRNA for metallothionein isoform 1R.7.61203282941CB1155904g35995214.00E−77musculin [Homo sapiens]7.61212733135CB12806166g2918880cathepsin B [Homo sapiens]7.51222176269CB11405940g58522950lysyl hydroxylase isoform 2 [Mus musculus]7.51231218607CB1477045g10300531.00E−149rtvp-1 [Homo sapiens]7.51241553795CB11906574g10223230collagen alpha-2(IV) chain [Mus musculus]7.4125238538.221906574g81017249.00E−40canstatin [Homo sapiens]7.4126246546.92936505g30435970Homo sapiens mRNA for KIAA0537 protein, complete cds.7.4127234223.141672442g2195100collagen alpha 1(V) chain precursor [Homo sapiens]7.21282054053CB12054053g1811231.00E−105cleavage signal 1 protein [Homo sapiens]7.11291613766CB11640161g65632522.00E−32G-protein gamma-12 subunit [Homo sapiens]7.1130233454.31636639g45196211.00E−67OASIS protein [Mus musculus]7.1131347699.131358285g551220ufo [Mus musculus]71323531583CB11358285g2387750putative tyrosine kinase receptor = UFO [human, NIH3T3,7Peptide, 894 aa] [Homo sapiens]133407096.14630625g28323460thioredoxin reductase [Homo sapiens]7134482411.262304121g9994540TX protease precursor [Homo sapiens]6.9135482411.252304121g9039340cysteine protease [Homo sapiens]6.91361258943CB1434771g13778940OB-cadherin-1 [Homo sapiens]6.91371327030.1450574g8296233.00E−86myosin regulatory light chain [Homo sapiens]6.9138025595.221962971g81707140laminin beta 2 chain; S-laminin [Homo sapiens]6.9139995174.11975129g65804116.00E−39dJ467L1.2 (vesicle-associated membrane protein 36.8(cellubrevin)) [Homo sapiens]1401709732CB1269456g296260CALLA protein (AA 1-750) [Homo sapiens]6.71411040610.4692827g363861.00E−131SB-2-beta precursor polypeptide (aa −29 to 229) [Homo sapiens]6.7142055498.61865767g27695620ZYG homologue [Homo sapiens]6.7143181172CB12503037g4840511.00E−121placental protein 5 (PP5) [Homo sapiens]6.61442705515CB11846209g1846570transfer RNA-Trp synthetase [Homo sapiens]6.5145480228.31997250g3398990Human transposon-like element mRNA.6.5146360929.39063038g3383361.00E−98spermidine/spermine N1-acetyltransferase [Homo sapiens]6.51471989087CB11603057g372650TRAM protein [Homo sapiens]6.5148995068.161904994g5460880CAP, 38 kda intracellular serine proteinase inhibitor6.4[Homo sapiens]1491217216.11976279g12973301.00E−140DOC-2 [Homo sapiens]6.4150474426.51281473g19463470RNA polymerase II elongation factor ELL2 [Homo sapiens]6.4151350521.222078364g37218780DR5 [Homo sapiens]6.41521075592.61686585g5055891.00E−154insulin-like growth factor binding protein 5 (IGFBP5)6.4gene product [Homo sapiens]1531485867CB11959565g54997210eRF1 [Homo sapiens]6.41542515360CB11887959g23702020procollagen alpha 2(V) [Homo sapiens]6.31553290944CB13598222g61658820collagen type XI alpha-1 isoform A [Homo sapiens]6.3156441206.152849603g64921300urokinase receptor-associated protein uPARAP [Homo sapiens]6.31571712327CB11712327g2024040Wnt-5a [Mus musculus]6.21581393778CB12056987g2123830myosin heavy chain [Gallus gallus]6.1159480127.442056987g1890367.00E−11nonmuscle myosin heavy chain (NMHC) [Homo sapiens]6.11601870941CB11870941g2974080P63 protein [Homo sapiens]6.11612495110CB13176845g38691130TRKA [Homo sapiens]6.1162034711.33425195g70206110unnamed protein product [Homo sapiens]6.1163251776.14418731g34786970integrin beta-5 [Mus musculus]6.1164239511.51821971g1897300platelet-derived growth factor receptor [Homo sapiens]6.1165989878.11997703g65634080connexin 43 [Homo sapiens]61661558664CB12018222g346561.00E−113MHC-encoded proteasome subunit gene [Homo sapiens]61673602501CB13602501g2008820retinoid X receptor-gamma [Mus musculus]61685549580CB11736926g294240beta-1,4-galactosyltransferase (AA −77 to 323) [Homo sapiens]61692687977CB11526282g6660431.00E−55NMB [Homo sapiens]61703168062CB11662688g13881971.00E−117low-Mr GTP-binding protein Rab32 [Homo sapiens]5.9171245367.23215205g40391179.00E−64PEA-15 protein [Cricetulus griseus]5.9172470587CB13940755g1799480cathepsin D [Homo sapiens]5.91731631074CB1064286g42398830Glutamine: fructose-6-phosphate amidotransferase [Homo sapiens]5.8174347829.12185448g5171790YAP65 (Yes-associated protein of 65 kDa MW) [Mus musculus]5.7175347699.112058242g2387750putative tyrosine kinase receptor = UFO [human,5.7NIH3T3, Peptide, 894 aa] [Homo sapiens]1761251672.11453450g372270tenascin [Homo sapiens]5.61771291022CB11291022g13360270ICE-LAP6 [Homo sapiens]5.5178237405.192380381g6027031.00E−1752,4-dienoyl-CoA reductase [Homo sapiens]5.51792685676CB12513883g5173500H. sapiens MTIX gene for metallothionein IX.5.5180010672CB1549196g1801300cell adhesion molecule [Homo sapiens]5.4181234630.58549196g77051570CD44R4 [Homo sapiens]5.4182332595.53249851g29794200PCDH7 (BH-Pcdh)b [Homo sapiens]5.4183332595.83249851g35133120BH-protocadherin-a [Mus musculus]5.4184335086.13602403g15430680CHASE [Homo sapiens]5.41851342493CB11453748g63819890adipocyte-derived leucine aminopeptidase [Homo sapiens]5.4186232691.202505425g1908771.00E−102ras-like protein [Homo sapiens]5.4187238814.21417211g14187820erm[Homo sapiens]5.3188201571.1959745g20721810Rat osteoprotegerin (OPG) protein, complete sequence5.3[Rattus norvegicus]189199882.51449824g26686150similar to drosophila peroxidasin precursor (PID:g531385)5.3[Caenorhabditis elegans]190237487.222380042g7910472.00E−60gamma subunit of sodium potassium ATPase like [Homo sapiens]5.2191237487.212380042g7910473.00E−51gamma subunit of sodium potassium ATPase like [Homo sapiens]5.2192305557CB1029564g26657925.00E−91caveolin-2 [Homo sapiens]5.21931378745CB1147184g1627790calpactin 1 heavy chain (p36) [Bos taurus]5.21941818836CB11818836g9842870NDP52 [Homo sapiens]5.2195137946.3690994g72430271.00E−168KIAA1323 protein [Homo sapiens]5.21962110909CB12825369g1650090progesterone-induced protein [Oryctolagus cuniculus]5.2197200578.11397926Incyte Unique5.2198259592CB1197207g59118571.00E−145hypothetical protein [Homo sapiens]5.21995584521CB11965863g2199251.00E−76MGC-24 precursor [Homo sapiens]5.1200399428.71491445g37775450nonsyndromic hearing impairment protein [Mus musculus]5.1201117509.43012290g10549031.00E−156gamma-sarcoglycan [Homo sapiens]5.12023255458CB11597330g28042730alpha actinin 4 [Homo sapiens]5.12031430889CB11856520g41074331.00E−120hypothetical protein [Homo sapiens]5204445048.61856520g42181851.00E−124hypothetical protein [Homo sapiens]52054946593CB12852818g3377670cerebroside sulfate activator protein [Homo sapiens]5206350605.454114209g4531800lamin A [Rattus norvegicus]52071413644CB11413644g9753110adenyl cyclase-associated protein 2 [Rattus norvegicus]5208984009.21446475g12259790H. sapiens mRNA for HMGI-C protein.4.9209627662CB11631511g41021820phosphoenolpyruvate carboxykinase [Mus musculus]4.92101382932.112175008g11550110nidogen[Homo sapiens]4.92112721850CB11624024g15428831.00E−86progression associated protein [Homo sapiens]4.9212994902.12059691g25847894.00E−18vacuolar proton-ATPase subunit M9.2 [Homo sapiens]4.9213442744.171610993g1830020guanylate binding protein isoform I [Homo sapiens]4.8214442744.211610993g1934400guanylate binding protein isoform I [Mus musculus]4.82151908920CB12134356g16948282.00E−40S100 calcium-binding protein A13 (S100A13) [Homo sapiens]4.8216399101.312134356g16948281.00E−40S100 calcium-binding protein A13 (S100A13) [Homo sapiens]4.8217183198CB1924319g7938411.00E−172nuclear protein [Homo sapiens]4.82181397781.71522716g3402190vimentin [Homo sapiens]4.8219899496.9812141g1836130granulin [Homo sapiens]4.82202111330CB11975209g16173194.00E−81vasodilator-stimulated phosphoprotein [Homo sapiens]4.8221331591.12452650g26550396.00E−61tumor suppressing STF cDNA 3 [Homo sapiens]4.8222337119.82488567g1794071.00E−134brain-derived neurotrophic factor [Homo sapiens]4.8223245011.112232471g41939461.00E−87p35srj [Homo sapiens]4.72241988468CB12232471g41939462.00E−88p35srj [Homo sapiens]4.7225331470.81457726g30437080KIAA0592 protein [Homo sapiens]4.7226411388CB1591358g1824831.00E−112prefibroblast collagenase inhibitor [Homo sapiens]4.7227253450.91347232g44266290protocadherin [Rattus norvegicus]4.7228351209.163686211g1790950acid sphingomyelinase [Homo sapiens]4.72292124320CB12204916g65732560coatomer protein gamma2-COP [Mus musculus]4.7230903876.1548019g82488540JAK1 protein tyrosine kinase [Mus sp.]4.72311238339CB12108793g5416131.00E−119platelet-endothelial tetraspan antigen 3 [Homo sapiens]4.6232245310.362108793g5416131.00E−125platelet-endothelial tetraspan antigen 3 [Homo sapiens]4.62332696735CB12696735g17832051.00E−160calponin [Homo sapiens]4.6234338036.21449054g8089151.00E−156tumor necrosis factor receptor type 1 associated4.6protein [Homo sapiens]235236484.151922533g66364980signal transducer and activator of transcription 1;4.6STAT1 [Rattus norvegicus]236232719.2537580g25828300alpha 1 integrin [Gallus gallus]4.6237462249.11830083g54102740hypothetical 19.5 kDa protein [Homo sapiens]4.62381187408.1030672g1780841.00E−125adenylyl cyclase-associated protein [Homo sapiens]4.6239627856CB11559756g26655190tyrosyl-tRNA synthetase [Homo sapiens]4.6240553078CB11985104g58235910adipophilin [Bos taurus]4.6241048612.151975268g2060670phosphoenolpyruvate carboxykinase [Rattus norvegicus]4.5242048612.121975268g3073331.00E−100phosphoenolpyruvate carboxykinase [Homo sapiens]4.52431099779.11612306g26623750oligosaccharyltransferase [Homo sapiens]4.52441520855CB1179929g14776510plectin [Homo sapiens]4.52451179282.12870970Incyte Unique4.52462770449CB11658320g4971740beta-hexosaminidase [Mus musculus]4.52471430336CB1030291g1786991.00E−180annexin IV (placental anticoagulant protein II) [Homo sapiens]4.5248903105.6544213g70214491.00E−126steroid sensitive gene-1 protein [Rattus norvegicus]4.52491327417.142211625g1865133.00E−28interferon-gamma [Homo sapiens]4.42501327417.102211625g1865131.00E−121interferon-gamma [Homo sapiens]4.4251230712.242814551g22749660Cdc42-interacting protein 4 [Homo sapiens]4.4252982520.12986240g75735324.00E−93dJ136O14.2 (collagen, type X, alpha 1) [Homo sapiens]4.4253311807CB1821141g72958550CG17259 gene product [Drosophila melanogaster]4.42541479370CB11626460g499440mannosyl-oligosaccharide 1,3-1,6-alpha-mannosidase4.4[Mus musculus]2552993696CB12884613g69001040glucose-regulated protein [Homo sapiens]4.42564004223CB11810945g4523201.00E−114rab 13 [Homo sapiens]4.4257453835.191723035g68222728.00E−69Ras negative regulator Rabex-5/Rin2 [Mus musculus]4.4258391741.161634279g360610peptide transporter [Homo sapiens]4.3259391741.641634279g360610peptide transporter [Homo sapiens]4.32601382958.263876715g3001690APPH = amyloid precursor protein homolog [human,4.3placenta, Peptide, 763 aa] [Homo sapiens]261232567.41577614g4040240follistatin [Bos taurus]4.32621720770CB12189762g76886991.00E−116RER1 protein [Homo sapiens]4.3263253987.19700559g3953382.00E−55helix-loop-helix protein [Homo sapiens]4.22642047630CB11381654g33417150asparagine synthetase [Homo sapiens]4.2265238203.11999864g3402370vinculin [Homo sapiens]4.2266899410.51724967g41653260plasma membrane calcium ATPase isoform 1 [Homo sapiens]4.2267474311.32736056g16577520FE65-like protein [Homo sapiens]4.22682169835CB11003486g5589990Shcp52 [Mus musculus]4.1269290021.111003486g18990551.00E−153p66shc [Homo sapiens]4.1270267324CB12132217g19058740carboxyl terminal LIM domain protein [Homo sapiens]4.12712119372CB11889060g4026660calpain II 80 kDa subunit [Rattus norvegicus]4.12722818482CB12668334g360610peptide transporter [Homo sapiens]4.12731330231.112594308g4307561.00E−116ME491/CD63 antigen [Homo sapiens]4.12741330117.5692201g17912891.00E−137MHC class II HLA-DQ [Homo sapiens]4.1275233402.32056290g16637260MNB [Homo sapiens]4.12761622313CB11901061g58803170lysyl hydroxylase 3 [Mus musculus]4.12772939887CB11375115g12355590responsible for hereditary multiple exotosis [Mus musculus]4.12781804120CB11901095g49597050fibulin-2 [Mus musculus]4279245485.121901095g48841201.00E−129hypothetical protein [Homo sapiens]42801285395CB1015834g30642630protein 4.1G [Mus musculus]4281036391.3399035g370741.00E−159transcription elongation factor [Homo sapiens]−4282036391.13399035g370741.00E−171transcription elongation factor [Homo sapiens]−4283474435.161610523g3071552.00E−86MAC30 [Homo sapiens]−42842495292CB12495292g22675850transcription intermediary factor 1 [Homo sapiens]−4.1285251651.41645766g4560902.00E−76effector cell protease receptor 1 [Homo sapiens]−4.22865408483CB13493061g2975290NF-M [Mus musculus]−4.2287347876.6103669g11841070DNA replication initiator protein [Xenopus laevis]−4.22881289007CB11986737g66900951.00E−145tetraspanin protein [Homo sapiens]−4.3289233301.18814216g1801730putative [Homo sapiens]−4.32902157771CB12825656g1990230microtubule associated protein 2 [Mus musculus]−4.32912958028CB11569804g629660NF-E1 [Gallus gallus]−4.4292233811.81569804g6395940GATA-2 transcription factor {3' flanking region,−4.4exon 6 }[Homo sapiens].2931270302CB11486358g2148620beta-tubulin [Xenopus laevis]−4.4294067163CB11384823g299794.00E−43Cks1 protein homologue [Homo sapiens]−4.4295002387CB12781405g3870051.00E−139proliferating cell nuclear antigen (PCNA) [Homo sapiens]−4.42962798854CB14385292g52625840hypothetical protein [Homo sapiens]−4.42971292280CB13496395g41643810nicotinic acetylcholine receptor alpha-3 subunit [Homo sapiens]−4.5298979248.23496395Incyte Unique−4.5299236240.31850531g43251801.00E−109tetraspan NET-6 [Homo sapiens]−4.5300234427.41616315g15076720GS3955 [Homo sapiens]−4.6301234427.71616315g15076729.00E−60GS3955 [Homo sapiens]−4.6302411205.16160410g28655200protein regulating cytokinesis 1 (PRC1) mRNA, complete cds−4.6[Homo sapiens]303411205.5160410g28655210protein regulating cytokinesis 1; PRC1 [Homo sapiens]−4.6304238854.231369473g7707424100E−117syntaxin 18 [Homo sapiens]−4.6305405008.1726201g59267034.00E−17Homo sapiens genomic DNA, chromosome 6p21.3, HLA Class I−4.6region, section 15/20.306372981.91576329g39012723.00E−56ZW10 interactor Zwint [Homo sapiens]−4.7307345125.8180439g1904260protein phosphatase-2A subunit-beta [Homo sapiens]−4.8308345125.17180439g17773733.00E−45B-regulatory subunit of protein phosphatase 2A [Rattus norvegicus]−4.83091723834CB11723834g4347530KIAA0030 [Homo sapiens]−4.9310407588.21640108g10350151.00E−112H. sapiens CpG island DNA genomic Mse1 fragment,−5clone 71a7, reverse read cpg71a7.rt1a3111970111CB11970111g2860130KIAA0008 [Homo sapiens]−5.1312058208CB1467621g8822230triadin [Homo sapiens]−5.2313333461.24003342g5597150KIAA0074 [Homo sapiens]−5.2314002940CB1161207g34022930aurora and IPL1-like midbody-associated protein kinase-1−5.4[Homo sapiens]315365153CB12375329g3395601.00E−178bone morphogenetic protein 5 [Homo sapiens]−5.6316034181CB11316528g1902670poly(ADP-ribose) polymerase [Homo sapiens]−5.7317264633.201709017g43780222.00E−84putative WHSC1 protein [Homo sapiens]−5.8318264633.191709017g66838080MMSET type I [Homo sapiens]−583191760566CB12657680g19073932.00E−79proneurotensin/proneuromedin N [Homo sapiens]−5.83203296553CB11739904g609535066 kDa neurofilament protein NF-66 [Mus musculus]−5.8321199471.22414624g15755341.00E−112Mad2 [Homo sapiens]−5.83221558165CB11403041g6875902.00E−36transmembrane protein [Homo sapiens]−5.9323988665.62219234g28272031.00E−114general transcription factor 2-I [Homo sapiens]−6324988665.102219234g28271804.00E−19general transcription factor 2-I; alternative splice−6product [Homo sapiens]325334634.13230940g22245770KIAA0318 [Homo sapiens]−63262823239CB1940823g2074090tyrosine hydroxylase (EC 1 14.16.2) [Rattus norvegicus]−6327021413CB11629861g14884131.00E−13N8 gene product = D52 homolog/leucine zipper protein−6.1[Homo sapiens]328637182CB13771476g2936890lamin B [Mus musculus]−6.13291297347CB11813133g4371023.00E−88HMG-1 [Mus musculus]−6.2330149914.152446238g5050981.00E−113KIAA0069 [Homo sapiens]−6.2331418689CB11646294g510530GATA-3 factor [Mus musculus]−6.33322232180CB1039817g2201360thymidylate synthase [Homo sapiens]−6.4333092267CB11932189g16990460Delta1 [Rattus norvegicus]−6.9334227432.21617878g36413000potassium channel [Rattus norvegicus]−7.1335227432.22617878g28014520potassium channel; KvEBN1 [Homo sapiens]−7.1336253570.301516301g17788400INS-1 winged helix [Rattus norvegicus]−7.3337253570.321516301g18422550hepatocyte nuclear factor-3/fork head homolog 11B−7.3[Homo sapiens]3383332616CB11502188g12444082.00E−17neuronatin alpha [Homo sapiens]−7.43391832346CB11721744g3399480tropomodulin [Homo sapiens]−7.4340221500.11672676g21306320synaptotagmin XI [Rattus norvegicus]−7.53411794861CB11515980g5572720HYL tyrosine kinase [Homo sapiens]−7.6342202239.13812392g2007680ribonucleotide reductase subunit M2 [Mus musculus]−7.63434181211CB1661492g13026580neural cell adhesion molecule L1 [Homo sapiens]−7.7344331051.4661492g3478073.00E−96cell adhesion molecule L1 [Homo sapiens]−7.73451454418CB11525795g298391.00E−172CDC2 polypeptide (CDC2) (AA 1-297) [Homo sapiens]−7.7346242309.61403636g8821470GRMP-62 [Gallus gallus]−7.8347232888.4129009g26684140topoisomersae II [Sus scrofa]−7.8348978190.83856893g9762350kinesin family protein KIF1a [Mus musculus]−7.83492700132CB12470485g11775280Ki-67 [Mus musculus]−8350343934.11267860g36416710doublecortin [Mus musculus]−8.33513145862CB13176609g17632590collapsin response mediator 1 [Mus musculus]−8.33521292191CB12821341g58345666.00E−65chromogranin B (secretogranin 1, SCG1) [Homo sapiens]−8.3353988660.321921393g514422.00E−09putative [Mus musculus]−9.23542522352CB1986752g25068360DNA replication licensing factor MCM7 (CDC47 homolog)−9.9[Homo sapiens]355244622.11412749g48367237.00E−90HMP19 protein [Homo sapiens]−10.63561555752CB13596853g31928790MAD3-like protein kinase [Homo sapiens]−10.73572324155CB11730052g2921661.00E−15469 kD autoanuigen [Homo sapiens]−11.23581100140.72916753g1842363.00E−83high mobility group2 protein [Homo sapiens]−13.83591100140.122916753g1842361.00E−98high mobility group 2 protein [Homo sapiens]−13.83603393396CB1494905g630990B-myb [Gallus gallus]−13.8361026662.3485111g63191780LEK1 [Mus musculus]−19.23621315515CB12821036g3380510secretogranin II [Homo sapiens]−19.4363406387.12373263g56894391.00E−161KIAA1051 protein [Homo sapiens]−19.83641610121CB12820985g1815210aromatic amino acid (dopa) decarboxylase [Homo sapiens]−21.1365330839.12811651g3869831.00E−164N-myc [Homo sapiens]−37.9


[0199]

2









TABLE 2








SEQ ID NO
Template ID
Clone ID
Start
Stop



















1
1497123CB1
1497123
300
1261


2
2985802CB1
3553729
6501
7088


3
475532.4
2859033
1
142


4
3138290CB1
1870965
34
5098


5
474310.40
1672744
1418
3957


6
410580.16
1445767
643
2168


7
337518.25
1674454
1234
3035


8
1303785CB1
79576
1100
1335


9
1044033.4
1514989
326
1982


10
1000222.31
690313
969
1529


11
403873.4
2329216
624
1460


12
1383105.12
4049957
136
1158


13
1383354.13
1572533
317
1260


14
697785CB1
2495131
21
332


15
420115CB1
1904751
1420
1958


16
1101453.2
2949427
72
682


17
1399366.20
2055534
4836
5848


18
3072333CB1
1447903
427
2440


19
12706811
1804548
1382
2242


20
1505038CB1
1987358
1199
3647


21
1035602.5
1854220
356
804


22
1330167.3
1001730
108
340


23
1003386CB1
1664320
547
1101


24
1097334.1
2483605
132
619


25
959142CB1
2804667
2455
5356


26
1359783CB1
1798209
736
2873


27
063646CB1
557012
544
1852


28
1519595CB1
2056395
938
2657


29
2054176CB1
3142736
660
2545


30
1312325CB1
1319608
2199
2722


31
022404.25
1319608
3805
4401


32
1787335CB1
1958902
726
1992


33
1193648.7
1851696
268
858


34
1193648.1
1851696
6053
6167


35
1867861CB1
1448051
8022
9703


36
5511889CB1
1650238
89
2294


37
3094768CB1
2902903
160
778


38
1256895CB1
1720056
721
2419


39
2019981CB1
1733490
2424
3921


40
2708240CB1
3084122
1730
3549


41
1092427.1
1313183
3747
5208


42
351841.7
1852047
2927
3756


43
022221.43
1314882
8395
10551


44
2190217CB1
78783
207
370


45
410910.3
2518178
2047
4796


46
1966280CB1
1700077
930
1514


47
430669.39
1572555
110
495


48
430669.23
1572555
1213
1596


49
1870753CB1
782235
4047
4691


50
3173735CB1
521139
322
2050


51
1330185.14
2868138
738
1146


52
2314132CB1
3118643
1118
2410


53
2508205CB1
2057296
480
2127


54
3326672CB1
1995380
27
565


55
234202.34
1995380
372
865


56
078242CB1
1319020
1743
1880


57
220943.20
417451
2272
2861


58
1383320.13
1558081
1275
3419


59
3526170CB1
2242648
533
1276


60
184081.24
27775
188
424


61
1821331CB1
1394401
525
2809


62
3660006CB1
1720114
2517
4383


63
089172.13
3693273
1727
4633


64
3084563CB1
2852042
87
1804


65
241227.17
1402715
515
1496


66
348151.2
1618422
3955
6433


67
1720808CB1
2606307
668
3615


68
998552.6
459651
663
1847


69
040652.35
2508261
541
981


70
040652.36
2508261
2420
2860


71
082155CB1
522294
542
1439


72
190144CB1
1668794
26
812


73
234537.3
1718651
3060
3637


74
1088425.1
162769
25
1178


75
254547.1
3134070
241
1028


76
2676170CB1
3820761
627
1978


77
1092181.1
2105963
37
514


78
471362.33
1720149
443
926


79
471362.27
1720149
319
771


80
1162416.1
2102320
1
157


81
252151.12
1599344
1054
1635


82
252151.7
1599344
1
579


83
358892.1
2057260
3649
3810


84
1296867CB1
3506985
209
793


85
337518.7
3506985
285
2721


86
1344279CB1
2771046
1591
3649


87
2731776CB1
2057601
1553
2381


88
1090035.1
1994472
40
450


89
1089929.9
2683564
646
778


90
2723092CB1
2633207
793
1058


91
2174489CB1
2173208
2599
3196


92
1253978CB1
1867652
606
2740


93
2274011CB1
1909488
606
1154


94
3119737CB1
2733928
183
414


95
1384695.102
1514318
2903
3243


96
257332CB1
1402228
1435
3048


97
2972880CB1
1636171
1163
1832


98
550425CB1
550425
482
1771


99
014284CB1
1822716
696
1862


100
1091854.7
1708528
4484
8482


101
138709.5
2844989
793
2272


102
375954.1
1404153
1066
3654


103
1262781CB1
3511216
947
1540


104
282761.16
3511216
2458
2924


105
3090708CB1
1283532
849
1996


106
230062.4
1711206
4892
6599


107
483043CB1
1854277
294
756


108
348205.9
1854277
564
977


109
1256295.18
1702350
828
1323


110
875668CB1
1418741
302
747


111
1180189.1
2664388
425
958


112
3109992CB1
2483173
5
1537


113
1250434CB1
1711151
2302
2787


114
1327838.1
3721987
39
327


115
2021477CB1
2190284
139
873


116
235171.20
1940994
3600
3851


117
149832CB1
604856
466
1002


118
1759127CB1
1759127
2503
3425


119
2048551CB1
2048551
1
558


120
3282941CB1
155904
993
1501


121
2733135CB1
2806166
523
1903


122
2176269CB1
1405940
790
2462


123
1218607CB1
477045
300
889


124
1553795CB1
1906574
2283
2722


125
238538.22
1906574
1044
1569


126
246546.9
2936505
358
860


127
234223.14
1672442
4807
6314


128
2054053CB1
2054053
332
869


129
1613766CB1
1640161
1176
1567


130
233454.3
1636639
930
1580


131
347699.13
1358285
2709
3065


132
3531583CB1
1358285
2458
3191


133
407096.14
630625
2268
3868


134
482411.26
2304121
29
519


135
482411.25
2304121
1168
1318


136
1258943CB1
434771
1739
3350


137
1327030.1
450574
353
1225


138
025595.22
1962971
4003
5656


139
995174.1
1975129
568
2327


140
1709732CB1
269456
1219
3491


141
1040610.4
692827
557
1390


142
055498.6
1865767
3980
4262


143
181172CB1
2503037
8
1168


144
2705515CB1
1846209
677
2204


145
480228.3
1997250
555
1014


146
360929.39
63038
85
296


147
1989087CB1
1603057
651
1324


148
995068.16
1904994
462
925


149
1217216.1
1976279
12140
12702


150
474426.5
1281473
306
1426


151
350521.22
2078364
1076
1891


152
1075592.6
1686585
3257
4529


153
1485867CB1
1959565
1635
2290


154
2515360CB1
1887959
4110
4667


155
3290944CB1
3598222
2388
3877


156
441206.15
2849603
967
2806


157
1712327CB1
1712327
1675
2673


158
1393778CB1
2056987
5339
5789


159
480127.44
2056987
1037
1246


160
1870941CB1
1870941
686
2204


161
2495110CB1
3176845
568
815


162
034711.3
3425195
2588
3084


163
251776.14
418731
2757
3391


164
239511.5
1821971
3967
5570


165
989878.1
1997703
2259
3042


166
1558664CB1
2018222
354
799


167
3602501CB1
3602501
1141
1784


168
5549580CB1
1736926
1057
2249


169
2687977CB1
1526282
23
2808


170
3168062CB1
1662688
520
1064


171
245367.2
3215205
452
2440


172
470587CB1
3940755
1735
2236


173
1631074CB1
64286
1477
1652


174
347829.12
185448
492
2202


175
347699.11
2058242
3889
4665


176
1251672.1
1453450
6281
7539


177
1291022CB1
1291022
1115
2019


178
237405.19
2380381
273
1420


179
2685676CB1
2513883
465
882


180
010672CB1
549196
119
635


181
234630.58
549196
213
788


182
3325955
3249851
160
733


183
332595.8
3249851
4129
4680


184
335086.1
3602403
3018
3387


185
1342493CB1
1453748
1684
2248


186
232691.20
2505425
554
1692


187
238814.2
1417211
1660
4002


188
201571.1
959745
278
1827


189
199882.5
1449824
4198
5529


190
237487.22
2380042
250
443


191
237487.21
2380042
595
756


192
305557CB1
29564
55
257


193
1378745CB1
147184
979
1422


194
1818836CB1
1818836
34
2284


195
1379463
690994
5224
6258


196
2110909CB1
2825369
938
2140


197
200578.1
1397926
1163
2312


198
259592CB1
197207
443
751


199
5584521CB1
1965863
131
653


200
399428.7
1491445
1662
2204


201
1175094
3012290
72
1586


202
3255458CB1
1597330
123
615


203
1430889CB1
1856520
236
669


204
4450486
1856520
497
923


205
4946593CB1
2852818
1624
2578


206
350605.45
4114209
1448
1913


207
1413644CB1
1413644
782
2021


208
9840092
1446475
68
808


209
627662CB1
1631511
1128
2113


210
1382932.11
2175008
4209
4703


211
2721850CB1
1624024
1162
2690


212
994902.1
2059691
530
1222


213
442744.17
1610993
1331
1866


214
442744.21
1610993
1571
2138


215
1908920CB1
2134356
755
1192


216
399101.31
2134356
514
1144


217
183198CB1
924319
774
1255


218
1397781.7
1522716
1328
1966


219
899496.9
812141
1463
2552


220
2111330CB1
1975209
1034
1781


221
331591.1
2452650
50
429


222
337119.8
2488567
742
1319


223
245011.11
2232471
879
1228


224
1988468CB1
2232471
886
1876


225
331470.8
1457726
3107
3620


226
411388CB1
591358
486
842


227
253450.9
1347232
12543
14884


228
351209.16
3686211
1030
2447


229
2124320CB1
2204916
3
2226


230
903876.1
548019
2801
5026


231
1238339CB1
2108793
179
677


232
245310.36
2108793
900
1367


233
2696735CB1
2696735
72
1519


234
338036.2
1449054
661
1416


235
236484.15
1922533
3229
4192


236
232719.2
537580
2541
3462


237
462249.1
1830083
2469
3616


238
1187408.1
30672
1252
1462


239
627856CB1
1559756
311
829


240
553078CB1
1985104
731
1832


241
048612.15
1975268
2605
2853


242
048612.12
1975268
1114
1662


243
1099779.1
1612306
1043
1732


244
1520855CB1
179929
4384
6269


245
1179282.1
2870970
730
1328


246
2770449CB1
1658320
843
1776


247
1430336CB1
30291
650
805


248
903105.6
544213
4275
4675


249
1327417.14
2211625
1
435


250
1327417.10
2211625
405
995


251
230712.24
2814551
407
940


252
982520.1
2986240
18
3113


253
311807CB1
821141
641
1853


254
1479370CB1
1626460
2350
3565


255
2993696CB1
2884613
13
2488


256
4004223CB1
1810945
425
955


257
453835.19
1723035
4103
5200


258
391741.16
1634279
1823
2346


259
391741.64
1634279
2996
3441


260
138295826
3876715
591
902


261
232567.4
1577614
511
1140


262
1720770CB1
2189762
323
811


263
253987.19
700559
493
1406


264
2047630CB1
1381654
815
1897


265
238203.11
999864
4229
5092


266
899410.5
1724967
4019
4432


267
4743113
2736056
3931
6660


268
2169835CB1
1003486
1792
2109


269
290021.11
1003486
2586
3038


270
267324CB1
2132217
1179
1454


271
2119372CB1
1889060
1770
3137


272
2818482CB1
2668334
404
1219


273
1330231.11
2594308
333
1159


274
1330117.5
692201
649
1299


275
233402.3
2056290
5739
6369


276
1622313CB1
1901061
1640
2538


277
2939887CB1
1375115
2059
2667


278
1804120CB1
1901095
2380
2963


279
245485.12
1901095
630
1083


280
1285395CB1
15834
1238
1533


281
036391.3
399035
1109
1635


282
036391.13
399035
2283
2683


283
474435.16
1610523
1326
2035


284
2495292CB1
2495292
1974
3637


285
251651.4
1645766
881
1434


286
5408483CB1
3493061
489
3217


287
347876.6
103669
290
2962


288
1289007CB1
1986737
942
1758


289
233301.18
814216
2032
2585


290
2157771CB1
2825656
5088
5612


291
2958028CB1
1569804
1259
1854


292
233811.8
1569804
316
734


293
1270302CB1
1486358
1376
2197


294
067163CB1
1384823
66
639


295
002387CB1
2781405
884
1288


296
2798854CB1
4385292
1174
3091


297
1292280CB1
3496395
1271
1842


298
979248.2
3496395
1
192


299
236240.3
1850531
472
1929


300
234427.4
1616315
911
1423


301
234427.7
1616315
1
625


302
411205.16
160410
195
679


303
411205.5
160410
1903
3093


304
238854.23
1369473
913
1296


305
405008.1
726201
69
488


306
372981.9
1576329
62
417


307
345125.8
180439
552
1110


308
345125.17
180439
270
840


309
1723834CB1
1723834
2901
3240


310
407588.2
1640108
1458
1771


311
1970111CB1
1970111
1059
2805


312
058208CB1
467621
609
1459


313
333461.2
4003342
1538
2064


314
002940CB1
161207
86
1222


315
365153CB1
2375329
509
1794


316
034181CB1
1316528
1946
3633


317
264633.20
1709017
1
562


318
264633.19
1709017
2523
3069


319
1760566CB1
2657680
536
798


320
3296553CB1
1739904
2395
2868


321
199471.2
2414624
125
1463


322
1558165CB1
1403041
431
1842


323
988665.6
2219234
878
1327


324
988665.10
2219234
276
797


325
334634.1
3230940
3713
5552


326
2823239CB1
940823
1174
1778


327
021413CB1
1629861
210
1669


328
637182CB1
3771476
1110
1582


329
1297347CB1
1813133
275
1067


330
149914.15
2446238
412
2382


331
418689CB1
1646294
1618
2260


332
2232180CB1
39817
798
963


333
092267CB1
1932189
805
1262


334
227432.21
617878
447
1001


335
227432.22
617878
843
1376


336
253570.30
1516301
2588
3043


337
253570.32
1516301
2747
3519


338
3332616CB1
1502188
15
547


339
1832346CB1
1721744
1109
2734


340
221500.1
1672676
3901
5215


341
1794861CB1
1515980
779
1949


342
202239.1
3812392
0
1680


343
4181211CB1
661492
2001
2372


344
331051.4
661492
1175
1676


345
1454418CB1
1525795
336
1776


346
242309.6
1403636
3247
3729


347
2328884
129009
3843
5647


348
978190.8
3856893
10
991


349
2700132CB1
2470485
412
985


350
343934.1
1267860
8048
9390


351
3145862CB1
3176609
1646
2820


352
1292191CB1
2821341
9
2541


353
988660.32
1921393
284
703


354
2522352CB1
986752
443
2551


355
244622.1
1412749
1845
2383


356
1555752CB1
3596853
27
3642


357
2324155CB1
1730052
112
1683


358
1100140.7
2916753
1
542


359
1100140.12
2916753
575
1152


360
3393396CB1
494905
166
2601


361
026662.3
485111
7806
10241


362
1315515CB1
2821036
31
2342


363
406387.1
2373263
5566
6690


364
1610121CB1
2820985
517
1895


365
330839.1
2811651
1054
2499










[0200]


Claims
  • 1. A combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs:1-365 that are differentially expressed in MYCN activated cells and the complements of the nucleic acid sequences of SEQ ID NOs:1-365.
  • 2. The combination of claim 1, wherein the MYCN activated cells are neuroblastoma.
  • 3. A method for using a combination comprising a plurality of cDNAs to detect expression of one or more nucleic acids in a sample, the method comprising: a) hybridizing the combination of claim 1 with nucleic acids of the sample, thereby forming one or more hybridization complexes; and b) detecting complex formation wherein complex formation indicates expression of at least one nucleic acid in the sample.
  • 4. The method of claim 3, wherein the combination is immobilized on a substrate.
  • 5. The method of claim 3, wherein the nucleic acids of the sample are amplified prior to hybridization.
  • 6. The method of claim 3, wherein the sample is from a subject with neuroblastoma and comparison with a standard defines the stage of that disorder.
  • 7. A method of using a combination comprising a plurality of cDNAs to screen a plurality of molecules or compounds to identify a ligand which specifically binds at least one cDNA of the combination, the method comprising: a) contacting the combination of claim 1 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between at least one cDNA and at least one molecule or compound, thereby identifying a ligand that specifically binds to a cDNA of the combination.
  • 8. The method of claim 7 wherein the plurality of molecules or compounds are selected from DNA molecules, RNA molecules, peptide nucleic acid molecules, mimetics, peptides, transcription factors, repressors, and regulatory proteins.
  • 9. An isolated cDNA selected from the SEQ ID NOs:1-365.
  • 10. A vector containing the cDNA of claim 9.
  • 11. A host cell containing the vector of claim 10.
  • 12. A method for producing a protein, the method comprising the steps of: a) culturing the host cell of claim 11 under conditions for expression of protein; and b) recovering the protein from the host cell culture.
  • 13. A method for using a cDNA to detect expression of a complementary nucleic acid in a sample, the method comprising: a) hybridizing the cDNA of claim 9 with the sample, thereby forming a hybridization complex; and b) detecting complex formation wherein complex formation indicates expression of a complementary nucleic acid in the sample.
  • 14. A method of using a cDNA to screen a plurality of molecules or compounds to identify a molecule or compound which specifically binds the cDNA, the method comprising: a) contacting the cDNA of claim 9 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between the cDNA and at least one molecule or compound, thereby identifying a molecule or compound that specifically binds the cDNA.
  • 15. A protein produced by the method of claim 12.
  • 16. A method for using a protein to screen a plurality of molecules or compounds to identify at least one ligand which specifically binds the protein, the method comprising: a) combining the protein of claim 15 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.
  • 17. The method of claim 16 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.
  • 18. An antibody which specifically binds the protein of claim 15.
  • 19. A method of using a protein to produce and purify an antibody, the method comprising: a) immunizing an animal with the protein of claim 15 under conditions to elicit an antibody response; b) isolating animal antibodies; c) contacting the protein with the isolated antibodies under conditions to allow specific binding; d) recovering the bound protein; and e) separating the protein from the antibody, thereby obtaining purified antibody.
  • 20. A method of using an antibody to detect a protein in a sample, the method comprising: a) contacting the antibody of claim 18 with a sample under condition for the formation of an antibody:protein complex, and b) detecting the antibody:protein complex wherein complex formation indicates the presence of the protein in the sample.
Parent Case Info

[0001] This application claims benefit of provisional application Serial No. 60/270,784, filed Feb. 23, 2001.

Provisional Applications (1)
Number Date Country
60270784 Feb 2001 US