WO2002068621A2 - Rax-related protein - Google Patents

Rax-related protein Download PDF

Info

Publication number
WO2002068621A2
WO2002068621A2 PCT/US2002/006179 US0206179W WO02068621A2 WO 2002068621 A2 WO2002068621 A2 WO 2002068621A2 US 0206179 W US0206179 W US 0206179W WO 02068621 A2 WO02068621 A2 WO 02068621A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
cdna
antibody
cancer
molecules
Prior art date
Application number
PCT/US2002/006179
Other languages
French (fr)
Other versions
WO2002068621A3 (en
Inventor
Preeti G. Lal
Henry Yue
Original Assignee
Incyte Genomics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Genomics, Inc. filed Critical Incyte Genomics, Inc.
Priority to AU2002248517A priority Critical patent/AU2002248517A1/en
Publication of WO2002068621A2 publication Critical patent/WO2002068621A2/en
Publication of WO2002068621A3 publication Critical patent/WO2002068621A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity

Definitions

  • This invention relates to cDNA which encodes a RAX-related protein and to the use of the cDNA and the encoded protein in the diagnosis and treatment of cell proliferative disorders including cancer and Crohn's disease.
  • Double-stranded RNA-dependent protein kinase mediates the anticancer and antiviral effects of interferon (Williams (1999) Oncogene 18:6112-6120).
  • interferon induces expression of PKR, a serine/threonine protein kinase that blocks viral replication by inhibiting protein synthesis and cell growth, and inducing apoptosis.
  • PKR is activated by binding to viral double-stranded RNA, which promotes autophosphorylation.
  • PKR inhibits protein synthesis at the stage of translation initiation by phosphorylating the ⁇ subunit of eukaryotic initiation factor-2 (eEF- 2 ).
  • RNA-binding is mediated by two double-stranded RNA-binding domains (DRBDs) at the N- terminus of PKR.
  • the DRBDs are also required for interaction of PKR with ribosomes and protein regulatory factors that contain DRBDs (Zhu et al. (1997) J Biol Chem 272:14434-14441; Patel and Sen (1998) EMBO J 17:4379-4390).
  • DRBDs RNA-binding domains
  • PKR has a role in promoting apoptosis.
  • Activation of PKR induces expression of members of the tumor necrosis factor receptor family, including Fas and Bax (Siddharth et al. (1998) EMBO J 17:6888-6902).
  • RAX PPR-associated protein X
  • RAX is a 35 kDa protein of 313 amino acids that has three DRBDs, which mediate interactions with double-stranded RNA and PKR.
  • RAX is phosphorylated at a serine residue by an unknown stress-activated protein kinase.
  • RAX activates PKR in the absence of dsRNA and may function in regulating protein translation, cell-proliferation, and apoptosis in non-virally infected cells.
  • PACT PVR activating protein
  • compositions which are useful in the diagnosis and treatment of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the invention is based on the discovery of a cDNA encoding RAX-related protein (RRP) which is useful in the diagnosis and treatment of cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • RRP RAX-related protein
  • the invention provides an isolated cDNA comprising a nucleic acid sequence encoding a protein having the amino acid sequence of SEQ ID NO:l.
  • the invention also provides an isolated cDNA or the complement thereof selected from the group consisting of a nucleic acid sequence of SEQ ID NO:2, a fragment of SEQ ID NO:2 selected from SEQ ID NOs:3-12, and a variant of SEQ ID NO:2 selected from SEQ ID NOs:13-15.
  • the invention additionally provides a composition, a substrate, and a probe comprising the cDNA, or the complement of the cDNA, encoding RRP.
  • the invention further provides a vector containing the cDNA, a host cell containing the vector and a method for using the cDNA to make RRP.
  • the invention still further provides a transgenic cell line or organism comprising the vector containing the cDNA encoding RRP.
  • the invention additionally provides a fragment, a variant, or the complement of the cDNA selected from the group consisting of
  • the invention provides a substrate containing at least one of these fragments or variants or the complements thereof.
  • the invention provides a probe comprising a cDNA or the complement thereof which can be used in methods of detection, screening, and purification.
  • the probe is a single-stranded complementary RNA or DNA molecule.
  • the invention provides a method for using a cDNA to detect the differential expression of a nucleic acid in a sample comprising hybridizing a probe to the nucleic acids, thereby forming hybridization complexes and comparing hybridization complex formation with a standard, wherein the comparison indicates the differential expression of the cDNA in the sample.
  • the method of detection further comprises amplifying the nucleic acids of the sample prior to hybridization.
  • the method showing differential expression of the cDNA is used to diagnose cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the cDNA or a fragment or a variant or the complements thereof may comprise an element on an array.
  • the invention additionally provides a method for using a cDNA or a fragment or a variant or the complements thereof to screen a library or plurality of molecules or compounds to identify at least one ligand which specifically binds the cDNA, the method comprising combining the cDNA with the molecules or compounds under conditions allowing specific binding, and detecting specific binding to the cDNA, thereby identifying a ligand which specifically binds the cDNA.
  • the molecules or compounds are selected from aptamers, DNA molecules, RNA molecules, peptide nucleic acids, artificial chromosome constructions, peptides, transcription factors, repressors, and regulatory molecules.
  • the invention provides a purified protein or a portion thereof selected from the group consisting of an amino acid sequence of SEQ ID NO:l, a variant having at least 89% identity to the amino acid sequence of SEQ ED NO:l, an antigenic epitope of SEQ ID NO:l, and a biologically active portion of SEQ ED NO:l.
  • the invention also provides a composition comprising the purified protein in conjunction with a pharmaceutical carrier.
  • the invention further provides a method of using the RRP to treat a subject with cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising administering to a patient in need of such treatment the composition containing the purified protein.
  • the invention still further provides a method for using a protein to screen a library or a plurality of molecules or compounds to identify at least one ligand, the method comprising combining the protein with the molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds the protein.
  • the molecules or compounds are selected from DNA molecules, RNA molecules, peptide nucleic acids, peptides, proteins, mimetics, agonists, antagonists, antibodies, immunoglobulins, inhibitors, and drugs.
  • the ligand is used to treat a subject with cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the invention provides a method of using a protein to screen a subject sample for antibodies which specifically bind the protein comprising isolating antibodies from the subject sample, contacting the isolated antibodies with the protein under conditions that allow specific binding, dissociating the antibody from the bound-protein, and comparing the quantity of antibody with known standards, wherein the presence or quantity of antibody is diagnostic of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • cancer particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the invention also provides a method of using a protein to prepare and purify antibodies comprising immunizing a animal with the protein under conditions to elicit an antibody response, isolating animal antibodies, attaching the protein to a substrate, contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein, dissociating the antibodies from the protein, thereby obtaining purified antibodies.
  • the invention provides a purified antibody which binds specifically to a protein which is expressed in cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the invention also provides a method of using an antibody to diagnose cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising combining the antibody comparing the quantity of bound antibody to known standards, thereby establishing the presence of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • cancer particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the invention further provides a method of using an antibody to treat cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising administering to a patient in need of such treatment a pharmaceutical composition comprising the purified antibody.
  • cancer particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease
  • the invention provides a method for inserting a heterologous marker gene into the genomic DNA of a mammal to disrupt the expression of the endogenous polynucleotide.
  • the invention also provides a method for using a cDNA to produce a mammalian model system, the method comprising constructing a vector containing the cDNA selected from SEQ ED NOs:2-15, tiansforming the vector into an embryonic stem cell, selecting a transformed embryonic stem, microinjecting the transformed embryonic stem cell into a mammalian blastocyst, thereby fo ⁇ ning a chimeric blastocyst, transferring the chimeric blastocyst into a pseudopregnant dam, wherein the dam gives birth to a chimeric offspring containing the cDNA in its germ line, and breeding the chimeric mammal to produce a homozygous, mammalian model system.
  • Figures 2A and 2B demonstrate the conserved chemical and structural similarities among the sequences and motifs of RRP (2754573; SEQ D NO:l) and human RAX (g4927416; SEQ ED NO: 16). The alignment was produced using the MEGALIGN program of LASERGENE software
  • Tables 1 and 2 show the northern analysis for RRP produced using the LEFESEQ Gold database (Incyte Genomics, Palo Alto CA).
  • the first column presents the tissue categories; the second column, the total number of clones in the tissue category; the third column, the ratio of the number of libraries in which at least one transcript was found to the total number of libraries; the fourth column, absolute clone abundance of the transcript; and the fifth column, percent abundance of the transcript.
  • Table 2 shows expression of RRP in various tissues from patients with cancer or Crohn's disease.
  • the first column lists the library name, the second column, the number of clones sequenced for that library; the third column, the description of the tissue from which the library was derived; the fourth column, the absolute abundance of the transcript; and the fifth column, the percent abundance of the transcript.
  • RRP refers to a purified protein obtained from any mammalian species, including bovine, canine, murine, ovine, porcine, rodent, simian, and preferably the human species, and from any source, whether natural, synthetic, semi-synthetic, or recombinant.
  • Array refers to an ordered arrangement of at least two cDNAs on a substrate. At least one of the cDNAs represents a control or standard, and the other, a cDNA of diagnostic or therapeutic interest. The arrangement of from about two to about 40,000 cDNAs on the substrate assures that the size and signal intensity of each labeled hybridization complex formed between each cDNA and at least one sample nucleic acid is individually distinguishable.
  • the "complement” of a cDNA of the Sequence Listing refers to a nucleic acid molecule which is completely complementary over its full length and which will hybridize to the cDNA or an mRNA under conditions of maximal stringency.
  • cDNA refers to an isolated polynucleotide, nucleic acid molecule, or any fragment or complement thereof. It may have originated recombinantly or synthetically, may be double-stranded or single-stranded, represents coding and noncoding 3' or 5' sequence, and generally lacks introns.
  • cDNA encoding a protein refers to a nucleotide 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) which provides identity within the conserved region (Altschul (1993) J Mol Evol 36: 290-300; Altschul et al. (1990) J Mol Biol 215:403-410).
  • BLAST Basic Local Alignment Search Tool
  • composition comprises the polynucleotide and a labeling moiety or a purified protein in conjunction with a pharmaceutical carrier.
  • “Derivative” refers to a cDNA or a protein that has been subjected to a chemical modification. Derivatization of a cDNA can involve substitution of a nontraditional base such as queosine or of an analog such as hypoxanthine. These substitutions are well known in the art.
  • Derivatization of a protein involves the replacement of a hydrogen by an acetyl, acyl, alkyl, amino, formyl, or morpholino group.
  • Derivative molecules retain the biological activities of the naturally occurring molecules but may confer advantages such as longer lifespan or enhanced activity.
  • “Differential expression” refers to an increased, upregulated or present, or decreased, downregulated or absent, gene expression as detected by presence, absence or at least two-fold changes in the amount of transcribed messenger RNA or translated protein in a sample.
  • disorder refers to conditions, diseases or syndromes in which the cDNAs and RRP are differentially expressed.
  • a disorder includes cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • Fragments refers to a chain of consecutive nucleotides from about 50 to about 4000 base pairs in length. Fragments may be used in PCR or hybridization technologies to identify related nucleic acid molecules and in binding assays to screen for a ligand. Such ligands are useful as therapeutics to regulate replication, transcription or translation.
  • 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'.
  • Hybridization conditions, degree of complementarity and the use of nucleotide analogs affect the efficiency and stringency of hybridization reactions.
  • "Labeling moiety” refers to any visible or radioactive label than can be attached to or incorporated into a cDNA or protein.
  • Visible labels include but are not limited to anthocyanins, green fluorescent protein (GFP), ⁇ glucuronidase, luciferase, Cy3 and Cy5, and the like.
  • Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like.
  • Ligand refers to any agent, molecule, or compound which will bind specifically to a polynucleotide or to an epitope of a protein. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic and/or organic substances including minerals, cofactors, nucleic acids, proteins, carbohydrates, fats, and lipids.
  • Oligomer 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. Substantially equivalent terms are amplimer, primer, and oligomer.
  • Portion refers to any part of a protein used for any purpose; but especially, to an epitope for the screening of ligands or for the production of antibodies.
  • 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.
  • 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 with reporter molecules for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.
  • 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 epitope of the protein identified using Kyte-Doolittle algorithms of the PROTEAN program (DNASTAR, Madison WI).
  • 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.
  • “Purified” refers to any molecule or compound that is separated from its natural environment and is from about 60% free to about 90% free from other components with which it is naturally associated.
  • 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; a tissue print; a fingerprint, buccal cells, skin, or hair; and the like.
  • 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 or the binding between an epitope of a protein and an agonist, antagonist, or antibody.
  • Similarity 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) or BLAST2 (Altschul et al. (1997) Nucleic Acids Res 25:3389-3402).
  • BLAST2 maybe 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. Particularly in proteins, similarity is greater than identity in that conservative substitutions, for example, valine for leucine or isoleucine, are counted in calculating the reported percentage. Substitutions which are considered to be conservative are well known in the art.
  • Substrate refers to any rigid or semi-rigid support to which cDNAs or proteins are bound and includes membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, capillaries or other tubing, plates, polymers, and microparticles with a variety of surface forms including wells, trenches, pins, channels and pores.
  • “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 or its secondary, tertiary, or quaternary structure. THE INVENTION
  • the invention is based on the discovery of a cDNA which encodes RRP and on the use of the cDNA, or fragments thereof, and protein, or portions thereof, directly or as compositions in the characterization, diagnosis, and treatment of cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • Nucleic acids encoding the RRP of the present invention were first identified in Incyte Clone 2754573 from the THP-1 promonocyte cell cDNA library (THP1AZS08) using a computer search for nucleotide and/or amino acid sequence alignments.
  • SEQ ED NO:2 was derived from the following overlapping and/or extended nucleic acid sequences (SEQ ED NO:3-12): Incyte Clones 2754573H1
  • THP1AZS08 491644H1 (HNT2AGT01), 412307R1 (BRSTNOT01), 263630H1 (HNT2AGT01), 412307T6 (BRSTNOT01), 1253094H1 (LUNGFET03), 2280508H1 (PROSNON01), 2270603H1 (PROSNON01), 2375670H1 (ISLTNOTOl), and 3151587H1 (ADRENON04).
  • Table 1 shows expression of RRP across various tissue categories (also listed in Example VEEI).
  • Table 2 shows expression of RRP specifically in tissues from patients with cancer or Crohn's disease.
  • RRP is found in libraries associated with various cancers, in particular, an adipose tissue library (CONFTUPOl) from a patient with liposarcoma, a mesentery tissue library (CONUTUTOl) from a patient with metastasizing mixed-mullerian tumors, a spinal soft tissue library (CONNTUT04) from a patient with schwannoma, a gallbladder tissue library (GBLATUT01) from a patient with squamous cell carcinoma, and lung tissue libraries (LUNLTUT04, LUNGTUT09, and LUNGTUT08) from patients with lung cancer.
  • CONFTUPOl adipose tissue library
  • CONUTUTOl mesentery tissue library
  • GLATUT01 gallbladder tissue library
  • lung tissue libraries LUNLTUT04, LUNGTUT09, and LUNGTUT08
  • RRP shows overexpression in a brain tissue library (BRAFTUE03) from a patient with astrocytoma compared to a library (BRAENOT14) from matched (m) microscopically normal tissue from the same donor.
  • RRP is overexpressed in a lung tissue library (LUNGTUT03) from a patient with squamous cell carcinoma compared to a library (LUNGNOT15) from matched (m) microscopically normal tissue from the same donor.
  • RRP is notably overexpressed in a bladder tissue library (BLADTUT05) from a patient with transitional cell carcinoma compared to a library (BLADNOT06) from matched (m) microscopically normal tissue from the same donor.
  • RRP is underexpressed in prostate tissue libraries (PROETUP02, PROSTUT16, PROSTUT18) from patients with prostate cancer compared to libraries (PROETMPOl, PROSNOT28, and PROSTMT03) from matched (m) microscopically normal tissue from the same donors.
  • RRP is overexpressed in a colon tissue library (COLNCRT01) from a patient with Crohn's disease compared to a library (COLNNOT05) from matched (m) microscopically normal tissue from the same donor.
  • the transcript is therefore useful in diagnostic assays for cancers, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • a fragment of the cDNA from about nucleotide 60 to about nucleotide 120 is also useful in diagnostic assays.
  • the invention encompasses a polypeptide comprising the amino acid sequence of SEQ ED NO:l as shown in Figures 1A, IB, IC, ID, IE, and IF.
  • RRP is 352 amino acids in length and has two potential N-glycosylation sites at N141 and N294; one potential myristoylation site at G12; five potential casein kinase II phosphorylation sites at T110, S169, S206, S260, and T296; and eight potential protein kinase C phosphorylation sites at S8, T67, T106, T121, S122, T210, S215, and S256.
  • PFAM analysis indicates that the regions of RRP from P74 to L138, from P166 to F231, and from Y280 to L345 are similar to a double-stranded RNA binding motif.
  • BLOCKS analysis indicates that the regions of RRP from Gl 18 to L138 and from N165 to Y184 are similar to
  • Ribonuclease EQ family motifs Ribonuclease EQ family motifs; and the region of RRP from T13 to T24 is similar to an RNA 3'- terminal phosphate cyclase motif.
  • RRP has chemical and structural similarity with human RAX (g4927416; SEQ ID NO:16).
  • RRP and RAX share about 88% identity and three double-stranded RNA binding motifs.
  • RRP differs from the human RAX primarily by an additional N-terminal extension of about 40 amino acid residues.
  • Useful antigenic epitopes extend from about A68 to about G82, from about D150 to about G168, and from about R258 to about S271; and biologically active portions of RRP extend from about P74 to about L138, from about P166 to about F231, and from about Y280 to about L345.
  • An antibody which specifically binds RRP is useful in an diagnostic assay to identify cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease
  • Mammalian variants of the cDNA encoding RRP were identified using BLAST2 with default parameters and the ZOOSEQ databases (incyte Genomics). These preferred variants have from about 86% to about 92% identity as shown in the table below.
  • the first column shows the SEQ ED for the human cDNA (SEQ ED H ); the second column, the SEQ ED for the variant cDNAs (SEQ ED H ); the SEQ ED for the variant cDNAs (SEQ ED H ); the SEQ ED for the variant cDNAs (SEQ
  • the third column the clone number for the variant cDNAs (Clone ⁇ ); the fourth column, the library name; the fifth column, the alignment of the variant cDNA to the human cDNA (includes the alignment of different regions of the variant cDNA with different regions of the human cDNA in some cases); and the sixth column, the percent identity to the human cDNA.
  • cDNAs, SEQ ED NOS:13-15 are particularly useful for producing transgenic cell lines or organisms.
  • the cDNAs of SEQ ED NOs:2-15 may be used in hybridization, amplification, and screening technologies to identify and distinguish among SEQ ED NO:2 and related molecules in a sample.
  • the mammalian cDNAs may be used to produce transgenic cell lines or organisms which are model systems for human cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease and upon which the toxicity and efficacy of potential therapeutic treatments may be tested. Toxicology studies, clinical trials, and subject/patient treatment profiles may be performed and monitored using the cDNAs, proteins, antibodies and molecules and compounds identified using the cDNAs and proteins of the present invention.
  • rriRNA is isolated from mammalian cells and tissues using methods which are well known to those skilled in the art and used to prepare the cDNA libraries.
  • the Incyte cDNAs were isolated from mammalian cDNA libraries aprepared as described in the EXAMPLES.
  • the consensus sequences are chemically and/or electronically assembled from fragments including Incyte cDNAs and extension and/or shotgun sequences using computer programs such as PHRAP (P Green, University of Washington, Seattle WA), and AUTOASSEMBLER application (Applied Biosystems, Foster City CA). After verification of the 5' and 3' sequence, at least one representative cDNA which encodes RRP is designated a reagent. Sequencing
  • sequence preparation is automated with machines such as the MICROLAB 2200 system (Hamilton, Reno NV) and the DNA ENGINE thermal cycler (MJ Research, Watertown MA).
  • Machines commonly used for sequencing include the ABI PRISM 3700, 377 or 373 DNA sequencing systems (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (APB), and the like.
  • the sequences may be analyzed using a variety of algorithms well known in the art and described in Ausubel et al. (1997; Short Protocols in Molecular Biology, John Wiley & Sons, New York NY, unit 7.7) and in Meyers (1995; Molecular Biology and Biotechnology. Wiley VCH, New York NY, pp. 856-853).
  • Shotgun sequencing may also be used to complete the sequence of a particular cloned insert of interest. Shotgun strategy involves randomly breaking the original insert into segments of various sizes and cloning these fragments into vectors.
  • the fragments are sequenced and reassembled using overlapping ends until the entire sequence of the original insert is known.
  • Shotgun sequencing methods are well known in the art and use thermostable DNA polymerases, heat-labile DNA polymerases, and primers chosen from representative regions flanking the cDNAs of interest.
  • Incomplete assembled sequences are inspected for identity using various algorithms or programs such as CONSED (Gordon (1998) Genome Res 8:195-202) which are well known in the art.
  • Contaminating sequences, including vector or chimeric sequences, or deleted sequences can be removed or restored, respectively, organizing the incomplete assembled sequences into finished sequences.
  • sequences of the invention may be extended using various PCR-based methods known in the art.
  • the XL-PCR kit (Applied Biosystems), nested primers, and commercially available cDNA or genomic DNA libraries may be used to extend the nucleic acid sequence.
  • primers may be designed using commercially available software, such as
  • OLIGO primer analysis software (Molecular Biology Insights, Cascade CO) to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to a target molecule at temperatures from about 55C to about 68C.
  • genomic, rather than cDNA libraries it is preferable to use genomic, rather than cDNA libraries.
  • a probe may be designed or derived from unique regions such as the 5' regulatory region or from a nonconserved region (i.e., 5 ' or 3 ' of the nucleotides encoding the conserved catalytic domain of the protein) and used in protocols to identify naturally occurring molecules encoding the RRP, allelic variants, or related molecules.
  • the probe may be DNA or RNA, may be single-stranded, and should have at least 50% sequence identity to any of the nucleic acid sequences, SEQ ED NOs:2-15.
  • Hybridization probes may be produced using oligolabeling, nick translation, end-labeling, or PCR amplification in the presence of a reporter molecule.
  • a vector containing the cDNA or a fragment thereof may be used to produce an mRNA probe in vitro by addition of an RNA polymerase and labeled nucleotides. These procedures may be conducted using commercially available kits such as those provided by APB.
  • the stringency of hybridization is determined by G+C content of the probe, salt concentration, and temperature. In particular, stringency can be increased by reducing the concentration of salt or raising the hybridization temperature.
  • Hybridization can be performed at low stringency with buffers, such as 5xSSC with 1% sodium dodecyl sulfate (SDS) at 60C, which permits the formation of a hybridization complex between nucleic acid sequences that contain some mismatches. Subsequent washes are performed at higher stringency with buffers such as 0.2xSSC with 0.1% SDS at either 45C (medium stringency) or 68C (high stringency). At high stringency, hybridization complexes will remain stable only where the nucleic acids are completely complementary.
  • formamide can be added to the hybridization solution to reduce the temperature at which hybridization is performed, and background signals can 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 (supra) and
  • Arrays may be prepared and analyzed using methods well known in the art. Oligonucleotides or cDNAs may be used as hybridization probes or targets to monitor the expression level of large numbers of genes simultaneously or to identify genetic variants, mutations, and single nucleotide polymorphisms. Arrays may be used to determine gene function; to understand the genetic basis of a condition, disease, or disorder; to diagnose a condition, disease, or disorder; and to develop and monitor the activities of therapeutic agents. (See, e.g., Brennan et al. (1995) USPN 5,474,796; Schena et al. (1996) Proc Natl Acad Sci 93:10614-10619; Heller et al. (1997) Proc Natl Acad Sci 94:2150-2155; and Heller et al. (1997) USPN 5,605,662.)
  • Hybridization probes are also useful in mapping the naturally occurring genomic sequence.
  • the probes may be hybridized to a particular chromosome, a specific region of a chromosome, or an artificial chromosome construction.
  • Such constructions include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC), bacterial PI constructions, or the cDNAs of libraries made from single chromosomes.
  • Any one of a multitude of cDNAs encoding RRP may be cloned into a vector and used to express the protein, or portions thereof, in host cells.
  • the nucleic acid sequence can be engineered by such methods as DNA shuffling (USPN 5,830,721) and site-directed mutagenesis to create new restriction sites, alter glycosylation patterns, change codon preference to increase expression in a particular host, produce splice variants, extend half-life, and the like.
  • the expression vector may contain transcriptional and translational control elements (promoters, enhancers, specific initiation signals, and polyadenylated 3 ' sequence) from various sources which have been selected for their efficiency in a particular host.
  • the vector, cDNA, and regulatory elements are combined using in vitro recombinant DNA techniques, synthetic techniques, and/or in vivo genetic recombination techniques well known in the art and described in Sambrook (supra, ch. 4, 8, 16 and 17).
  • a variety of host systems may be transformed with an expression vector. These include, but are not limited to, bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems transformed with baculovirus expression vectors; plant cell systems transformed with expression vectors containing viral and/or bacterial elements, or animal cell systems (Ausubel supra, unit 16).
  • an adenovirus transcription/translation complex may be utilized in mammalian cells. After sequences are ligated into the El or E3 region of the viral genome, the infective virus is used to transform and express the protein in host cells.
  • the Rous sarcoma virus enhancer or SV40 or EBV-based vectors may also be used for high-level protein expression.
  • Routine cloning, subcloning, and propagation of nucleic acid sequences can be achieved using the multifunctional PBLUESCREPT vector (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Introduction of a nucleic acid sequence into the multiple cloning site of these vectors disrupts the lacZ gene and allows colorimetric screening for transformed bacteria. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence.
  • the vector can be stably transformed into cell lines along with a selectable or visible marker gene on the same or on a separate vector. After transformation, cells are allowed to grow for about 1 to 2 days in enriched media and then are transfe ⁇ ed to selective media. Selectable markers, antimetabolite, antibiotic, or herbicide resistance genes, confer resistance to the relevant selective agent and allow growth and recovery of cells which successfully express the introduced sequences. Resistant clones identified either by survival on selective media or by the expression of visible markers may be propagated using culture techniques. Visible markers are also used to estimate the amount of protein expressed by the introduced genes. Verification that the host cell contains the desired cDNA is based on DNA-DNA or DNA-RNA hybridizations or PCR amplification techniques.
  • the host cell may be chosen for its ability to modify a recombinant protein in a desired fashion. Such modifications include acetylation, carboxylation, glycosylation, phosphorylation, lipidation, acylation and the like. Post-translational processing which cleaves a "prepro" form may also be used to specify protein targeting, folding, and/or activity. Different host cells available from the ATCC (Manassas VA) which have specific cellular machinery and characteristic mechanisms for post-translational activities may be chosen to ensure the co ⁇ ect modification and processing of the recombinant protein.
  • ATCC Manassas VA
  • Heterologous moieties engineered into a vector for ease of purification include glutathione S- transferase (GST), 6xHis, FLAG, MYC, and the like.
  • GST and 6-His are purified using commercially available affinity matrices such as immobilized glutathione and metal-chelate resins, respectively.
  • FLAG and MYC are purified using commercially available monoclonal and polyclonal antibodies.
  • a sequence encoding a proteolytic cleavage site may be part of the vector located between the protein and the heterologous moiety. Methods for recombinant protein expression and purification are discussed in Ausubel (supra, unit 16) and are commercially available. Chemical Synthesis of Peptides
  • Proteins or portions thereof may be produced not only by recombinant methods, but also by using chemical methods well known in the art.
  • Solid phase peptide synthesis may be carried out in a batchwise or continuous flow process which sequentially adds ⁇ -amino- and side chain-protected amino acid residues to an insoluble polymeric support via a linker group.
  • a linker group such as memylamine-derivatized polyethylene glycol is attached to poly(styrene-co-divinylbenzene) to form the support resin.
  • the amino acid residues are N- ⁇ -protected by acid labile Boc (t-butyloxycarbonyl) or base-labile Fmoc (9-fluorenylmethoxycarbonyl).
  • the carboxyl group of the protected amino acid is coupled to the amine of the linker group to anchor the residue to the solid phase support resin.
  • Trifluoroacetic acid or piperidine are used to remove the protecting group in the case of Boc or Fmoc, respectively.
  • Each additional amino acid is added to the anchored residue using a coupling agent or pre-activated amino acid derivative, and the resin is washed.
  • the full length peptide is synthesized by sequential deprotection, coupling of derivitized amino acids, and washing with dichloromethane and/or N, N-dimethylformamide. The peptide is cleaved between the peptide carboxy terminus and the linker group to yield a peptide acid or amide.
  • a protein or portion thereof may be substantially purified by preparative high performance liquid chromatography and its composition confirmed by amino acid analysis or by sequencing (Creighton (1984) Proteins, Structures and Molecular Properties, WH Freeman, New York NY).
  • Various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with RRP or any portion thereof.
  • Adjuvants such as Freund's, mineral gels, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemacyanin (KLH), and dinitrophenol may be used to increase immunological response.
  • the ohgopeptide, peptide, or portion of protein used to induce antibodies should consist of at least about five amino acids, more preferably ten amino acids, which are identical to a portion of the natural protein. Oligopeptides may be fused with proteins such as KLH in order to produce antibodies to the chimeric molecule.
  • Monoclonal antibodies may be prepared using any technique which provides for the production of antibodies by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler et al. (1975) Nature 256:495-497; Kozbor et al. (1985) J. Immunol Methods 81:31-42; Cote et al. (1983) Proc Natl Acad Sci 80:2026-2030; and Cole et al.
  • antibody fragments which contain specific binding sites for epitopes of the protein may also be generated.
  • fragments include, but are not limited to, F(ab 2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e.g., Huse et al. (1989) Science 246:1275-1281.)
  • the RRP or a portion thereof may be used in screening assays of phagemid or B-lymphocyte immunoglobulin libraries to identify antibodies having the desired specificity.
  • Numerous protocols for competitive binding or immunoassays using either polyclonal or monoclonal antibodies with estabhshed specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between the protein and its specific antibody.
  • a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes is preferred, but a competitive binding assay may also be employed (Pound (1998) Immunochemical Protocols, Humana Press, Totowa NJ).
  • reporter molecules and conjugation techniques are known by those skilled in the art and maybe used in various nucleic acid, amino acid, and antibody assays. Synthesis of labeled molecules may be achieved using commercially available kits (Promega, Madison WI) for incorporation of a labeled nucleotide such as 32 P-dCTP (APB), Cy3-dCTP or Cy5-dCTP (Operon Technologies, Alameda CA), or amino acid such as 35 S-methionine (APB).
  • APB 32 P-dCTP
  • Cy3-dCTP Cy3-dCTP
  • Cy5-dCTP Opon Technologies, Alameda CA
  • amino acid such as 35 S-methionine (APB).
  • Nucleotides and amino acids may be directly labeled with a variety of substances including fluorescent, chemiluminescent, or chromogenic agents, and the like, by chemical conjugation to amines, thiols and other groups present in the molecules using reagents such as BIODEPY or FTTC (Molecular Probes, Eugene OR).
  • the cDNAs, fragments, oligonucleotides, complementary RNA and DNA molecules, and PNAs may be used to detect and quantify differential gene expression for diagnosis of a disorder.
  • antibodies which specifically bind RRP may be used to quantitate the protein.
  • Disorders associated with differential expression include cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the diagnostic assay may use hybridization or amplification technology to compare gene expression in a biological sample from a patient to standard samples in order to detect differential gene expression. Qualitative or quantitative methods for this comparison are well known in the art.
  • the cDNA or probe may be labeled by standard methods and added to a biological sample from a patient under conditions for the formation of hybridization complexes. After an incubation period, the sample is washed and the amount of label (or signal) associated with hybridization complexes, is quantified and compared with a standard value. If 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.
  • normal and disease expression profiles are estabhshed. This is accomplished by combining a sample taken from normal subjects, either animal or human, with a cDNA under conditions for hybridization to occur. Standard hybridization complexes may be quantified by comparing the values obtained using normal subjects with values from an experiment in which a known amount of a purified sequence is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who were diagnosed with a particular condition, disease, or disorder. Deviation from standard values toward those associated with a particular disorder is used to diagnose that disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies or in clinical trials or to monitor the treatment of an individual patient.
  • 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.
  • Detection and quantification of a protein using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (REAs), and fluorescence activated cell sorting (FACS).
  • ELISAs enzyme-linked immunosorbent assays
  • REAs radioimmunoassays
  • FACS fluorescence activated cell sorting
  • RRP Rasplasia
  • human RAX g4927416; SEQ ED NO: 16
  • RRP and RAX share three double-stranded RNA binding motifs.
  • differential expression of RRP is associated with cancer in various tissues and Crohn's disease as shown in Table 2.
  • RRP clearly plays a role in cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
  • the an inhibitor, antagonist or antibody of the protein may be administered to a subject to treat a condition associated with increased expression or activity.
  • a pharmaceutical composition comprising an inhibitor, antagonist or antibody in conjunction with a pharmaceutical carrier may be administered to a subject to treat a condition associated with the increased expression or activity of the endogenous protein.
  • a vector expressing the complement of the cDNA or fragments thereof may be administered to a subject to treat the disorder.
  • the protein, an agonist or enhancer may be administered to a subject to treat a condition associated with decreased expression or activity.
  • a pharmaceutical composition comprising the protein, an agonist or enhancer in conjunction with a pharmaceutical carrier may be administered to a subject to treat a condition associated with the decreased expression or activity of the endogenous protein.
  • a vector expressing cDNA may be administered to a subject to treat the disorder.
  • any of the cDNAs, complementary molecules, or fragments thereof, proteins or portions thereof, vectors delivering these nucleic acid molecules or expressing the proteins, and their ligands may be administered in combination with other therapeutic agents. Selection of the agents for use in combination therapy may be made by one of ordinary skill in the art according to conventional pharmaceutical principles. A combination of therapeutic agents may act synergistically to affect treatment of a particular disorder at a lower dosage of each agent. Modification of Gene Expression Using Nucleic Acids
  • Gene expression may be modified by designing complementary or antisense molecules (DNA, RNA, or PNA) to the control, 5', 3', or other regulatory regions of the gene encoding RRP.
  • DNA complementary or antisense molecules
  • Oligonucleotides designed to inhibit transcription initiation are preferred. Similarly, inhibition can be achieved using triple helix base-pairing which inhibits the binding of polymerases, transcription factors, or regulatory molecules (Gee et al. In: Huber and Carr (1994) Molecular and Immunologic Approaches, Futura Pubhshing, Mt. Kisco NY, pp. 163-177).
  • a complementary molecule may also be designed to block translation by preventing binding between ribosomes and mRNA.
  • a library or plurality of cDNAs may be screened to identify those which specifically bind a regulatory, nontranslated sequence.
  • Ribozymes enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA followed by endonucleolytic cleavage at sites such as GUA,
  • oligonucleotide with the same sequence may be evaluated for secondary structural features which would render the oligonucleotide inoperable.
  • the suitability of candidate targets may also be evaluated by testing their hybridization with complementary oligonucleotides using ribonuclease protection assays.
  • Complementary nucleic acids and ribozymes of the invention may be prepared via recombinant expression, in vitro or in vivo, or using sohd phase phosphoramidite chemical synthesis.
  • RNA molecules may be modified to increase intracellular stability and half-life by addition of flanking sequences at the 5 ' and/or 3 ' ends of the molecule or by the use of phosphorothioate or 2 ' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. Modification is inherent in the production of PNAs and can be extended to other nucleic acid molecules.
  • the cDNA encoding RRP may be used to screen a library of molecules or compounds for specific binding affinity.
  • the libraries may be aptamers, DNA molecules, RNA molecules, PNAs, peptides, proteins such as transcription factors, enhancers, repressors, and other ligands which regulate the activity, replication, transcription, or translation of the endogenous gene.
  • the assay involves combining a polynucleotide with a library of molecules under conditions allowing specific binding, and detecting specific binding to identify at least one molecule which specifically binds the single-stranded or double-stranded molecule.
  • the cDNA of the invention may be incubated with a plurality of purified molecules or compounds and binding activity determined by methods well known in the art, e.g., a gel- retardation assay (USPN 6,010,849) or a reticulocyte lysate transcriptional assay.
  • 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 and may be later confirmed by recovering and raising antibodies against that molecule or compound. When these antibodies are added into the assay, they cause a supershift in the gel-retardation assay.
  • the cDNA may be used to purify a molecule or compound using affinity chromatography methods well known in the art.
  • 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.
  • the protein or a portion thereof may be used to purify a Ugand from a sample.
  • a method for using a protein or a portion thereof to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, detecting specific binding between the protein and Ugand, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified Ugand.
  • RRP may be used to screen a pluraUty of molecules or compounds in any of a variety of screening assays.
  • the portion of the protein employed in such screening may be free in solution, affixed to an abiotic or biotic substrate (e.g. borne on a cell surface), or located intraceUularly.
  • viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a peptide on their cell surface can be used in screening assays.
  • the cells are screened against a pluraUty or libraries of Ugands, and the specificity of binding or formation of complexes between the expressed protein and the Ugand may be measured. Specific binding between the protein and molecule may be measured.
  • the assay may be used to identify DNA molecules, RNA molecules, peptide nucleic acids, peptides, proteins, mimetics, agonists, antagonists, antibodies, immunoglobuUns, inhibitors, and drugs or any other Ugand, which specifically binds the protein.
  • this invention comtemplates a method for high throughput screening using very small assay volumes and very small amounts of test compound as described in USPN 5,876,946, incorporated herein by reference.
  • this method is used to screen large numbers of molecules and compounds via specific binding.
  • 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.
  • Molecules or compounds identified by screening may be used in a mammaUan model system to evaluate their toxicity, diagnostic, or therapeutic potential.
  • compositions are those substances wherein the active ingredients are contained in an effective amount to achieve a desired and intended purpose.
  • the determination of an effective dose is well within the capabihty of those skilled in the art.
  • the therapeuticaUy effective dose may be estimated initially either in ceU culture assays or in animal models. The animal model is also used to achieve a desirable concentration range and route of administration. Such information may then be used to determine useful doses and routes for administration in humans.
  • a therapeuticaUy effective dose refers to that amount of protein or inhibitor which ameUorates the symptoms or condition.
  • Therapeutic efficacy and toxicity of such agents may be determined by standard pharmaceutical procedures in ceU cultures or experimental animals, e.g., ED 50 (the dose therapeuticaUy effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, and it may be expressed as the ratio, LD 50 /ED 50 .
  • Pharmaceutical compositions which exhibit large therapeutic indexes are prefe ⁇ ed. The data obtained from ceU culture assays and animal studies are used in formulating a range of dosage for human use.
  • Model Systems 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, Ufespan, reproductive potential, and abundant reference Uterature. Inbred and outbred rodent strains provide a convenient model for investigation of the physiological consequences of under- or over-expression of genes of interest and for the development of methods for diagnosis and treatment of diseases. A mammal inbred to over- express a particular gene (for example, secreted in milk) may also serve as a convenient source of the protein expressed by that gene. Toxicology
  • Toxicology is the study of the effects of agents on Uving systems. The majority of toxicity studies are performed on rats or mice. Observation of quaUtative and quantitative changes in physiology, behavior, homeostatic processes, and lethaUty in the rats or mice are used to generate a toxicity profile and to assess potential consequences on human health foUowing exposure to the agent. Genetic toxicology identifies and analyzes the effect of an agent on the rate of endogenous, spontaneous, and induced genetic mutations. Genotoxic agents usuaUy have common chemical or physical properties that faciUtate interaction with nucleic acids and are most harmful when chromosomal aberrations are transmitted to progeny.
  • Toxicological studies may identify agents that increase the frequency of structural or functional abnormaUties in the tissues of the progeny if administered to either parent before conception, to the mother during pregnancy, or to the developing organism. Mice and rats are most frequently used in these tests because their short reproductive cycle aUows the production of the numbers of organisms needed to satisfy statistical requirements.
  • Acute toxicity tests are based on a single administration of an agent to the subject to determine the symptomology or lethaUty of the agent. Three experiments are conducted: 1) an initial dose-range-finding experiment, 2) an experiment to narrow the range of effective doses, and 3) a final experiment for estabUshing the dose-response curve.
  • Subchronic toxicity tests are based on the repeated administration of an agent. Rat and dog are commonly used in these studies to provide data from species in different famiUes. With the exception of carcinogenesis, there is considerable evidence that daily administration of an agent at high-dose concentrations for periods of three to four months wiU reveal most forms of toxicity in adult animals.
  • Transgenic rodents that over-express or under-express a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents.
  • the introduced gene may be activated at a specific time in a specific tissue type during fetal or postnatal development. Expression of the tiansgene 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 chaUenge with experimental drug therapies.
  • Embryonic Stem CeUs Embryonic (ES) stem cells isolated from rodent embryos retain the potential to form embryonic tissues.
  • ES ceUs are the prefe ⁇ ed ceUs used in the creation of experimental knockout and knockin rodent strains.
  • Mouse ES ceUs such as the mouse 129/SvJ ceU line, are derived from the early mouse embryo and are grown under culture conditions well known in the art.
  • Vectors used to produce a transgenic strain contain a disease gene candidate and a marker gen, the latter serves to identify the presence of the introduced disease gene.
  • the vector is transformed into ES ceUs by methods weU known in the art, and transformed ES cells are identified and microinjected into mouse ceU blastocysts such as those from the C57BL/6 mouse strain.
  • the blastocysts are surgicaUy transfe ⁇ ed to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains.
  • ES ceUs derived from human blastocysts may be manipulated in vitro to differentiate into at least eight separate ceU Uneages. These Uneages are used to study the differentiation of various cell types and tissues in vitro, and they include endoderm, mesoderm, and ectodermal ceU types which differentiate into, for example, neural ceUs, hematopoietic Uneages, and cardiomyocytes. Knockout Analysis
  • a region of a mammaUan gene is enzymatically modified to include a non-mammaUan gene 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.
  • Transformed cells are injected into rodent blastulae, and the blastulae are implanted into pseudopregnant dams.
  • Transgenic progeny are crossbred to obtain homozygous inbred lines which lack a functional copy of the mammaUan gene.
  • the mammaUan gene is a human gene.
  • Knockin Analysis ES ceUs can be used to create knockin humanized animals (pigs) or transgenic animal models (mice or rats) 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 ceU genome. Transformed ceUs are injected into blastulae and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of the analogous human condition. These methods have been used to model several human diseases.
  • NHPs are the first choice test animal.
  • NHPs and individual humans exhibit differential sensitivities to many drugs and toxins and can be classified as a range of phenotypes from "extensive metaboUzers" to "poor metaboUzers" of these agents.
  • the cDNAs which encode the protein may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of cDNAs that are cu ⁇ ently known, including, but not Umited to, such properties as the triplet genetic code and specific base pair interactions.
  • the LUNGFET03 hbrary was constructed from fetal lung tissue obtained from an anencephaUc Caucasian female fetus (specimen #RU95-10-0739) who died at 20 weeks gestation.
  • the frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments, Westbury NJ) in guanidinium isothiocyanate solution.
  • the lysate was centrifuged over a 5J M CsCl cushion using an SW28 rotor in an L8-70M ultracentrifuge (Beckman Coulter, FuUerton CA) for 18 hours at 25,000 rpm at ambient temperature.
  • the RNA was extracted with acid phenol, pH 4.7, precipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in
  • RNAse-free water and treated with DNAse at 37C.
  • the RNA was reextracted and precipitated as before.
  • the mRNA was isolated with the OLIGOTEX kit (Qiagen, Chatsworth CA) and used to construct the cDNA Ubrary.
  • the mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies) which contains a Notl primer-adaptor designed to prime the first strand cDNA synthesis at the poly(A) tail of mRNAs. Double stranded cDNA was blunted, Ugated to EcoRI adaptors and digested with Notl (New England Biolabs, Beverly MA). The cDNAs were fractionated on a SEPHAROSE CL4B column (APB), and those cDNAs exceeding 400 bp were Ugated into pENCY plasmid (Incyte Genomics). The plasmid pENCY was subsequently transformed into DH5 ⁇ competent ceUs (Life Technologies).
  • the normaUzation of the human prostate Ubrary is described.
  • the PROSNON01 normaUzed cDNA Ubrary was constructed from microscopically normal prostate tissue obtained from a 28-year-old Caucasian male (specimen #RA95-09-0667). The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments) in guanidinium isothiocyanate solution. The lysate was extracted with acid phenol at pH 4.7 per Stratagene's RNA isolation protocol (Stratagene).
  • the primer to template ratio in the primer extension reaction was increased from 2:1 to 10:1.
  • the ddNTP concentration in this reaction was reduced to 150 ⁇ M each ddNTP to aUow generation of longer primer extension products.
  • the reannealing hybridization was extended from 13 to 48 hours.
  • the single stranded DNA circles of the normaUzed Ubrary were purified by hydroxyapatite chromatography and converted to partiaUy double-stranded by random priming, followed by electroporation into DH10B competent bacteria (Life Technologies). Subtracted Promonocyte Cells
  • THP1AZS08 the subtracted THP-1 promonocyte cell Ubrary
  • THP1AZS08 was constructed from THP-1 promonocyte ceUs treated for three days with 5-aza-2'-deoxycytidine.
  • THP-1 (ATCC TEB 202) is a human promonocyte cell line derived from peripheral blood of a 1 -year-old Caucasian male with acute monocytic leukemia (Int J Cancer (1980) 26:171). The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments) in guanidinium isothiocyanate solution.
  • the lysate was extracted with acid phenol at pH 4.7 per Stratagene's RNA isolation protocol (Stratagene).
  • the RNA was extracted with an equal volume of acid phenol, reprecipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in DEPC-treated water, and treated with DNase for 25 min at 37C. Extraction and precipitation were repeated as before.
  • the mRNA was isolated using the OLIGOTEX kit (Qiagen) and used to construct the cDNA Ubrary. The mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies).
  • cDNAs were fractionated on a SEPHAROSE CL4B column (APB), and those cDNAs exceeding 400 bp were Ugated into PSPORT I plasmid (Life Technologies). The plasmid was transformed into competent ELECTROMAX DH10B cells (Life Technologies).
  • THP1AZSO8 was constructed by subtraction of an untreated control THP1 ceU line Ubrary (5 x 10 6 clones THP1NOT02 clones) from a 5-aza-2'-deoxycytidine-treated THP-1 ceU Une Ubrary (5.76 x 10 6 THP1AZT02 clones) as foUows.
  • These plasmid Ubraries were grown in DH12S cells (Life Technologies) Uquid culture under carbenicilUn (25mg/l) and methicilUn (lmg/ml) selection foUowing transformation by electroporation.
  • the culture was aUowed to grow to an OD600 of 0.2 as monitored with a DU-7 spectrophotometer (Beckman Coulter) and then superinfected with a 5-fold excess of the helper phage M13K07 according to the method of Vieira et al. (1987; Methods Enzymol 153:3-11).
  • the cDNA Ubrary was then subtracted in two rounds of hybridization using a methodology adapted from Soares et al. (supra); Swaroop et al. (1991; Nucleic Acid Res 19:1954); and Bonaldo et al. (1996; Genome Research 6:791- 806).
  • the THP1AZT02 single-stranded Ubrary was gel and hydroxyapatite purified according to the method described in Soares et al. (supra).
  • THP1NOT02 The hybridization probe for subtraction, THP1NOT02 was generated by in vitro transcription using the MEGASCREPT kit (Ambion, Austin TX) with SP6 RNA polymerase and 40% biotin-14-CTP (Life Technologies) foUowing linearization of the double stranded plasmid DNA with Eco RI.
  • the purified single-stranded template DNA was prehybridized according to the method of Bonaldo (supra); and hybridized as described in Soares (supra).
  • the single stranded cDNA Ubrary derived from the 5-aza-2'-deoxycytidine-treated ceUs was hybridized for 48 hours with a 300:1 molar ratio of biotinylated riboprobe derived from the control ceU Ubrary, THP1NOT02. FoUowing each hybridization step, the single stranded DNA (subtracted
  • the plasmid was constructed by digesting the PSPORT1 plasmid (Life Technologies) with EcoRI restriction enzyme (New England Biolabs, Beverly MA) and filling the overhanging ends using Klenow enzyme (New England Biolabs) and 2'-deoxynucleotide 5 -triphosphates (dNTPs).
  • the plasmid was self-Ugated and transformed into the bacterial host, E. coU strain JM109.
  • An intermediate plasmid, pSPORT 1- ⁇ RI which showed no digestion with EcoRI, was digested with Hind HI (New England Biolabs); and the overhanging ends were fiUed in with Klenow and dNTPs.
  • a linker sequence was phosphorylated, Ugated onto the 5 'blunt end, digested with EcoRI, and self-Ugated. FoUowing transformation into JM109 host ceUs, plasmids were isolated and tested for preferential digestibiUty with EcoRI, but not with Hind HI. A single colony that met this criteria was designated pENCY plasmid.
  • the plasmid After testing the plasmid for its abiUty to incorporate cDNAs from a Ubrary prepared using Notl and EcoRI restriction enzymes, several clones were sequenced; and a single clone containing an insert of approximately 0.8 kb was selected from which to prepare a large quantity of the plasmid. After digestion with Notl and EcoRI, the plasmid was isolated on an agarose gel and purified using a QIAQUICK column (Qiagen) for use in Ubrary construction.
  • QIAQUICK column Qiagen
  • Plasmid DNA was released from the ceUs and purified using either the IVTINEPREP kit (Edge Biosystems, Gaithersburg MD) or the REAL PREP 96 plasmid kit (Qiagen).
  • a kit consists of a 96- weU block with reagents for 960 purifications.
  • the recommended protocol was employed except for the foUowing changes: 1) the bacteria were cultured in 1 ml of sterile TERRIFIC BROTH (APB) with carbenicilUn at 25 mg/1 and glycerol at 0.4%; 2) after inoculation, the ceUs were cultured for 19 hours and then lysed with 0.3 ml of lysis buffer; and 3) foUowing isopropanol precipitation, the plasmid DNA peUet was resuspended in 0.1 ml of distilled water. After the last step in the protocol, samples were transfe ⁇ ed to a 96-weU block for storage at 4C.
  • the cDNAs were prepared for sequencing using the MICROLAB 2200 system (Hamilton) in combination with the DNA ENGINE thermal cycler (MJ Research).
  • the cDNAs were sequenced by the method of Sanger and Coulson (1975; J Mol Biol 94:441-448) using an ABI PRISM 377 sequencing system (AppUed Biosystems) or the MEGABACE 1000 DNA sequencing system (APB). Most of the isolates were sequenced according to standard ABI protocols and kits (AppUed
  • the cDNAs were extended using the cDNA clone and oUgonucleotide 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 commerciaUy available primer analysis software 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 68C to about 72C. Any stretch of nucleotides that would result in hairpin structures and primer-primer dimerizations was avoided. Selected cDNA Ubraries were used as templates to extend the sequence. If more than one extension was necessary, additional or nested sets of primers were designed.
  • Prefe ⁇ ed Ubraries have been size-selected to include larger cDNAs and random primed to contain more sequences with 5' or upstream regions of genes. Genomic Ubraries are used to obtain regulatory elements, especially extension into the 5' promoter binding region. High fidehty ampUfication was obtained by PCR using methods such as that taught in USPN
  • PCR was performed in 96-weU plates using the DNA ENGINE thermal cycler (MJ Research).
  • the reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg 2+ , (NH jS ,, and ⁇ -mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94C, three min; Step 2: 94C, 15 sec; Step
  • Step 3 60C, one min; Step 4: 68C, two min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68C, five min; Step 7: storage at 4C.
  • the parameters for primer pair T7 and SK+ were as follows: Step 1: 94C, three min; Step 2: 94C, 15 sec; Step 3: 57C, one min; Step 4: 68C, two min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68C, five min; Step 7: storage at 4C.
  • the concentration of DNA in each weU was determined by dispensing 100 ⁇ l PICOGREEN quantitation reagent (0.25% reagent in lx TE, v/v; Molecular Probes) and 0.5 ⁇ l of undiluted PCR product into each weU of an opaque fluorimeter plate (Corning, Acton MA) and aUowing 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 ⁇ l to 10 ⁇ l aUquot of the reaction mixture was analyzed by electrophoresis on a 1% agarose minigel to determine which reactions were successful in extending the sequence.
  • the extended clones were desalted, concentrated, transfe ⁇ ed to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to reUgation into pUC18 vector (APB).
  • CviJI cholera virus endonuclease Molecular Biology Research, Madison WI
  • APIB pUC18 vector
  • Extended clones were rehgated using T4 DNA Ugase (New England Biolabs) into pUC18 vector (APB), treated with Pfu DNA polymerase (Stratagene) to fiU-in restriction site overhangs, and transfected into E. coU competent ceUs. Transformed ceUs were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37C in 384-well plates in LB/2x carbenicilUn Uquid media.
  • the ceUs were lysed, and DNA was amplified using primers, Taq DNA polymerase (APB), and Pfu DNA polymerase (Stratagene) with the foUowing parameters: Step 1: 94C, three min; Step 2: 94C, 15 sec; Step 3: 60C, one min; Step 4: 72C, two min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72C, five min; Step 7: storage at 4C.
  • DNA was quantified using PICOGREEN quantitation reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamphfied using the conditions described above.
  • Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the ABI PRISM BIGDYE terminator cycle sequencing kit (AppUed Biosystems).
  • DMSO dimethylsulfoxide
  • APIB DYENAMIC DIRECT cycle sequencing kit
  • ABI PRISM BIGDYE terminator cycle sequencing kit AppUed Biosystems
  • the cDNAs of the Sequence Listing or their deduced amino acid sequences were used to query databases such as GenBank, SwissProt, BLOCKS, and the like. These databases that contain previously identified and annotated sequences or domains were searched using BLAST or BLAST2 to produce aUgnments and to determine which sequences were exact matches or homologs.
  • the aUgnments were to sequences of prokaryotic (bacterial) or eukaryotic (animal, fungal, or plant) origin.
  • algorithms such as the one described in Smith and Smith (1992, Protein Engineering 5:35-51) could have been used to deal with primary sequence patterns and secondary structure gap penalties.
  • AU of the sequences disclosed in this appUcation have lengths of at least 49 nucleotides, and no more than 12% uncaUed bases (where N is recorded rather than A, C, G, or T).
  • BLAST matches between a query sequence and a database sequence were evaluated statisticaUy and only reported when they satisfied the threshold of 10 25 for nucleotides and 10" 14 for peptides. Homology was also evaluated by product score calculated as foUows: the % nucleotide or amino acid identity [between the query and reference sequences] in
  • BLAST is multipUed by the % maximum possible BLAST score [based on the lengths of query and reference sequences] and then divided by 100. In comparison with hybridization procedures used in the laboratory, the stringency for an exact match was set from a lower limit of about 40 (with 1-2% e ⁇ or due to uncaUed bases) to a 100% match of about 70.
  • the BLAST software suite (NCBI, Bethesda MD; http://www.ncbi.nln .nm.gov/gorf/bl2.html), includes various sequence analysis programs including 'blastn" that is used to aUgn nucleotide sequences and BLAST2 that is used for direct pairwise comparison of either nucleotide or amino acid sequences.
  • BLAST programs are commonly used with gap and other parameters set to default settings, e.g.: Matrix: BLOSUM62; Reward for match: 1; Penalty for mismatch: -2; Open Gap: 5 and Extension Gap: 2 penalties; Gap x drop-off: 50; Expect: 10;
  • cDNAs of this appUcation were compared with assembled consensus sequences or templates found in the LEFESEQ GOLD database (Incyte Genomics).
  • Component sequences from cDNA, extension, fuU length, and shotgun sequencing projects were subjected to PHRED analysis and assigned a quaUty score.
  • AU sequences with an acceptable quaUty score were subjected to various pre-processing and editing pathways to remove low quaUty 3' ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, and bacterial contamination sequences.
  • Edited sequences had to be at least 50 bp in length, and low-information sequences and repetitive elements such as dinucleotide repeats, Alu repeats, and the like, were replaced by "Ns" or masked. Edited sequences were subjected to assembly procedures in which the sequences were assigned to gene bins. Each sequence could only belong to one bin, and sequences in each bin were assembled to produce a template. Newly sequenced components were added to existing bins using BLAST and CROSSMATCH. To be added to a bin, the component sequences had to have a BLAST quaUty score greater than or equal to 150 and an aUgnment of at least 82% local identity. The sequences in each bin were assembled using PHRAP. Bins with several overlapping component sequences were assembled using DEEP PHRAP. The orientation of each template was determined based on the number and orientation of its component sequences.
  • Bins were compared to one another, and those having local similarity of at least 82% were combined and reassembled. Bins having templates with less than 95% local identity were spUt. Templates were subjected to analysis by S lTl ' CHER/EXON MAPPER algorithms that determine the probabiUties of the presence of spUce variants, alternatively spUced exons, spUce junctions, differential expression of alternative spUced genes across tissue types or disease states, and the like. Assembly procedures were repeated periodicaUy, and templates were annotated using BLAST against GenBank databases such as GBpri. An exact match was defined as having from 95% local identity over 200 base pairs through 100% local identity over 100 base pairs and a homolog match as having an E-value
  • the cDNA was further analyzed using MACDNASIS PRO software (Hitachi Software Engineering), and LASERGENE software (DNASTAR) and queried against pubUc databases such as the GenBank rodent, mammalian, vertebrate, prokaryote, and eukaryote databases, SwissProt, BLOCKS, PRINTS, PFAM, and Prosite.
  • the cDNAs are appUed to a substrate by one of the foUowing methods.
  • a mixture of cDNAs is fractionated by gel electrophoresis and transfe ⁇ ed to a nylon membrane by capillary transfer.
  • the cDNAs are individuaUy Ugated to a vector and inserted into bacterial host cells to form a Ubrary.
  • the cDNAs are then a ⁇ anged on a substrate by one of the foUowing methods.
  • bacterial ceUs containing individual clones are robotically picked and a ⁇ anged on a nylon membrane.
  • the membrane is placed on LB agar containing selective agent (carbenicilUn, kanamycin, ampicilUn, or chloramphenicol depending on the vector used) and incubated at 37C 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 ), neutraUzing solution (1.5 M NaCl, 1 M Tris, pH 8.0), and twice in 2xSSC for 10 min each.
  • the membrane is then UV i ⁇ adiated in a STRATALENKER UV- crossUnker (Stratagene).
  • cDNAs are ampUfied from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert.
  • PCR ampUfication increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 ⁇ g.
  • AmpUfied nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are a ⁇ anged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobiUzed by denaturation, neutiaUzation, and UV irradiation as described above.
  • Purified nucleic acids are roboticaUy a ⁇ anged and immobiUzed on polymer-coated glass sUdes using the procedure described in USPN 5,807,522.
  • Polymer-coated sUdes are prepared by cleaning glass microscope shdes (Corning, Acton MA) by ultrasound in 0.1% SDS and acetone, etching in 4% hydrofluoric acid (VWR Scientific Products, West Chester PA), coating with 0.05% aminopropyl silane (Sigma Aldrich) in 95% ethanol, and curing in a 110C oven. The sUdes are washed extensively with distiUed water between and after treatments.
  • the nucleic acids are a ⁇ anged on the sUde and then immobiUzed by exposing the a ⁇ ay to UV i ⁇ adiation using a STRATALENKER UV- crosshnker (Stratagene). Arrays are then washed at room temperature in 0.2% SDS and rinsed three times in distiUed water. Non-specific binding sites are blocked by incubation of a ⁇ ays in 0.2% casein in phosphate buffered saUne (PBS; Tropix, Bedford MA) for 30 min at 60C; then the a ⁇ ays are washed in 0.2% SDS and rinsed in distiUed water as before. Probe Preparation for Membrane Hybridization
  • Hybridization probes derived from the 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 ⁇ l TE buffer, denaturing by heating to 100C for five min, and briefly centrifuging. The denatured cDNA is then added to a RED ⁇ PREME tube (APB), gently mixed until blue color is evenly distributed, and briefly centi ⁇ fuged. Five ⁇ l of [ 32 P]dCTP is added to the tube, and the contents are incubated at 37C for 10 min.
  • APB RED ⁇ PREME tube
  • probe is purified from unincorporated nucleotides using a PROBEQUANT G-50 microcolumn (APB).
  • APB PROBEQUANT G-50 microcolumn
  • Hybridization probes derived from mRNA isolated from samples are employed for screening cDNAs of the Sequence Listing in a ⁇ ay-based hybridizations.
  • Probe is prepared using the GEMbright kit (Incyte Genomics) by diluting mRNA to a concentration of 200 ng in 9 ⁇ l TE buffer and adding 5 ⁇ l
  • Yeast control mRNAs are synthesized by in vitro transcription from noncoding yeast genomic DNA (W. Lei, unpubhshed). As quantitative controls, one set of control mRNAs at 0.002 ng, 0.02 ng, 0.2 ng, and 2 ng are diluted into reverse transcription reaction mixture at ratios of 1:100,000, 1:10,000, 1:1000, and 1:100 (w/w) to sample mRNA respectively.
  • RNA differential expression patterns a second set of control mRNAs are diluted into reverse transcription reaction mixture at ratios of 1:3, 3:1, 1:10, 10:1, 1:25, and 25:1 (w/w). The reaction mixture is mixed and incubated at 37C for two hr. The reaction mixture is then incubated for 20 min at 85C, and probes are purified using two successive CHROMA SPEN+TE 30 columns (Clontech, Palo Alto CA). Purified probe is ethanol precipitated by diluting probe to 90 ⁇ l in
  • Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and lxhigh phosphate buffer (0.5 M NaCl, 0.1 M Na 2 HPO 4 , 5 mM EDTA, pH 7) at 55C 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 55C for 16 hr. FoUowing hybridization, the membrane is washed for 15 min at 25C in lmM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 min each at 25C in lmM Tris (pH 8.0).
  • XOMAT-AR film Eastman Kodak, Rochester NY
  • Polymer Coated SUde-based Hybridization Probe is heated to 65C for five min, centrifuged five min at 9400 rpm in a 5415C microcentrifuge (Eppendorf Scientific, Westbury NY), and then 18 ⁇ l is aUquoted onto the array surface and covered with a coversUp.
  • the a ⁇ ays are transfe ⁇ ed to a waterproof chamber having a cavity just sUghtly larger than a microscope sUde.
  • the chamber is kept at 100% humidity internaUy by the addition of 140 ⁇ l of 5xSSC in a corner of the chamber.
  • the chamber containing the a ⁇ ays is incubated for about 6.5 hr at 60C.
  • the a ⁇ ays are washed for 10 min at 45C in lxSSC, 0.1% SDS, and three times for 10 min each at 45C in O.lxSSC, and dried.
  • Hybridization reactions are performed in absolute or differential hybridization formats.
  • absolute hybridization format probe from one sample is hybridized to array elements, and signals are detected after hybridization complexes form. Signal strength co ⁇ elates with probe mRNA levels in the sample.
  • differential hybridization format differential expression of a set of genes in two biological samples is analyzed. Probes from the two samples are prepared and labeled with different labeling moieties. A mixture of the two labeled probes is hybridized to the a ⁇ ay elements, and signals are examined under conditions in which the emissions from the two different labels are individuaUy detectable. Elements on the a ⁇ ay that are hybridized to substantiaUy equal numbers of probes derived from both biological samples give a distinct combined fluorescence (Shalon WO95/35505).
  • Hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Santa Clara CA) capable of generating spectral lines at 488 run for excitation of Cy3 and at 632 nm for excitation of Cy5.
  • the excitation laser Ught is focused on the a ⁇ ay using a 20X microscope objective (Nikon, MelviUe NY).
  • the sUde containing the a ⁇ ay is placed on a computer-controUed X-Y stage on the microscope and raster-scanned past the objective with a resolution of 20 micrometers.
  • the two fluorophores are sequentiaUy excited by the laser.
  • Emitted Ught is spUt, based on wavelength, into two photomultipUer tube detectors (PMT R1477, Hamamatsu Photonics Systems, Bridgewater NJ) co ⁇ esponding to the two fluorophores.
  • Appropriate filters positioned between the a ⁇ ay and the photomultipUer tubes are used to filter the signals.
  • the emission maxima of the fluorophores used are 565 nm for Cy3 and 650 nm for Cy5.
  • the sensitivity of the scans is caUbrated using the signal intensity generated by the yeast control mRNAs added to the probe mix.
  • a specific location on the a ⁇ ay contains a complementary DNA sequence, allowing the intensity of the signal at that location to be co ⁇ elated with a weight ratio of hybridizing species of 1 : 100,000.
  • the output of the photomultipUer tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood MA) instaUed in an IBM-compatible PC computer.
  • the digitized data are displayed as an image where the signal intensity is mapped using a Unear 20-color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal).
  • the data is also analyzed quantitatively. Where two different fluorophores are excited and measured simultaneously, the data are first co ⁇ ected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using the emission spectrum for each fluorophore.
  • a grid is superimposed over the fluorescence signal image such that the signal from each spot is centered in each element of the grid.
  • the fluorescence signal within each element is then integrated to obtain a numerical value corresponding to the average intensity of the signal.
  • the software used for signal analysis is the GEMTOOLS program (Incyte Genomics).
  • BLAST was used to search for identical or related molecules in the GenBank or LEFESEQ databases (Incyte Genomics).
  • the product score for human and rat sequences was calculated as follows: the BLAST score is multiphed by the % nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences), such that a 100% aUgnment over the length of the shorter sequence gives a product score of 100.
  • the product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match wiU be exact within a 1% to 2% e ⁇ or, and with a product score of at least 70, the match wiU be exact. Similar or related molecules are usuaUy identified by selecting those which show product scores between 8 and 40.
  • AU sequences and cDNA Ubraries in the LEFESEQ database were categorized by system, organ/tissue and ceU type. The categories included cardiovascular system, connective tissue, digestive system, embryonic structures, endocrine system, exocrine glands, female and male genitaUa, germ ceUs, hemic/immune system, Uver, musculoskeletal system, nervous system, pancreas, respiratory system, sense organs, skin, stomatognathic system, unclassified/mixed, and the urinary tract. For each category, the number of Ubraries in which the sequence was expressed were counted and shown over the total number of Ubraries in that category. In a non-normaUzed Ubrary, expression levels of two or more are significant.
  • Molecules complementary to the cDNA from about 5 (PNA) to about 5000 bp (complement of a cDNA insert), are used to detect or inhibit gene expression. Detection is described in Example VQ.
  • the complementary molecule is designed to bind to the most unique 5' sequence and includes nucleotides of the 5' UTR upstream of the initiation codon of the open reading frame.
  • Complementary molecules include genomic sequences (such as enhancers or introns) and are used in "triple heUx" base pairing to compromise the abiUty of the double heUx to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules.
  • a complementary molecule is designed to prevent ribosomal binding to the mRNA encoding the protein.
  • Complementary molecules are placed in expression vectors and used to transform a cell line to test efficacy; into an organ, tumor, synovial cavity, or the vascular system for transient or short term therapy; or into a stem ceU, zygote, or other reproducing lineage for long term or stable gene therapy.
  • Transient expression lasts for a month or more with a non-repUcating vector and for three months or more if appropriate elements for inducing vector repUcation are used in the transformation/expression system.
  • X Expression of RRP Expression and purification of the protein are achieved using either a mammaUan ceU expression system or an insect cell expression system.
  • the pUB6/V5-His vector system (Invitrogen, Carlsbad CA) is used to express RRP in CHO cells.
  • the vector contains the selectable bsd gene, multiple cloning sites, the promoter/enhancer sequence from the human ubiquitin C gene, a C-terminal V5 epitope for antibody detection with anti-V5 antibodies, and a C-terminal polyhistidine (6xHis) sequence for rapid purification on PROBOND resin (Invitrogen).
  • Transformed ceUs are selected on media containing blasticidin.
  • Spodoptera frugiperda (Sf9) insect ceUs are infected with recombinant Autographica caUfornica nuclear polyhedrosis virus (baculovirus).
  • the polyhedrin gene is replaced with the cDNA by homologous recombination and the polyhedrin promoter drives cDNA transcription.
  • the protein is synthesized as a fusion protein with 6xhis which enables purification as described above. Purified protein is used in the foUowing activity and to make antibodies
  • RRP is purified using polyacrylamide gel electrophoresis and used to immunize mice or rabbits. Antibodies are produced using the protocols below. Alternatively, the amino acid sequence of RRP is analyzed using LASERGENE software (DNASTAR) to determine regions of high antigenicity. An antigenic epitope, usuaUy found near the C-terminus or in a hydrophiUc region is selected, synthesized, and used to raise antibodies.
  • epitopes of about 15 residues in length are produced using an ABI 431 A peptide synthesizer (AppUed Biosystems) using Fmoc-chemistry and coupled to KLH (Sigma- Aldrich) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester to increase antigenicity.
  • ABI 431 A peptide synthesizer AppUed Biosystems
  • KLH Sigma- Aldrich
  • Rabbits are immunized with the epitope-KLH complex in complete Freund's adjuvant. Immunizations are repeated at intervals thereafter in incomplete Freund's adjuvant. After a minimum of seven weeks for mouse or twelve weeks for rabbit, antisera are drawn and tested for antipeptide activity. Testing involves binding the peptide to plastic, blocking with 1% bovine serum albumin, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG. Methods weU known in the art are used to determine antibody titer and the amount of complex formation.
  • the cDNA, or fragments thereof, or the protein, or portions thereof, are labeled with 32 P- dCTP, Cy3-dCTP, or Cy5-dCTP (APB), or with BIODIPY or FTTC (Molecular Probes, Eugene OR), respectively.
  • Libraries of candidate molecules or compounds previously a ⁇ anged on a substrate are incubated in the presence of labeled cDNA or protein. After incubation under conditions for either a nucleic acid or amino acid sequence, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed, and the Ugand is identified. 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.
  • a yeast two-hybrid system MATCHMAKER LexA Two-Hybrid system (Clontech Laboratories, Palo Alto CA), is used to screen for peptides that bind the protein of the invention.
  • a cDNA encoding the protein is inserted into the multiple cloning site of a pLexA vector, Ugated, and transformed into E. coU.
  • cDNA, prepared from mRNA is inserted into the multiple cloning site of a pB42AD vector, Ugated, and transformed into E. coU to construct a cDNA Ubrary.
  • the pLexA plasmid and pB42AD-cDNA Ubrary constructs are isolated from E.
  • coU coU and used in a 2:1 ratio to co- transform competent yeast EGY48[p8op-lacZ] ceUs using a polyethylene glycol/Uthium acetate protocol.
  • Transformed yeast ceUs are plated on synthetic dropout (SD) media lacking histidine (-His), tryptophan (-Trp), and uracil (-Ura), and incubated at 30C until the colonies have grown up and are counted.
  • SD synthetic dropout
  • the colonies are pooled in a minimal volume of lx TE (pH 7.5), replated on SD/-His/-Leu/- TrpAUra media supplemented with 2% galactose (Gal), 1% raffinose (Raf), and 80 mg/ml 5-bromo-4- chloro-3-indolyl ⁇ -d-galactopyranoside (X-Gal), and subsequently examined for growth of blue colonies. Interaction between expressed protein and cDNA fusion proteins activates expression of a LEU2 reporter gene in EGY48 and produces colony growth on media lacking leucine (-Leu).
  • Interaction also activates expression of ⁇ -galactosidase from the p8op-lacZ reporter construct that produces blue color in colonies grown on X-Gal.
  • Positive interactions between expressed protein and cDNA fusion proteins are verified by isolating individual positive colonies and growing them in SD/-Trp/-Ura Uquid medium for 1 to 2 days at 30C.
  • a sample of the culture is plated on SD/-Trp/-Ura media and incubated at 30C until colonies appear.
  • the sample is repUca-plated on SD/-Trp/-Ura and SD/-His/-Trp/-Ura plates. Colonies that grow on SD containing histidine but not on media lacking histidine have lost the pI_ «xA plasmid.
  • Histidine-requiring colonies are grown on SD/Gal/Raf/X-Gal/-Trp/-Ura, and white colonies are isolated and propagated.
  • the pB42AD-cDNA plasmid which contains a cDNA encoding a protein that physicaUy interacts with the protein, is isolated from the yeast ceUs and characterized.
  • RRP activity is dete ⁇ nined in assays for PKR binding.
  • RRP is labeled with 125 I Bolton-Hunter reagent (Bolton and Hunter (1973) Biochem J 133:529-539).
  • PKR previously a ⁇ ayed in the weUs of a multi-weU plate, is incubated with the 125 I-labeled RRP, washed, and any weUs with labeled RRP-PKR complex are assayed. Data obtained using different concentrations of RRP are used to calculate values for the number, affinity, and association of RRP with PKR.
  • CONFTUP01 1234 adipose tumor, lipoSAR, 3' CGAP 1 0.0810
  • PROSNOT28 3822 prostate, AH, mw/adenoCA, 55M, m/PROSTUT16 1 0.0262
  • PROETUP02 3615 prostate tumor, cancer, 45M, m/PROETMP01/02, CGAP
  • BRAINOT14 5199 brain frontal, mw/astrocytoma, 40F, m/BRAITUT12/ BRAFTUE03
  • BLADNOT06 3739 bladder, mw/TC CA, aw/prostate TC CA, 66M, m/BLADTUT05

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention provides cDNA which encodes a RAX-related protein. It also provides for the use of the cDNA, fragments, complements, and variants thereof and of the encoded protein, portions thereof and antibodies thereto for diagnosis and treatment of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. The invention additionally provides expression vectors and host cells for the production of the protein and a transgenic model system.

Description

RAX-RELATED PROTEIN
FIELD OF THE INVENTION
This invention relates to cDNA which encodes a RAX-related protein and to the use of the cDNA and the encoded protein in the diagnosis and treatment of cell proliferative disorders including cancer and Crohn's disease.
BACKGROUND OF THE INVENTION Phylogenetic relationships among organisms have been demonstrated many times, and studies from a diversity of prokaryotic and eukaryotic organisms suggest a more or less gradual evolution of molecules, biochemical and physiological mechanisms, and metabolic pathways. Despite different evolutionary pressures, the proteins of nematode, fly, rat, and man have common chemical and structural features and generally perform the same cellular function. Comparisons of the nucleic acid and protein sequences from organisms where structure and/or function are known accelerate the investigation of human sequences and allow the development of model systems for testing diagnostic and therapeutic agents for human conditions, diseases, and disorders.
Double-stranded RNA-dependent protein kinase (PKR) mediates the anticancer and antiviral effects of interferon (Williams (1999) Oncogene 18:6112-6120). During viral infection, interferon induces expression of PKR, a serine/threonine protein kinase that blocks viral replication by inhibiting protein synthesis and cell growth, and inducing apoptosis. PKR is activated by binding to viral double-stranded RNA, which promotes autophosphorylation. PKR inhibits protein synthesis at the stage of translation initiation by phosphorylating the α subunit of eukaryotic initiation factor-2 (eEF- 2 ). RNA-binding is mediated by two double-stranded RNA-binding domains (DRBDs) at the N- terminus of PKR. The DRBDs are also required for interaction of PKR with ribosomes and protein regulatory factors that contain DRBDs (Zhu et al. (1997) J Biol Chem 272:14434-14441; Patel and Sen (1998) EMBO J 17:4379-4390). In addition to regulating protein translation, PKR has a role in promoting apoptosis. Activation of PKR induces expression of members of the tumor necrosis factor receptor family, including Fas and Bax (Siddharth et al. (1998) EMBO J 17:6888-6902). Mutations that inactivate PKR or decrease expression of PKR cause tumorigenesis in mice (Meurs et al. (1993) Proc Natl Acad Sci 90:232-236). Murine RAX (PKR-associated protein X) was identified in a screen using the yeast two- hybrid system for proteins that interact with PKR (Ito et al. (1999) J Biol Chem 274:15427-15432). RAX is a 35 kDa protein of 313 amino acids that has three DRBDs, which mediate interactions with double-stranded RNA and PKR. The association of RAX with PKR is enhanced in cells where IL-3 concentrations are decreased or in response to chemical stresses such as sodium arsenite, thapsigargin, and peroxide. Under these conditions, RAX is phosphorylated at a serine residue by an unknown stress-activated protein kinase. RAX activates PKR in the absence of dsRNA and may function in regulating protein translation, cell-proliferation, and apoptosis in non-virally infected cells. PACT (PKR activating protein) is a human homolog of RAX that also binds to and activates PKR in response to cellular stresses (Patel and Sen (supra); Chandrashekhar et al. (2000) J Biol
Chem 275:37993-37998). Overexpression of PACT in mammalian cells is associated with phosphorylation of eIF-2 , inhibition of protein synthesis, and apoptosis. In response to stress signals such as serum starvation or treatment with peroxide or arsenite, PACT becomes phosphorylated and binds to PKR with increased affinity. Like RAX, PACT activates PKR in the absence of double- stranded RNA. Both PACT and RAX apparently participate in signal transduction pathways responsive to cellular stress that induce inhibition of protein synthesis and apoptosis.
The discovery of a cDNA encoding RAX-related protein satisfies a need in the art by providing compositions which are useful in the diagnosis and treatment of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
SUMMARY OF THE INVENTION The invention is based on the discovery of a cDNA encoding RAX-related protein (RRP) which is useful in the diagnosis and treatment of cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
The invention provides an isolated cDNA comprising a nucleic acid sequence encoding a protein having the amino acid sequence of SEQ ID NO:l. The invention also provides an isolated cDNA or the complement thereof selected from the group consisting of a nucleic acid sequence of SEQ ID NO:2, a fragment of SEQ ID NO:2 selected from SEQ ID NOs:3-12, and a variant of SEQ ID NO:2 selected from SEQ ID NOs:13-15. The invention additionally provides a composition, a substrate, and a probe comprising the cDNA, or the complement of the cDNA, encoding RRP. The invention further provides a vector containing the cDNA, a host cell containing the vector and a method for using the cDNA to make RRP. The invention still further provides a transgenic cell line or organism comprising the vector containing the cDNA encoding RRP. The invention additionally provides a fragment, a variant, or the complement of the cDNA selected from the group consisting of
SEQ ID Nos:2-15. In one aspect, the invention provides a substrate containing at least one of these fragments or variants or the complements thereof. In a second aspect, the invention provides a probe comprising a cDNA or the complement thereof which can be used in methods of detection, screening, and purification. In a further aspect, the probe is a single-stranded complementary RNA or DNA molecule.
The invention provides a method for using a cDNA to detect the differential expression of a nucleic acid in a sample comprising hybridizing a probe to the nucleic acids, thereby forming hybridization complexes and comparing hybridization complex formation with a standard, wherein the comparison indicates the differential expression of the cDNA in the sample. In one aspect, the method of detection further comprises amplifying the nucleic acids of the sample prior to hybridization. In another aspect, the method showing differential expression of the cDNA is used to diagnose cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. In another aspect, the cDNA or a fragment or a variant or the complements thereof may comprise an element on an array.
The invention additionally provides a method for using a cDNA or a fragment or a variant or the complements thereof to screen a library or plurality of molecules or compounds to identify at least one ligand which specifically binds the cDNA, the method comprising combining the cDNA with the molecules or compounds under conditions allowing specific binding, and detecting specific binding to the cDNA, thereby identifying a ligand which specifically binds the cDNA. In one aspect, the molecules or compounds are selected from aptamers, DNA molecules, RNA molecules, peptide nucleic acids, artificial chromosome constructions, peptides, transcription factors, repressors, and regulatory molecules. The invention provides a purified protein or a portion thereof selected from the group consisting of an amino acid sequence of SEQ ID NO:l, a variant having at least 89% identity to the amino acid sequence of SEQ ED NO:l, an antigenic epitope of SEQ ID NO:l, and a biologically active portion of SEQ ED NO:l. The invention also provides a composition comprising the purified protein in conjunction with a pharmaceutical carrier. The invention further provides a method of using the RRP to treat a subject with cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising administering to a patient in need of such treatment the composition containing the purified protein. The invention still further provides a method for using a protein to screen a library or a plurality of molecules or compounds to identify at least one ligand, the method comprising combining the protein with the molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds the protein. In one aspect, the molecules or compounds are selected from DNA molecules, RNA molecules, peptide nucleic acids, peptides, proteins, mimetics, agonists, antagonists, antibodies, immunoglobulins, inhibitors, and drugs. In another aspect, the ligand is used to treat a subject with cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
The invention provides a method of using a protein to screen a subject sample for antibodies which specifically bind the protein comprising isolating antibodies from the subject sample, contacting the isolated antibodies with the protein under conditions that allow specific binding, dissociating the antibody from the bound-protein, and comparing the quantity of antibody with known standards, wherein the presence or quantity of antibody is diagnostic of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. The invention also provides a method of using a protein to prepare and purify antibodies comprising immunizing a animal with the protein under conditions to elicit an antibody response, isolating animal antibodies, attaching the protein to a substrate, contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein, dissociating the antibodies from the protein, thereby obtaining purified antibodies. The invention provides a purified antibody which binds specifically to a protein which is expressed in cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. The invention also provides a method of using an antibody to diagnose cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising combining the antibody comparing the quantity of bound antibody to known standards, thereby establishing the presence of cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. The invention further provides a method of using an antibody to treat cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease comprising administering to a patient in need of such treatment a pharmaceutical composition comprising the purified antibody.
The invention provides a method for inserting a heterologous marker gene into the genomic DNA of a mammal to disrupt the expression of the endogenous polynucleotide. The invention also provides a method for using a cDNA to produce a mammalian model system, the method comprising constructing a vector containing the cDNA selected from SEQ ED NOs:2-15, tiansforming the vector into an embryonic stem cell, selecting a transformed embryonic stem, microinjecting the transformed embryonic stem cell into a mammalian blastocyst, thereby foπning a chimeric blastocyst, transferring the chimeric blastocyst into a pseudopregnant dam, wherein the dam gives birth to a chimeric offspring containing the cDNA in its germ line, and breeding the chimeric mammal to produce a homozygous, mammalian model system.
BRIEF DESCRIPTION OF THE FIGURES AND TABLE Figures 1A, IB, 1C, ID, IE, and IF show the RRP (SEQ ED NO:l) encoded by the cDNA
(SEQ ED NO:2). The translation was produced using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA).
Figures 2A and 2B demonstrate the conserved chemical and structural similarities among the sequences and motifs of RRP (2754573; SEQ D NO:l) and human RAX (g4927416; SEQ ED NO: 16). The alignment was produced using the MEGALIGN program of LASERGENE software
(DNASTAR, Madison WI).
Tables 1 and 2 show the northern analysis for RRP produced using the LEFESEQ Gold database (Incyte Genomics, Palo Alto CA). In Table 1, the first column presents the tissue categories; the second column, the total number of clones in the tissue category; the third column, the ratio of the number of libraries in which at least one transcript was found to the total number of libraries; the fourth column, absolute clone abundance of the transcript; and the fifth column, percent abundance of the transcript. Table 2 shows expression of RRP in various tissues from patients with cancer or Crohn's disease. The first column lists the library name, the second column, the number of clones sequenced for that library; the third column, the description of the tissue from which the library was derived; the fourth column, the absolute abundance of the transcript; and the fifth column, the percent abundance of the transcript.
DESCRIPTION OF THE INVENTION It is understood that this invention is not limited to the particular machines, materials and methods described. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments and is not intended to limit the scope of the present invention which will be limited only by the appended claims. As used herein, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. For example, a reference to "a host cell" includes a plurality of such host cells known to those skilled in the art.
Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs.
All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention. Definitions
"RRP" refers to a purified protein obtained from any mammalian species, including bovine, canine, murine, ovine, porcine, rodent, simian, and preferably the human species, and from any source, whether natural, synthetic, semi-synthetic, or recombinant. "Array" refers to an ordered arrangement of at least two cDNAs on a substrate. At least one of the cDNAs represents a control or standard, and the other, a cDNA of diagnostic or therapeutic interest. The arrangement of from about two to about 40,000 cDNAs on the substrate assures that the size and signal intensity of each labeled hybridization complex formed between each cDNA and at least one sample nucleic acid is individually distinguishable. The "complement" of a cDNA of the Sequence Listing refers to a nucleic acid molecule which is completely complementary over its full length and which will hybridize to the cDNA or an mRNA under conditions of maximal stringency.
"cDNA" refers to an isolated polynucleotide, nucleic acid molecule, or any fragment or complement thereof. It may have originated recombinantly or synthetically, may be double-stranded or single-stranded, represents coding and noncoding 3' or 5' sequence, and generally lacks introns.
The phrase "cDNA encoding a protein" refers to a nucleotide 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) which provides identity within the conserved region (Altschul (1993) J Mol Evol 36: 290-300; Altschul et al. (1990) J Mol Biol 215:403-410).
A "composition" comprises the polynucleotide and a labeling moiety or a purified protein in conjunction with a pharmaceutical carrier.
"Derivative" refers to a cDNA or a protein that has been subjected to a chemical modification. Derivatization of a cDNA can involve substitution of a nontraditional base such as queosine or of an analog such as hypoxanthine. These substitutions are well known in the art.
Derivatization of a protein involves the replacement of a hydrogen by an acetyl, acyl, alkyl, amino, formyl, or morpholino group. Derivative molecules retain the biological activities of the naturally occurring molecules but may confer advantages such as longer lifespan or enhanced activity.
"Differential expression" refers to an increased, upregulated or present, or decreased, downregulated or absent, gene expression as detected by presence, absence or at least two-fold changes in the amount of transcribed messenger RNA or translated protein in a sample.
"Disorder" refers to conditions, diseases or syndromes in which the cDNAs and RRP are differentially expressed. Such a disorder includes cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
"Fragment" refers to a chain of consecutive nucleotides from about 50 to about 4000 base pairs in length. Fragments may be used in PCR or hybridization technologies to identify related nucleic acid molecules and in binding assays to screen for a ligand. Such ligands are useful as therapeutics to regulate replication, transcription or translation.
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'. Hybridization conditions, degree of complementarity and the use of nucleotide analogs affect the efficiency and stringency of hybridization reactions. "Labeling moiety" refers to any visible or radioactive label than can be attached to or incorporated into a cDNA or protein. Visible labels include but are not limited to anthocyanins, green fluorescent protein (GFP), β glucuronidase, luciferase, Cy3 and Cy5, and the like. Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like.
"Ligand" refers to any agent, molecule, or compound which will bind specifically to a polynucleotide or to an epitope of a protein. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic and/or organic substances including minerals, cofactors, nucleic acids, proteins, carbohydrates, fats, and lipids.
"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. Substantially equivalent terms are amplimer, primer, and oligomer.
"Portion" refers to any part of a protein used for any purpose; but especially, to an epitope for the screening of ligands or for the production of antibodies.
"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.
"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 with reporter molecules for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.
"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 epitope of the protein identified using Kyte-Doolittle algorithms of the PROTEAN program (DNASTAR, Madison WI). 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.
"Purified" refers to any molecule or compound that is separated from its natural environment and is from about 60% free to about 90% free from other components with which it is naturally associated.
"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; a tissue print; a fingerprint, buccal cells, skin, or hair; and the like.
"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 or the binding between an epitope of a protein and an agonist, antagonist, or antibody. "Similarity" as applied to sequences, refers to the quantification (usually percentage) of nucleotide or residue matches between at least two sequences aligned using a standardized algorithm such as Smith- Waterman alignment (Smith and Waterman (1981) J Mol Biol 147:195-197) or BLAST2 (Altschul et al. (1997) Nucleic Acids Res 25:3389-3402). BLAST2 maybe 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. Particularly in proteins, similarity is greater than identity in that conservative substitutions, for example, valine for leucine or isoleucine, are counted in calculating the reported percentage. Substitutions which are considered to be conservative are well known in the art.
"Substrate" refers to any rigid or semi-rigid support to which cDNAs or proteins are bound and includes membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, capillaries or other tubing, plates, polymers, and microparticles with a variety of surface forms including wells, trenches, pins, channels and pores.
"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 or its secondary, tertiary, or quaternary structure. THE INVENTION
The invention is based on the discovery of a cDNA which encodes RRP and on the use of the cDNA, or fragments thereof, and protein, or portions thereof, directly or as compositions in the characterization, diagnosis, and treatment of cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
Nucleic acids encoding the RRP of the present invention were first identified in Incyte Clone 2754573 from the THP-1 promonocyte cell cDNA library (THP1AZS08) using a computer search for nucleotide and/or amino acid sequence alignments. SEQ ED NO:2 was derived from the following overlapping and/or extended nucleic acid sequences (SEQ ED NO:3-12): Incyte Clones 2754573H1
(THP1AZS08), 491644H1 (HNT2AGT01), 412307R1 (BRSTNOT01), 263630H1 (HNT2AGT01), 412307T6 (BRSTNOT01), 1253094H1 (LUNGFET03), 2280508H1 (PROSNON01), 2270603H1 (PROSNON01), 2375670H1 (ISLTNOTOl), and 3151587H1 (ADRENON04). Table 1 shows expression of RRP across various tissue categories (also listed in Example VEEI). Table 2 shows expression of RRP specifically in tissues from patients with cancer or Crohn's disease. RRP is found in libraries associated with various cancers, in particular, an adipose tissue library (CONFTUPOl) from a patient with liposarcoma, a mesentery tissue library (CONUTUTOl) from a patient with metastasizing mixed-mullerian tumors, a spinal soft tissue library (CONNTUT04) from a patient with schwannoma, a gallbladder tissue library (GBLATUT01) from a patient with squamous cell carcinoma, and lung tissue libraries (LUNLTUT04, LUNGTUT09, and LUNGTUT08) from patients with lung cancer. RRP shows overexpression in a brain tissue library (BRAFTUE03) from a patient with astrocytoma compared to a library (BRAENOT14) from matched (m) microscopically normal tissue from the same donor. RRP is overexpressed in a lung tissue library (LUNGTUT03) from a patient with squamous cell carcinoma compared to a library (LUNGNOT15) from matched (m) microscopically normal tissue from the same donor. RRP is notably overexpressed in a bladder tissue library (BLADTUT05) from a patient with transitional cell carcinoma compared to a library (BLADNOT06) from matched (m) microscopically normal tissue from the same donor. RRP is underexpressed in prostate tissue libraries (PROETUP02, PROSTUT16, PROSTUT18) from patients with prostate cancer compared to libraries (PROETMPOl, PROSNOT28, and PROSTMT03) from matched (m) microscopically normal tissue from the same donors. RRP is overexpressed in a colon tissue library (COLNCRT01) from a patient with Crohn's disease compared to a library (COLNNOT05) from matched (m) microscopically normal tissue from the same donor. The transcript is therefore useful in diagnostic assays for cancers, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. A fragment of the cDNA from about nucleotide 60 to about nucleotide 120 is also useful in diagnostic assays.
In one embodiment, the invention encompasses a polypeptide comprising the amino acid sequence of SEQ ED NO:l as shown in Figures 1A, IB, IC, ID, IE, and IF. RRP is 352 amino acids in length and has two potential N-glycosylation sites at N141 and N294; one potential myristoylation site at G12; five potential casein kinase II phosphorylation sites at T110, S169, S206, S260, and T296; and eight potential protein kinase C phosphorylation sites at S8, T67, T106, T121, S122, T210, S215, and S256. PFAM analysis indicates that the regions of RRP from P74 to L138, from P166 to F231, and from Y280 to L345 are similar to a double-stranded RNA binding motif. BLOCKS analysis indicates that the regions of RRP from Gl 18 to L138 and from N165 to Y184 are similar to
Ribonuclease EQ family motifs; and the region of RRP from T13 to T24 is similar to an RNA 3'- terminal phosphate cyclase motif. As shown in Figures 2 A and 2B, RRP has chemical and structural similarity with human RAX (g4927416; SEQ ID NO:16). In particular, RRP and RAX share about 88% identity and three double-stranded RNA binding motifs. RRP differs from the human RAX primarily by an additional N-terminal extension of about 40 amino acid residues. Useful antigenic epitopes extend from about A68 to about G82, from about D150 to about G168, and from about R258 to about S271; and biologically active portions of RRP extend from about P74 to about L138, from about P166 to about F231, and from about Y280 to about L345. An antibody which specifically binds RRP is useful in an diagnostic assay to identify cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease
Mammalian variants of the cDNA encoding RRP were identified using BLAST2 with default parameters and the ZOOSEQ databases (incyte Genomics). These preferred variants have from about 86% to about 92% identity as shown in the table below. The first column shows the SEQ ED for the human cDNA (SEQ EDH); the second column, the SEQ ED for the variant cDNAs (SEQ
ED^); the third column, the clone number for the variant cDNAs (Clone^); the fourth column, the library name; the fifth column, the alignment of the variant cDNA to the human cDNA (includes the alignment of different regions of the variant cDNA with different regions of the human cDNA in some cases); and the sixth column, the percent identity to the human cDNA.
SEQ ID„ SEQ IDv.r Clonev-c Library Name NtH Alignment Identity
2 13 702148603H1 RALUTXT01 162 -545 92%
2 14 700491104F6 RABCNOT01 646 -1119 86 %
2 15 700148476H1 RANPNOT01 670 -807 91%
These cDNAs, SEQ ED NOS:13-15 are particularly useful for producing transgenic cell lines or organisms.
It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of cDNAs encoding RRP, some bearing minimal similarity to the cDNAs of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible variation of cDNA that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide encoding naturally occurring RRP, and all such variations are to be considered as being specifically disclosed.
The cDNAs of SEQ ED NOs:2-15 may be used in hybridization, amplification, and screening technologies to identify and distinguish among SEQ ED NO:2 and related molecules in a sample. The mammalian cDNAs may be used to produce transgenic cell lines or organisms which are model systems for human cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease and upon which the toxicity and efficacy of potential therapeutic treatments may be tested. Toxicology studies, clinical trials, and subject/patient treatment profiles may be performed and monitored using the cDNAs, proteins, antibodies and molecules and compounds identified using the cDNAs and proteins of the present invention.
The identification and characterization of the cDNAs and proteins, fragments or portions thereof, were described in USSN 08/933,750, incorporated by reference herein in their entirety. Characterization and Use of the Invention cDNA libraries
En a particular embodiment disclosed herein, rriRNA is isolated from mammalian cells and tissues using methods which are well known to those skilled in the art and used to prepare the cDNA libraries. The Incyte cDNAs were isolated from mammalian cDNA libraries aprepared as described in the EXAMPLES. The consensus sequences are chemically and/or electronically assembled from fragments including Incyte cDNAs and extension and/or shotgun sequences using computer programs such as PHRAP (P Green, University of Washington, Seattle WA), and AUTOASSEMBLER application (Applied Biosystems, Foster City CA). After verification of the 5' and 3' sequence, at least one representative cDNA which encodes RRP is designated a reagent. Sequencing
Methods for sequencing nucleic acids are well known in the art and may be used to practice any of the embodiments of the invention. These methods employ enzymes such as the Klenow fragment of DNA polymerase I, SEQUENASE, Taq DNA polymerase and thermostable T7 DNA polymerase (Amersham Pharmacia Biotech (APB), Piscataway NJ), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE amplification system (Life Technologies, Gaithersburg MD). Preferably, sequence preparation is automated with machines such as the MICROLAB 2200 system (Hamilton, Reno NV) and the DNA ENGINE thermal cycler (MJ Research, Watertown MA). Machines commonly used for sequencing include the ABI PRISM 3700, 377 or 373 DNA sequencing systems (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (APB), and the like. The sequences may be analyzed using a variety of algorithms well known in the art and described in Ausubel et al. (1997; Short Protocols in Molecular Biology, John Wiley & Sons, New York NY, unit 7.7) and in Meyers (1995; Molecular Biology and Biotechnology. Wiley VCH, New York NY, pp. 856-853). Shotgun sequencing may also be used to complete the sequence of a particular cloned insert of interest. Shotgun strategy involves randomly breaking the original insert into segments of various sizes and cloning these fragments into vectors. The fragments are sequenced and reassembled using overlapping ends until the entire sequence of the original insert is known. Shotgun sequencing methods are well known in the art and use thermostable DNA polymerases, heat-labile DNA polymerases, and primers chosen from representative regions flanking the cDNAs of interest.
Incomplete assembled sequences are inspected for identity using various algorithms or programs such as CONSED (Gordon (1998) Genome Res 8:195-202) which are well known in the art. Contaminating sequences, including vector or chimeric sequences, or deleted sequences can be removed or restored, respectively, organizing the incomplete assembled sequences into finished sequences.
Extension of a Nucleic Acid Sequence
The sequences of the invention may be extended using various PCR-based methods known in the art. For example, the XL-PCR kit (Applied Biosystems), nested primers, and commercially available cDNA or genomic DNA libraries may be used to extend the nucleic acid sequence. For all PCR-based methods, primers may be designed using commercially available software, such as
OLIGO primer analysis software (Molecular Biology Insights, Cascade CO) to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to a target molecule at temperatures from about 55C to about 68C. When extending a sequence to recover regulatory elements, it is preferable to use genomic, rather than cDNA libraries. Hybridization
The cDNA and fragments thereof can be used in hybridization technologies for various purposes. A probe may be designed or derived from unique regions such as the 5' regulatory region or from a nonconserved region (i.e., 5 ' or 3 ' of the nucleotides encoding the conserved catalytic domain of the protein) and used in protocols to identify naturally occurring molecules encoding the RRP, allelic variants, or related molecules. The probe may be DNA or RNA, may be single-stranded, and should have at least 50% sequence identity to any of the nucleic acid sequences, SEQ ED NOs:2-15. Hybridization probes may be produced using oligolabeling, nick translation, end-labeling, or PCR amplification in the presence of a reporter molecule. A vector containing the cDNA or a fragment thereof may be used to produce an mRNA probe in vitro by addition of an RNA polymerase and labeled nucleotides. These procedures may be conducted using commercially available kits such as those provided by APB.
The stringency of hybridization is determined by G+C content of the probe, salt concentration, and temperature. In particular, stringency can be increased by reducing the concentration of salt or raising the hybridization temperature. Hybridization can be performed at low stringency with buffers, such as 5xSSC with 1% sodium dodecyl sulfate (SDS) at 60C, which permits the formation of a hybridization complex between nucleic acid sequences that contain some mismatches. Subsequent washes are performed at higher stringency with buffers such as 0.2xSSC with 0.1% SDS at either 45C (medium stringency) or 68C (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 can be added to the hybridization solution to reduce the temperature at which hybridization is performed, and background signals can 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 (supra) and
Sambrook et al. (1989) Molecular Cloning. A Laboratory Manual, Cold Spring Harbor Press, Plainview NY.
Arrays may be prepared and analyzed using methods well known in the art. Oligonucleotides or cDNAs may be used as hybridization probes or targets to monitor the expression level of large numbers of genes simultaneously or to identify genetic variants, mutations, and single nucleotide polymorphisms. Arrays may be used to determine gene function; to understand the genetic basis of a condition, disease, or disorder; to diagnose a condition, disease, or disorder; and to develop and monitor the activities of therapeutic agents. (See, e.g., Brennan et al. (1995) USPN 5,474,796; Schena et al. (1996) Proc Natl Acad Sci 93:10614-10619; Heller et al. (1997) Proc Natl Acad Sci 94:2150-2155; and Heller et al. (1997) USPN 5,605,662.)
Hybridization probes are also useful in mapping the naturally occurring genomic sequence. The probes may be hybridized to a particular chromosome, a specific region of a chromosome, or an artificial chromosome construction. Such constructions include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC), bacterial PI constructions, or the cDNAs of libraries made from single chromosomes. Expression
Any one of a multitude of cDNAs encoding RRP may be cloned into a vector and used to express the protein, or portions thereof, in host cells. The nucleic acid sequence can be engineered by such methods as DNA shuffling (USPN 5,830,721) and site-directed mutagenesis to create new restriction sites, alter glycosylation patterns, change codon preference to increase expression in a particular host, produce splice variants, extend half-life, and the like. The expression vector may contain transcriptional and translational control elements (promoters, enhancers, specific initiation signals, and polyadenylated 3 ' sequence) from various sources which have been selected for their efficiency in a particular host. The vector, cDNA, and regulatory elements are combined using in vitro recombinant DNA techniques, synthetic techniques, and/or in vivo genetic recombination techniques well known in the art and described in Sambrook (supra, ch. 4, 8, 16 and 17).
A variety of host systems may be transformed with an expression vector. These include, but are not limited to, bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems transformed with baculovirus expression vectors; plant cell systems transformed with expression vectors containing viral and/or bacterial elements, or animal cell systems (Ausubel supra, unit 16). For example, an adenovirus transcription/translation complex may be utilized in mammalian cells. After sequences are ligated into the El or E3 region of the viral genome, the infective virus is used to transform and express the protein in host cells. The Rous sarcoma virus enhancer or SV40 or EBV-based vectors may also be used for high-level protein expression.
Routine cloning, subcloning, and propagation of nucleic acid sequences can be achieved using the multifunctional PBLUESCREPT vector (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Introduction of a nucleic acid sequence into the multiple cloning site of these vectors disrupts the lacZ gene and allows colorimetric screening for transformed bacteria. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence.
For long term production of recombinant proteins, the vector can be stably transformed into cell lines along with a selectable or visible marker gene on the same or on a separate vector. After transformation, cells are allowed to grow for about 1 to 2 days in enriched media and then are transfeπed to selective media. Selectable markers, antimetabolite, antibiotic, or herbicide resistance genes, confer resistance to the relevant selective agent and allow growth and recovery of cells which successfully express the introduced sequences. Resistant clones identified either by survival on selective media or by the expression of visible markers may be propagated using culture techniques. Visible markers are also used to estimate the amount of protein expressed by the introduced genes. Verification that the host cell contains the desired cDNA is based on DNA-DNA or DNA-RNA hybridizations or PCR amplification techniques.
The host cell may be chosen for its ability to modify a recombinant protein in a desired fashion. Such modifications include acetylation, carboxylation, glycosylation, phosphorylation, lipidation, acylation and the like. Post-translational processing which cleaves a "prepro" form may also be used to specify protein targeting, folding, and/or activity. Different host cells available from the ATCC (Manassas VA) which have specific cellular machinery and characteristic mechanisms for post-translational activities may be chosen to ensure the coπect modification and processing of the recombinant protein.
Recovery of Proteins from Cell Culture
Heterologous moieties engineered into a vector for ease of purification include glutathione S- transferase (GST), 6xHis, FLAG, MYC, and the like. GST and 6-His are purified using commercially available affinity matrices such as immobilized glutathione and metal-chelate resins, respectively. FLAG and MYC are purified using commercially available monoclonal and polyclonal antibodies. For ease of separation following purification, a sequence encoding a proteolytic cleavage site may be part of the vector located between the protein and the heterologous moiety. Methods for recombinant protein expression and purification are discussed in Ausubel (supra, unit 16) and are commercially available. Chemical Synthesis of Peptides
Proteins or portions thereof may be produced not only by recombinant methods, but also by using chemical methods well known in the art. Solid phase peptide synthesis may be carried out in a batchwise or continuous flow process which sequentially adds α-amino- and side chain-protected amino acid residues to an insoluble polymeric support via a linker group. A linker group such as memylamine-derivatized polyethylene glycol is attached to poly(styrene-co-divinylbenzene) to form the support resin. The amino acid residues are N-α-protected by acid labile Boc (t-butyloxycarbonyl) or base-labile Fmoc (9-fluorenylmethoxycarbonyl). The carboxyl group of the protected amino acid is coupled to the amine of the linker group to anchor the residue to the solid phase support resin. Trifluoroacetic acid or piperidine are used to remove the protecting group in the case of Boc or Fmoc, respectively. Each additional amino acid is added to the anchored residue using a coupling agent or pre-activated amino acid derivative, and the resin is washed. The full length peptide is synthesized by sequential deprotection, coupling of derivitized amino acids, and washing with dichloromethane and/or N, N-dimethylformamide. The peptide is cleaved between the peptide carboxy terminus and the linker group to yield a peptide acid or amide. (Novabiochem 1997/98 Catalog and Peptide Synthesis Handbook, San Diego CA pp. S1-S20). Automated synthesis may also be carried out on machines such as the ABI 431 A peptide synthesizer (Applied Biosystems). A protein or portion thereof may be substantially purified by preparative high performance liquid chromatography and its composition confirmed by amino acid analysis or by sequencing (Creighton (1984) Proteins, Structures and Molecular Properties, WH Freeman, New York NY).
Preparation and Screening of Antibodies
Various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with RRP or any portion thereof. Adjuvants such as Freund's, mineral gels, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemacyanin (KLH), and dinitrophenol may be used to increase immunological response. The ohgopeptide, peptide, or portion of protein used to induce antibodies should consist of at least about five amino acids, more preferably ten amino acids, which are identical to a portion of the natural protein. Oligopeptides may be fused with proteins such as KLH in order to produce antibodies to the chimeric molecule. Monoclonal antibodies may be prepared using any technique which provides for the production of antibodies by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler et al. (1975) Nature 256:495-497; Kozbor et al. (1985) J. Immunol Methods 81:31-42; Cote et al. (1983) Proc Natl Acad Sci 80:2026-2030; and Cole et al. (1984) Mol Cell Biol 62:109-120.) Alternatively, techniques described for antibody production may be adapted, using methods known in the art, to produce epitope-specific, single chain antibodies. Antibody fragments which contain specific binding sites for epitopes of the protein may also be generated. For example, such fragments include, but are not limited to, F(ab 2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e.g., Huse et al. (1989) Science 246:1275-1281.)
The RRP or a portion thereof may be used in screening assays of phagemid or B-lymphocyte immunoglobulin libraries to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoassays using either polyclonal or monoclonal antibodies with estabhshed specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between the protein and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes is preferred, but a competitive binding assay may also be employed (Pound (1998) Immunochemical Protocols, Humana Press, Totowa NJ).
Labeling of Molecules for Assay
A wide variety of reporter molecules and conjugation techniques are known by those skilled in the art and maybe used in various nucleic acid, amino acid, and antibody assays. Synthesis of labeled molecules may be achieved using commercially available kits (Promega, Madison WI) for incorporation of a labeled nucleotide such as 32P-dCTP (APB), Cy3-dCTP or Cy5-dCTP (Operon Technologies, Alameda CA), or amino acid such as 35S-methionine (APB). Nucleotides and amino acids may be directly labeled with a variety of substances including fluorescent, chemiluminescent, or chromogenic agents, and the like, by chemical conjugation to amines, thiols and other groups present in the molecules using reagents such as BIODEPY or FTTC (Molecular Probes, Eugene OR).
DIAGNOSTICS
The cDNAs, fragments, oligonucleotides, complementary RNA and DNA molecules, and PNAs and may be used to detect and quantify differential gene expression for diagnosis of a disorder. Similarly antibodies which specifically bind RRP may be used to quantitate the protein. Disorders associated with differential expression include cancer, particularly liposarcoma, mesentery mixed- mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease. The diagnostic assay may use hybridization or amplification technology to compare gene expression in a biological sample from a patient to standard samples in order to detect differential gene expression. Qualitative or quantitative methods for this comparison are well known in the art.
For example, the cDNA or probe may be labeled by standard methods and added to a biological sample from a patient under conditions for the formation of hybridization complexes. After an incubation period, the sample is washed and the amount of label (or signal) associated with hybridization complexes, is quantified and compared with a standard value. If 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.
In order to provide standards for establishing differential expression, normal and disease expression profiles are estabhshed. This is accomplished by combining a sample taken from normal subjects, either animal or human, with a cDNA under conditions for hybridization to occur. Standard hybridization complexes may be quantified by comparing the values obtained using normal subjects with values from an experiment in which a known amount of a purified sequence is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who were diagnosed with a particular condition, disease, or disorder. Deviation from standard values toward those associated with a particular disorder is used to diagnose that disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies or in clinical trials or to monitor the treatment of an individual patient. Once the presence of a condition is estabhshed 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. Immunological Methods
Detection and quantification of a protein using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (REAs), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes is prefeπed, but a competitive binding assay may be employed. (See, e.g., Coligan et al. (1997) Cuπent Protocols in Immunology, Wiley-lnterscience, New York NY; and Pound, supra.) THERAPEUTICS
Chemical and structural similarity exists between regions of RRP (SEQ ED NO:l) and human RAX (g4927416; SEQ ED NO: 16) as shown in Figures 2A and 2B. In particular, RRP and RAX share three double-stranded RNA binding motifs. In addition, differential expression of RRP is associated with cancer in various tissues and Crohn's disease as shown in Table 2. RRP clearly plays a role in cancer, particularly liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
In the treatment of conditions associated with increased expression of the RRP such as liposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gallbladder cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease, it is desirable to decrease expression or protein activity. In one embodiment, the an inhibitor, antagonist or antibody of the protein may be administered to a subject to treat a condition associated with increased expression or activity. In another embodiment, a pharmaceutical composition comprising an inhibitor, antagonist or antibody in conjunction with a pharmaceutical carrier may be administered to a subject to treat a condition associated with the increased expression or activity of the endogenous protein. In an additional embodiment, a vector expressing the complement of the cDNA or fragments thereof may be administered to a subject to treat the disorder.
In the treatment of conditions associated with decreased expression of the RRP such as prostate cancer, it is desirable to increase expression or protein activity. In one embodiment, the protein, an agonist or enhancer may be administered to a subject to treat a condition associated with decreased expression or activity. In another embodiment, a pharmaceutical composition comprising the protein, an agonist or enhancer in conjunction with a pharmaceutical carrier may be administered to a subject to treat a condition associated with the decreased expression or activity of the endogenous protein. In an additional embodiment, a vector expressing cDNA may be administered to a subject to treat the disorder.
Any of the cDNAs, complementary molecules, or fragments thereof, proteins or portions thereof, vectors delivering these nucleic acid molecules or expressing the proteins, and their ligands may be administered in combination with other therapeutic agents. Selection of the agents for use in combination therapy may be made by one of ordinary skill in the art according to conventional pharmaceutical principles. A combination of therapeutic agents may act synergistically to affect treatment of a particular disorder at a lower dosage of each agent. Modification of Gene Expression Using Nucleic Acids
Gene expression may be modified by designing complementary or antisense molecules (DNA, RNA, or PNA) to the control, 5', 3', or other regulatory regions of the gene encoding RRP.
Oligonucleotides designed to inhibit transcription initiation are preferred. Similarly, inhibition can be achieved using triple helix base-pairing which inhibits the binding of polymerases, transcription factors, or regulatory molecules (Gee et al. In: Huber and Carr (1994) Molecular and Immunologic Approaches, Futura Pubhshing, Mt. Kisco NY, pp. 163-177). A complementary molecule may also be designed to block translation by preventing binding between ribosomes and mRNA. In one alternative, a library or plurality of cDNAs may be screened to identify those which specifically bind a regulatory, nontranslated sequence.
Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA followed by endonucleolytic cleavage at sites such as GUA,
GUU, and GUC. Once such sites are identified, an oligonucleotide with the same sequence may be evaluated for secondary structural features which would render the oligonucleotide inoperable. The suitability of candidate targets may also be evaluated by testing their hybridization with complementary oligonucleotides using ribonuclease protection assays. Complementary nucleic acids and ribozymes of the invention may be prepared via recombinant expression, in vitro or in vivo, or using sohd phase phosphoramidite chemical synthesis. In addition, RNA molecules may be modified to increase intracellular stability and half-life by addition of flanking sequences at the 5 ' and/or 3 ' ends of the molecule or by the use of phosphorothioate or 2 ' O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. Modification is inherent in the production of PNAs and can be extended to other nucleic acid molecules. Either the inclusion of nontraditional bases such as inosine, queosine, and wybutosine, and or the modification of adenine, cytidine, guanine, thymine, and uridine with acetyl-, methyl-, thio- groups renders the molecule less available to endogenous endonucleases. Screening and Purification Assays
The cDNA encoding RRP may be used to screen a library of molecules or compounds for specific binding affinity. The libraries may be aptamers, DNA molecules, RNA molecules, PNAs, peptides, proteins such as transcription factors, enhancers, repressors, and other ligands which regulate the activity, replication, transcription, or translation of the endogenous gene. The assay involves combining a polynucleotide with a library of molecules under conditions allowing specific binding, and detecting specific binding to identify at least one molecule which specifically binds the single-stranded or double-stranded molecule.
In one embodiment, the cDNA of the invention may be incubated with a plurality of purified molecules or compounds and binding activity determined by methods well known in the art, e.g., a gel- retardation assay (USPN 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 and may be later confirmed by recovering and raising antibodies against that molecule or compound. When these antibodies are added into the assay, they cause a supershift in the gel-retardation assay.
En 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.
In a further embodiment, the protein or a portion thereof may be used to purify a Ugand from a sample. A method for using a protein or a portion thereof to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, detecting specific binding between the protein and Ugand, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified Ugand.
In a prefeπed embodiment, RRP may be used to screen a pluraUty of molecules or compounds in any of a variety of screening assays. The portion of the protein employed in such screening may be free in solution, affixed to an abiotic or biotic substrate (e.g. borne on a cell surface), or located intraceUularly. For example, in one method, viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a peptide on their cell surface can be used in screening assays. The cells are screened against a pluraUty or libraries of Ugands, and the specificity of binding or formation of complexes between the expressed protein and the Ugand may be measured. Specific binding between the protein and molecule may be measured. Depending on the particular kind of Ubrary being screened, the assay may be used to identify DNA molecules, RNA molecules, peptide nucleic acids, peptides, proteins, mimetics, agonists, antagonists, antibodies, immunoglobuUns, inhibitors, and drugs or any other Ugand, which specifically binds the protein. En one aspect, this invention comtemplates a method for high throughput screening using very small assay volumes and very small amounts of test compound as described in USPN 5,876,946, incorporated herein by reference. This method is used to screen large numbers of molecules and compounds via specific binding. In another aspect, 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. Molecules or compounds identified by screening may be used in a mammaUan model system to evaluate their toxicity, diagnostic, or therapeutic potential. Pharmacology
Pharmaceutical compositions are those substances wherein the active ingredients are contained in an effective amount to achieve a desired and intended purpose. The determination of an effective dose is well within the capabihty of those skilled in the art. For any compound, the therapeuticaUy effective dose may be estimated initially either in ceU culture assays or in animal models. The animal model is also used to achieve a desirable concentration range and route of administration. Such information may then be used to determine useful doses and routes for administration in humans.
A therapeuticaUy effective dose refers to that amount of protein or inhibitor which ameUorates the symptoms or condition. Therapeutic efficacy and toxicity of such agents may be determined by standard pharmaceutical procedures in ceU cultures or experimental animals, e.g., ED50 (the dose therapeuticaUy effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it may be expressed as the ratio, LD50/ED50. Pharmaceutical compositions which exhibit large therapeutic indexes are prefeπed. The data obtained from ceU culture assays and animal studies are used in formulating a range of dosage for human use. Model Systems 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, Ufespan, reproductive potential, and abundant reference Uterature. Inbred and outbred rodent strains provide a convenient model for investigation of the physiological consequences of under- or over-expression of genes of interest and for the development of methods for diagnosis and treatment of diseases. A mammal inbred to over- express a particular gene (for example, secreted in milk) may also serve as a convenient source of the protein expressed by that gene. Toxicology
Toxicology is the study of the effects of agents on Uving systems. The majority of toxicity studies are performed on rats or mice. Observation of quaUtative and quantitative changes in physiology, behavior, homeostatic processes, and lethaUty in the rats or mice are used to generate a toxicity profile and to assess potential consequences on human health foUowing exposure to the agent. Genetic toxicology identifies and analyzes the effect of an agent on the rate of endogenous, spontaneous, and induced genetic mutations. Genotoxic agents usuaUy have common chemical or physical properties that faciUtate interaction with nucleic acids and are most harmful when chromosomal aberrations are transmitted to progeny. Toxicological studies may identify agents that increase the frequency of structural or functional abnormaUties in the tissues of the progeny if administered to either parent before conception, to the mother during pregnancy, or to the developing organism. Mice and rats are most frequently used in these tests because their short reproductive cycle aUows the production of the numbers of organisms needed to satisfy statistical requirements.
Acute toxicity tests are based on a single administration of an agent to the subject to determine the symptomology or lethaUty of the agent. Three experiments are conducted: 1) an initial dose-range-finding experiment, 2) an experiment to narrow the range of effective doses, and 3) a final experiment for estabUshing the dose-response curve.
Subchronic toxicity tests are based on the repeated administration of an agent. Rat and dog are commonly used in these studies to provide data from species in different famiUes. With the exception of carcinogenesis, there is considerable evidence that daily administration of an agent at high-dose concentrations for periods of three to four months wiU reveal most forms of toxicity in adult animals.
Chronic toxicity tests, with a duration of a year or more, are used to demonstrate either the absence of toxicity or the carcinogenic potential of an agent. When studies are conducted on rats, a minimum of three test groups plus one control group are used, and animals are examined and monitored at the outset and at intervals throughout the experiment. Transgenic Animal Models
Transgenic rodents that over-express or under-express a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents. (See, e.g., USPN 5,175,383 and USPN 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 tiansgene 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 chaUenge with experimental drug therapies. Embryonic Stem CeUs Embryonic (ES) stem cells isolated from rodent embryos retain the potential to form embryonic tissues. When ES cells are placed inside a carrier embryo, they resume normal development and contribute to tissues of the Uve-born animal. ES ceUs are the prefeπed ceUs used in the creation of experimental knockout and knockin rodent strains. Mouse ES ceUs, such as the mouse 129/SvJ ceU line, are derived from the early mouse embryo and are grown under culture conditions well known in the art. Vectors used to produce a transgenic strain contain a disease gene candidate and a marker gen, the latter serves to identify the presence of the introduced disease gene. The vector is transformed into ES ceUs by methods weU known in the art, and transformed ES cells are identified and microinjected into mouse ceU blastocysts such as those from the C57BL/6 mouse strain. The blastocysts are surgicaUy transfeπed to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains.
ES ceUs derived from human blastocysts may be manipulated in vitro to differentiate into at least eight separate ceU Uneages. These Uneages are used to study the differentiation of various cell types and tissues in vitro, and they include endoderm, mesoderm, and ectodermal ceU types which differentiate into, for example, neural ceUs, hematopoietic Uneages, and cardiomyocytes. Knockout Analysis
In gene knockout analysis, a region of a mammaUan gene is enzymatically modified to include a non-mammaUan gene 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. Transformed cells are injected into rodent blastulae, and the blastulae are implanted into pseudopregnant dams. Transgenic progeny are crossbred to obtain homozygous inbred lines which lack a functional copy of the mammaUan gene. In one example, the mammaUan gene is a human gene. Knockin Analysis ES ceUs can be used to create knockin humanized animals (pigs) or transgenic animal models (mice or rats) 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 ceU genome. Transformed ceUs are injected into blastulae and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of the analogous human condition. These methods have been used to model several human diseases.
Non-Human Primate Model
The field of animal testing deals with data and methodology from basic sciences such as physiology, genetics, chemistry, pharmacology and statistics. These data are paramount in evaluating the effects of therapeutic agents on non-human primates as they can be related to human health. Monkeys are used as human suπogates in vaccine and drug evaluations, and their responses are relevant to human exposures under similar conditions. Cynomolgus and Rhesus monkeys (Macaca fascicularis and Macaca mulatto, respectively) and Common Marmosets (CalUthrix iacchus) are the most common non-human primates (NHPs) used in these investigations. Since great cost is associated with developing and mamtaining a colony of NHPs, early research and toxicological studies are usually carried out in rodent models. En studies using behavioral measures such as drug addiction, NHPs are the first choice test animal. In addition, NHPs and individual humans exhibit differential sensitivities to many drugs and toxins and can be classified as a range of phenotypes from "extensive metaboUzers" to "poor metaboUzers" of these agents.
In additional embodiments, the cDNAs which encode the protein may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of cDNAs that are cuπently known, including, but not Umited to, such properties as the triplet genetic code and specific base pair interactions.
EXAMPLES The examples below are provided to iUustrate the subject invention and are not included for the purpose of Umiting the invention. The preparation of the fetal lung (LUNGFET03), normahzed prostate (PROSNON01), and subtracted THP-1 promonocyte ceU (THP1AZS08) Ubraries will be described.
I cDNA Library Construction
Fetal Lung
The LUNGFET03 hbrary was constructed from fetal lung tissue obtained from an anencephaUc Caucasian female fetus (specimen #RU95-10-0739) who died at 20 weeks gestation. The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments, Westbury NJ) in guanidinium isothiocyanate solution. The lysate was centrifuged over a 5J M CsCl cushion using an SW28 rotor in an L8-70M ultracentrifuge (Beckman Coulter, FuUerton CA) for 18 hours at 25,000 rpm at ambient temperature. The RNA was extracted with acid phenol, pH 4.7, precipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in
RNAse-free water, and treated with DNAse at 37C. The RNA was reextracted and precipitated as before. The mRNA was isolated with the OLIGOTEX kit (Qiagen, Chatsworth CA) and used to construct the cDNA Ubrary.
The mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies) which contains a Notl primer-adaptor designed to prime the first strand cDNA synthesis at the poly(A) tail of mRNAs. Double stranded cDNA was blunted, Ugated to EcoRI adaptors and digested with Notl (New England Biolabs, Beverly MA). The cDNAs were fractionated on a SEPHAROSE CL4B column (APB), and those cDNAs exceeding 400 bp were Ugated into pENCY plasmid (Incyte Genomics). The plasmid pENCY was subsequently transformed into DH5α competent ceUs (Life Technologies).
NormaUzed Prostate
For purposes of example, the normaUzation of the human prostate Ubrary (PROSNON01) is described. The PROSNON01 normaUzed cDNA Ubrary was constructed from microscopically normal prostate tissue obtained from a 28-year-old Caucasian male (specimen #RA95-09-0667). The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments) in guanidinium isothiocyanate solution. The lysate was extracted with acid phenol at pH 4.7 per Stratagene's RNA isolation protocol (Stratagene). The RNA was extracted with an equal volume of acid phenol, reprecipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in DEPC-treated water, and treated with DNase for 25 min at 37C. Extraction and precipitation were repeated as before. The mRNA was isolated using the OLIGOTEX kit (Qiagen) and used to construct the cDNA Ubrary. The mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies). cDNAs were fractionated on a SEPHAROSE CL4B column (APB), and those cDNAs exceeding 400 bp were Ugated into PSport I plasmid (Life Technologies). The plasmid was subsequently transformed into DH12S competent ceUs (Life Technologies).
4.4 x 106 independent clones were grown in Uquid culture under carbenicilUn (25mg/l) and methicilUn (lmg/ml) selection. The culture was aUowed to grow to an OD600 of 0.2 as monitored with a DU-7 spectrophotometer (Beckman Coulter) and then superinfected with a 5-fold excess of the helper phage M13K07 according to the method of Vieira et al. (1987; Methods Enzymol 153:3-11). To reduce the number of excess cDNA copies according to their abundance levels in the Ubrary, the cDNA Ubrary was then normaUzed in a single round according to the procedure of Soares et al. (1994; Proc Natl Acad Sci 91:9928-9232) with the foUowing modifications. The primer to template ratio in the primer extension reaction was increased from 2:1 to 10:1. The ddNTP concentration in this reaction was reduced to 150μM each ddNTP to aUow generation of longer primer extension products. The reannealing hybridization was extended from 13 to 48 hours. The single stranded DNA circles of the normaUzed Ubrary were purified by hydroxyapatite chromatography and converted to partiaUy double-stranded by random priming, followed by electroporation into DH10B competent bacteria (Life Technologies). Subtracted Promonocyte Cells
For purposes of example, the construction of the subtracted THP-1 promonocyte cell Ubrary (THP1AZS08) is described. THP1AZS08 was constructed from THP-1 promonocyte ceUs treated for three days with 5-aza-2'-deoxycytidine. THP-1 (ATCC TEB 202) is a human promonocyte cell line derived from peripheral blood of a 1 -year-old Caucasian male with acute monocytic leukemia (Int J Cancer (1980) 26:171). The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments) in guanidinium isothiocyanate solution. The lysate was extracted with acid phenol at pH 4.7 per Stratagene's RNA isolation protocol (Stratagene). The RNA was extracted with an equal volume of acid phenol, reprecipitated using 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in DEPC-treated water, and treated with DNase for 25 min at 37C. Extraction and precipitation were repeated as before. The mRNA was isolated using the OLIGOTEX kit (Qiagen) and used to construct the cDNA Ubrary. The mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies). cDNAs were fractionated on a SEPHAROSE CL4B column (APB), and those cDNAs exceeding 400 bp were Ugated into PSPORT I plasmid (Life Technologies). The plasmid was transformed into competent ELECTROMAX DH10B cells (Life Technologies).
THP1AZSO8 was constructed by subtraction of an untreated control THP1 ceU line Ubrary (5 x 106 clones THP1NOT02 clones) from a 5-aza-2'-deoxycytidine-treated THP-1 ceU Une Ubrary (5.76 x 106 THP1AZT02 clones) as foUows. These plasmid Ubraries were grown in DH12S cells (Life Technologies) Uquid culture under carbenicilUn (25mg/l) and methicilUn (lmg/ml) selection foUowing transformation by electroporation. The culture was aUowed to grow to an OD600 of 0.2 as monitored with a DU-7 spectrophotometer (Beckman Coulter) and then superinfected with a 5-fold excess of the helper phage M13K07 according to the method of Vieira et al. (1987; Methods Enzymol 153:3-11).
To enrich for 5-aza-2'-deoxycytidine induced transcripts, the cDNA Ubrary was then subtracted in two rounds of hybridization using a methodology adapted from Soares et al. (supra); Swaroop et al. (1991; Nucleic Acid Res 19:1954); and Bonaldo et al. (1996; Genome Research 6:791- 806). The THP1AZT02 single-stranded Ubrary was gel and hydroxyapatite purified according to the method described in Soares et al. (supra). The hybridization probe for subtraction, THP1NOT02 was generated by in vitro transcription using the MEGASCREPT kit (Ambion, Austin TX) with SP6 RNA polymerase and 40% biotin-14-CTP (Life Technologies) foUowing linearization of the double stranded plasmid DNA with Eco RI. The purified single-stranded template DNA was prehybridized according to the method of Bonaldo (supra); and hybridized as described in Soares (supra). In each round of subtraction, the single stranded cDNA Ubrary derived from the 5-aza-2'-deoxycytidine-treated ceUs was hybridized for 48 hours with a 300:1 molar ratio of biotinylated riboprobe derived from the control ceU Ubrary, THP1NOT02. FoUowing each hybridization step, the single stranded DNA (subtracted
Ubrary) was purified by streptavidin coated magnetic beads (Dynal Biotech, Oslo Norway) according to the manufacturers specifications. Following the second streptavidin separation, the single stranded subtracted Ubrary was converted to partiaUy double-stranded by random priming, foUowed by electroporation into DH10B competent bacteria (Life Technologies). II Construction of pINCY Plasmid
The plasmid was constructed by digesting the PSPORT1 plasmid (Life Technologies) with EcoRI restriction enzyme (New England Biolabs, Beverly MA) and filling the overhanging ends using Klenow enzyme (New England Biolabs) and 2'-deoxynucleotide 5 -triphosphates (dNTPs). The plasmid was self-Ugated and transformed into the bacterial host, E. coU strain JM109. An intermediate plasmid, pSPORT 1-ΔRI, which showed no digestion with EcoRI, was digested with Hind HI (New England Biolabs); and the overhanging ends were fiUed in with Klenow and dNTPs. A linker sequence was phosphorylated, Ugated onto the 5 'blunt end, digested with EcoRI, and self-Ugated. FoUowing transformation into JM109 host ceUs, plasmids were isolated and tested for preferential digestibiUty with EcoRI, but not with Hind HI. A single colony that met this criteria was designated pENCY plasmid.
After testing the plasmid for its abiUty to incorporate cDNAs from a Ubrary prepared using Notl and EcoRI restriction enzymes, several clones were sequenced; and a single clone containing an insert of approximately 0.8 kb was selected from which to prepare a large quantity of the plasmid. After digestion with Notl and EcoRI, the plasmid was isolated on an agarose gel and purified using a QIAQUICK column (Qiagen) for use in Ubrary construction.
Ill Isolation and Sequencing of cDNA Clones
Plasmid DNA was released from the ceUs and purified using either the IVTINEPREP kit (Edge Biosystems, Gaithersburg MD) or the REAL PREP 96 plasmid kit (Qiagen). A kit consists of a 96- weU block with reagents for 960 purifications. The recommended protocol was employed except for the foUowing changes: 1) the bacteria were cultured in 1 ml of sterile TERRIFIC BROTH (APB) with carbenicilUn at 25 mg/1 and glycerol at 0.4%; 2) after inoculation, the ceUs were cultured for 19 hours and then lysed with 0.3 ml of lysis buffer; and 3) foUowing isopropanol precipitation, the plasmid DNA peUet was resuspended in 0.1 ml of distilled water. After the last step in the protocol, samples were transfeπed to a 96-weU block for storage at 4C.
The cDNAs were prepared for sequencing using the MICROLAB 2200 system (Hamilton) in combination with the DNA ENGINE thermal cycler (MJ Research). The cDNAs were sequenced by the method of Sanger and Coulson (1975; J Mol Biol 94:441-448) using an ABI PRISM 377 sequencing system (AppUed Biosystems) or the MEGABACE 1000 DNA sequencing system (APB). Most of the isolates were sequenced according to standard ABI protocols and kits (AppUed
Biosystems) with solution volumes of 0.25x-1.0x concentrations. In the alternative, cDNAs were sequenced using solutions and dyes from APB. IV Extension of cDNA Sequences
The cDNAs were extended using the cDNA clone and oUgonucleotide 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 commerciaUy available primer analysis software 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 68C to about 72C. Any stretch of nucleotides that would result in hairpin structures and primer-primer dimerizations was avoided. Selected cDNA Ubraries were used as templates to extend the sequence. If more than one extension was necessary, additional or nested sets of primers were designed. Prefeπed Ubraries have been size-selected to include larger cDNAs and random primed to contain more sequences with 5' or upstream regions of genes. Genomic Ubraries are used to obtain regulatory elements, especially extension into the 5' promoter binding region. High fidehty ampUfication was obtained by PCR using methods such as that taught in USPN
5,932,451. PCR was performed in 96-weU plates using the DNA ENGINE thermal cycler (MJ Research). The reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg2+, (NH jS ,, and β-mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94C, three min; Step 2: 94C, 15 sec; Step
3: 60C, one min; Step 4: 68C, two min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68C, five min; Step 7: storage at 4C. Ln the alternative, the parameters for primer pair T7 and SK+ (Stratagene) were as follows: Step 1: 94C, three min; Step 2: 94C, 15 sec; Step 3: 57C, one min; Step 4: 68C, two min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68C, five min; Step 7: storage at 4C. The concentration of DNA in each weU was determined by dispensing 100 μl PICOGREEN quantitation reagent (0.25% reagent in lx TE, v/v; Molecular Probes) and 0.5 μl of undiluted PCR product into each weU of an opaque fluorimeter plate (Corning, Acton MA) and aUowing 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 aUquot of the reaction mixture was analyzed by electrophoresis on a 1% agarose minigel to determine which reactions were successful in extending the sequence.
The extended clones were desalted, concentrated, transfeπed to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to reUgation into pUC18 vector (APB). For shotgun sequences, the digested nucleotide sequences were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and the agar was digested with AGARACE enzyme (Promega). Extended clones were rehgated using T4 DNA Ugase (New England Biolabs) into pUC18 vector (APB), treated with Pfu DNA polymerase (Stratagene) to fiU-in restriction site overhangs, and transfected into E. coU competent ceUs. Transformed ceUs were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37C in 384-well plates in LB/2x carbenicilUn Uquid media.
The ceUs were lysed, and DNA was amplified using primers, Taq DNA polymerase (APB), and Pfu DNA polymerase (Stratagene) with the foUowing parameters: Step 1: 94C, three min; Step 2: 94C, 15 sec; Step 3: 60C, one min; Step 4: 72C, two min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72C, five min; Step 7: storage at 4C. DNA was quantified using PICOGREEN quantitation reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamphfied using the conditions described above. Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the ABI PRISM BIGDYE terminator cycle sequencing kit (AppUed Biosystems).
V Homology Searching of cDNA Clones and Their Deduced Proteins
The cDNAs of the Sequence Listing or their deduced amino acid sequences were used to query databases such as GenBank, SwissProt, BLOCKS, and the like. These databases that contain previously identified and annotated sequences or domains were searched using BLAST or BLAST2 to produce aUgnments and to determine which sequences were exact matches or homologs. The aUgnments were to sequences of prokaryotic (bacterial) or eukaryotic (animal, fungal, or plant) origin. Alternatively, algorithms such as the one described in Smith and Smith (1992, Protein Engineering 5:35-51) could have been used to deal with primary sequence patterns and secondary structure gap penalties. AU of the sequences disclosed in this appUcation have lengths of at least 49 nucleotides, and no more than 12% uncaUed bases (where N is recorded rather than A, C, G, or T).
As detailed in KarUn (supra), BLAST matches between a query sequence and a database sequence were evaluated statisticaUy and only reported when they satisfied the threshold of 1025 for nucleotides and 10"14 for peptides. Homology was also evaluated by product score calculated as foUows: the % nucleotide or amino acid identity [between the query and reference sequences] in
BLAST is multipUed by the % maximum possible BLAST score [based on the lengths of query and reference sequences] and then divided by 100. In comparison with hybridization procedures used in the laboratory, the stringency for an exact match was set from a lower limit of about 40 (with 1-2% eπor due to uncaUed bases) to a 100% match of about 70. The BLAST software suite (NCBI, Bethesda MD; http://www.ncbi.nln .nm.gov/gorf/bl2.html), includes various sequence analysis programs including 'blastn" that is used to aUgn nucleotide sequences and BLAST2 that is used for direct pairwise comparison of either nucleotide or amino acid sequences. BLAST programs are commonly used with gap and other parameters set to default settings, e.g.: Matrix: BLOSUM62; Reward for match: 1; Penalty for mismatch: -2; Open Gap: 5 and Extension Gap: 2 penalties; Gap x drop-off: 50; Expect: 10;
Word Size: 11 ; and Filter: on. Identity is measured over the entire length of a sequence. Brenner et al. (1998; Proc Natl Acad Sci 95:6073-6078, incorporated herein by reference) analyzed BLAST for its abiUty to identify structural homologs by sequence identity and found 30% identity is a reUable threshold for sequence aUgnments of at least 150 residues and 40%, for aUgnments of at least 70 residues.
The cDNAs of this appUcation were compared with assembled consensus sequences or templates found in the LEFESEQ GOLD database (Incyte Genomics). Component sequences from cDNA, extension, fuU length, and shotgun sequencing projects were subjected to PHRED analysis and assigned a quaUty score. AU sequences with an acceptable quaUty score were subjected to various pre-processing and editing pathways to remove low quaUty 3' ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, and bacterial contamination sequences. Edited sequences had to be at least 50 bp in length, and low-information sequences and repetitive elements such as dinucleotide repeats, Alu repeats, and the like, were replaced by "Ns" or masked. Edited sequences were subjected to assembly procedures in which the sequences were assigned to gene bins. Each sequence could only belong to one bin, and sequences in each bin were assembled to produce a template. Newly sequenced components were added to existing bins using BLAST and CROSSMATCH. To be added to a bin, the component sequences had to have a BLAST quaUty score greater than or equal to 150 and an aUgnment of at least 82% local identity. The sequences in each bin were assembled using PHRAP. Bins with several overlapping component sequences were assembled using DEEP PHRAP. The orientation of each template was determined based on the number and orientation of its component sequences.
Bins were compared to one another, and those having local similarity of at least 82% were combined and reassembled. Bins having templates with less than 95% local identity were spUt. Templates were subjected to analysis by S lTl'CHER/EXON MAPPER algorithms that determine the probabiUties of the presence of spUce variants, alternatively spUced exons, spUce junctions, differential expression of alternative spUced genes across tissue types or disease states, and the like. Assembly procedures were repeated periodicaUy, and templates were annotated using BLAST against GenBank databases such as GBpri. An exact match was defined as having from 95% local identity over 200 base pairs through 100% local identity over 100 base pairs and a homolog match as having an E-value
(or probabiUty score) of <1 x 10"8. The templates were also subjected to frameshift FASTx against GENPEPT, and homolog match was defined as having an E-value of <1 x 10"8. Template analysis and assembly was described in USSN 09/276,534, filed March 25, 1999.
FoUowing assembly, templates were subjected to BLAST, motif, and other functional analyses and categorized in protein hierarchies using methods described in USSN 08/812,290 and USSN
08/811,758, both filed March 6, 1997; in USSN 08/947,845, filed October 9, 1997; and in USSN 09/034,807, filed March 4, 1998. Then templates were analyzed by translating each template in aU three forward reading frames and searching each translation against the PFAM database of hidden Markov model-based protein famines and domains using the HMMER software package (Washington University School of Medicine, St. Louis MO; http://pfam.wustl.edu/). The cDNA was further analyzed using MACDNASIS PRO software (Hitachi Software Engineering), and LASERGENE software (DNASTAR) and queried against pubUc databases such as the GenBank rodent, mammalian, vertebrate, prokaryote, and eukaryote databases, SwissProt, BLOCKS, PRINTS, PFAM, and Prosite. VI Chromosome Mapping
Radiation hybrid and genetic mapping data available from pubUc resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Genethon are used to determine if any of the cDNAs presented in the Sequence Listing have been mapped. Any of the fragments of the cDNA encoding RRP that have been mapped result in the assignment of all related regulatory and coding sequences mapping to the same location. The genetic map locations are described as ranges, or intervals, of human chromosomes. The map position of an interval, in cM (which is roughly equivalent to 1 megabase of human DNA), is measured relative to the terminus of the chromosomal p-arm. VII Hybridization Technologies and Analyses ImmobiUzation of cDNAs on a Substrate
The cDNAs are appUed to a substrate by one of the foUowing methods. A mixture of cDNAs is fractionated by gel electrophoresis and transfeπed to a nylon membrane by capillary transfer. Alternatively, the cDNAs are individuaUy Ugated to a vector and inserted into bacterial host cells to form a Ubrary. The cDNAs are then aπanged on a substrate by one of the foUowing methods. In the first method, bacterial ceUs containing individual clones are robotically picked and aπanged on a nylon membrane. The membrane is placed on LB agar containing selective agent (carbenicilUn, kanamycin, ampicilUn, or chloramphenicol depending on the vector used) and incubated at 37C 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 ), neutraUzing solution (1.5 M NaCl, 1 M Tris, pH 8.0), and twice in 2xSSC for 10 min each. The membrane is then UV iπadiated in a STRATALENKER UV- crossUnker (Stratagene).
In the second method, cDNAs are ampUfied from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR ampUfication increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 μg. AmpUfied nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are aπanged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobiUzed by denaturation, neutiaUzation, and UV irradiation as described above. Purified nucleic acids are roboticaUy aπanged and immobiUzed on polymer-coated glass sUdes using the procedure described in USPN 5,807,522. Polymer-coated sUdes are prepared by cleaning glass microscope shdes (Corning, Acton MA) by ultrasound in 0.1% SDS and acetone, etching in 4% hydrofluoric acid (VWR Scientific Products, West Chester PA), coating with 0.05% aminopropyl silane (Sigma Aldrich) in 95% ethanol, and curing in a 110C oven. The sUdes are washed extensively with distiUed water between and after treatments. The nucleic acids are aπanged on the sUde and then immobiUzed by exposing the aπay to UV iπadiation using a STRATALENKER UV- crosshnker (Stratagene). Arrays are then washed at room temperature in 0.2% SDS and rinsed three times in distiUed water. Non-specific binding sites are blocked by incubation of aπays in 0.2% casein in phosphate buffered saUne (PBS; Tropix, Bedford MA) for 30 min at 60C; then the aπays are washed in 0.2% SDS and rinsed in distiUed water as before. Probe Preparation for Membrane Hybridization
Hybridization probes derived from the 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 100C for five min, and briefly centrifuging. The denatured cDNA is then added to a REDΓPREME tube (APB), gently mixed until blue color is evenly distributed, and briefly centiϊfuged. Five μl of [32P]dCTP is added to the tube, and the contents are incubated at 37C 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 100C for five min, snap cooled for two min on ice, and used in membrane-based hybridizations as described below. Probe Preparation for Polymer Coated SUde Hybridization
Hybridization probes derived from mRNA isolated from samples are employed for screening cDNAs of the Sequence Listing in aπay-based hybridizations. Probe is prepared using the GEMbright kit (Incyte Genomics) by diluting mRNA to a concentration of 200 ng in 9 μl TE buffer and adding 5 μl
5x buffer, 1 μl 0.1 M DTT, 3 μl Cy3 or Cy5 labeling mix, 1 μl RNase inhibitor, 1 μl reverse transcriptase, and 5 μl lx yeast control mRNAs. Yeast control mRNAs are synthesized by in vitro transcription from noncoding yeast genomic DNA (W. Lei, unpubhshed). As quantitative controls, one set of control mRNAs at 0.002 ng, 0.02 ng, 0.2 ng, and 2 ng are diluted into reverse transcription reaction mixture at ratios of 1:100,000, 1:10,000, 1:1000, and 1:100 (w/w) to sample mRNA respectively. To examine mRNA differential expression patterns, a second set of control mRNAs are diluted into reverse transcription reaction mixture at ratios of 1:3, 3:1, 1:10, 10:1, 1:25, and 25:1 (w/w). The reaction mixture is mixed and incubated at 37C for two hr. The reaction mixture is then incubated for 20 min at 85C, and probes are purified using two successive CHROMA SPEN+TE 30 columns (Clontech, Palo Alto CA). Purified probe is ethanol precipitated by diluting probe to 90 μl in
DEPC-treated water, adding 2 μl lmg/ml glycogen, 60 μl 5 M sodium acetate, and 300 μl 100% ethanol. The probe is centrifuged for 20 min at 20,800xg, and the peUet is resuspended in 12 μl resuspension buffer, heated to 65C for five min, and mixed thoroughly. The probe is heated and mixed as before and then stored on ice. Probe is used in high density aπay-based hybridizations as described below.
Membrane-based Hybridization
Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and lxhigh phosphate buffer (0.5 M NaCl, 0.1 M Na2HPO4, 5 mM EDTA, pH 7) at 55C 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 55C for 16 hr. FoUowing hybridization, the membrane is washed for 15 min at 25C in lmM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 min each at 25C in lmM Tris (pH 8.0). To detect hybridization complexes, XOMAT-AR film (Eastman Kodak, Rochester NY) is exposed to the membrane overnight at -70C, developed, and examined visually. Polymer Coated SUde-based Hybridization Probe is heated to 65C for five min, centrifuged five min at 9400 rpm in a 5415C microcentrifuge (Eppendorf Scientific, Westbury NY), and then 18 μl is aUquoted onto the array surface and covered with a coversUp. The aπays are transfeπed to a waterproof chamber having a cavity just sUghtly larger than a microscope sUde. The chamber is kept at 100% humidity internaUy by the addition of 140 μl of 5xSSC in a corner of the chamber. The chamber containing the aπays is incubated for about 6.5 hr at 60C. The aπays are washed for 10 min at 45C in lxSSC, 0.1% SDS, and three times for 10 min each at 45C in O.lxSSC, and dried.
Hybridization reactions are performed in absolute or differential hybridization formats. In the absolute hybridization format, probe from one sample is hybridized to array elements, and signals are detected after hybridization complexes form. Signal strength coπelates with probe mRNA levels in the sample. In the differential hybridization format, differential expression of a set of genes in two biological samples is analyzed. Probes from the two samples are prepared and labeled with different labeling moieties. A mixture of the two labeled probes is hybridized to the aπay elements, and signals are examined under conditions in which the emissions from the two different labels are individuaUy detectable. Elements on the aπay that are hybridized to substantiaUy equal numbers of probes derived from both biological samples give a distinct combined fluorescence (Shalon WO95/35505).
Hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Santa Clara CA) capable of generating spectral lines at 488 run for excitation of Cy3 and at 632 nm for excitation of Cy5. The excitation laser Ught is focused on the aπay using a 20X microscope objective (Nikon, MelviUe NY). The sUde containing the aπay is placed on a computer-controUed X-Y stage on the microscope and raster-scanned past the objective with a resolution of 20 micrometers. In the differential hybridization format, the two fluorophores are sequentiaUy excited by the laser. Emitted Ught is spUt, based on wavelength, into two photomultipUer tube detectors (PMT R1477, Hamamatsu Photonics Systems, Bridgewater NJ) coπesponding to the two fluorophores. Appropriate filters positioned between the aπay and the photomultipUer tubes are used to filter the signals. The emission maxima of the fluorophores used are 565 nm for Cy3 and 650 nm for Cy5. The sensitivity of the scans is caUbrated using the signal intensity generated by the yeast control mRNAs added to the probe mix. A specific location on the aπay contains a complementary DNA sequence, allowing the intensity of the signal at that location to be coπelated with a weight ratio of hybridizing species of 1 : 100,000.
The output of the photomultipUer tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood MA) instaUed in an IBM-compatible PC computer. The digitized data are displayed as an image where the signal intensity is mapped using a Unear 20-color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal). The data is also analyzed quantitatively. Where two different fluorophores are excited and measured simultaneously, the data are first coπected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using the emission spectrum for each fluorophore. A grid is superimposed over the fluorescence signal image such that the signal from each spot is centered in each element of the grid. The fluorescence signal within each element is then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis is the GEMTOOLS program (Incyte Genomics).
VIII Electronic Analysis
BLAST was used to search for identical or related molecules in the GenBank or LEFESEQ databases (Incyte Genomics). The product score for human and rat sequences was calculated as follows: the BLAST score is multiphed by the % nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences), such that a 100% aUgnment over the length of the shorter sequence gives a product score of 100. The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match wiU be exact within a 1% to 2% eπor, and with a product score of at least 70, the match wiU be exact. Similar or related molecules are usuaUy identified by selecting those which show product scores between 8 and 40.
Electronic northern analysis was performed at a product score of 70 and is shown in Tables 1 and 2. AU sequences and cDNA Ubraries in the LEFESEQ database were categorized by system, organ/tissue and ceU type. The categories included cardiovascular system, connective tissue, digestive system, embryonic structures, endocrine system, exocrine glands, female and male genitaUa, germ ceUs, hemic/immune system, Uver, musculoskeletal system, nervous system, pancreas, respiratory system, sense organs, skin, stomatognathic system, unclassified/mixed, and the urinary tract. For each category, the number of Ubraries in which the sequence was expressed were counted and shown over the total number of Ubraries in that category. In a non-normaUzed Ubrary, expression levels of two or more are significant.
IX Complementary Molecules
Molecules complementary to the cDNA, from about 5 (PNA) to about 5000 bp (complement of a cDNA insert), are used to detect or inhibit gene expression. Detection is described in Example VQ. To inhibit transcription by preventing promoter binding, the complementary molecule is designed to bind to the most unique 5' sequence and includes nucleotides of the 5' UTR upstream of the initiation codon of the open reading frame. Complementary molecules include genomic sequences (such as enhancers or introns) and are used in "triple heUx" base pairing to compromise the abiUty of the double heUx to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. To inhibit translation, a complementary molecule is designed to prevent ribosomal binding to the mRNA encoding the protein.
Complementary molecules are placed in expression vectors and used to transform a cell line to test efficacy; into an organ, tumor, synovial cavity, or the vascular system for transient or short term therapy; or into a stem ceU, zygote, or other reproducing lineage for long term or stable gene therapy.
Transient expression lasts for a month or more with a non-repUcating vector and for three months or more if appropriate elements for inducing vector repUcation are used in the transformation/expression system.
Stable transformation of appropriate dividing cells with a vector encoding the complementary molecule produces a transgenic ceU line, tissue, or organism (USPN 4,736,866). Those ceUs that assimilate and repUcate sufficient quantities of the vector to aUow stable integration also produce enough complementary molecules to compromise or entirely eliminate activity of the cDNA encoding the protein.
X Expression of RRP Expression and purification of the protein are achieved using either a mammaUan ceU expression system or an insect cell expression system. The pUB6/V5-His vector system (Invitrogen, Carlsbad CA) is used to express RRP in CHO cells. The vector contains the selectable bsd gene, multiple cloning sites, the promoter/enhancer sequence from the human ubiquitin C gene, a C-terminal V5 epitope for antibody detection with anti-V5 antibodies, and a C-terminal polyhistidine (6xHis) sequence for rapid purification on PROBOND resin (Invitrogen). Transformed ceUs are selected on media containing blasticidin.
Spodoptera frugiperda (Sf9) insect ceUs are infected with recombinant Autographica caUfornica nuclear polyhedrosis virus (baculovirus). The polyhedrin gene is replaced with the cDNA by homologous recombination and the polyhedrin promoter drives cDNA transcription. The protein is synthesized as a fusion protein with 6xhis which enables purification as described above. Purified protein is used in the foUowing activity and to make antibodies
XI Production of Antibodies
RRP is purified using polyacrylamide gel electrophoresis and used to immunize mice or rabbits. Antibodies are produced using the protocols below. Alternatively, the amino acid sequence of RRP is analyzed using LASERGENE software (DNASTAR) to determine regions of high antigenicity. An antigenic epitope, usuaUy found near the C-terminus or in a hydrophiUc region is selected, synthesized, and used to raise antibodies. Typically, epitopes of about 15 residues in length are produced using an ABI 431 A peptide synthesizer (AppUed Biosystems) using Fmoc-chemistry and coupled to KLH (Sigma- Aldrich) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester to increase antigenicity.
Rabbits are immunized with the epitope-KLH complex in complete Freund's adjuvant. Immunizations are repeated at intervals thereafter in incomplete Freund's adjuvant. After a minimum of seven weeks for mouse or twelve weeks for rabbit, antisera are drawn and tested for antipeptide activity. Testing involves binding the peptide to plastic, blocking with 1% bovine serum albumin, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG. Methods weU known in the art are used to determine antibody titer and the amount of complex formation.
XII Purification of Naturally Occurring Protein Using Specific Antibodies
NaturaUy occurring or recombinant protein is purified by immunoaffinity chromatography using antibodies which specifically bind the protein. An immunoaffinity column is constructed by covalently coupUng 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 aUow 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 coUected.
XIII Screening Molecules for Specific Binding with the cDNA or Protein
The cDNA, or fragments thereof, or the protein, or portions thereof, are labeled with 32P- dCTP, Cy3-dCTP, or Cy5-dCTP (APB), or with BIODIPY or FTTC (Molecular Probes, Eugene OR), respectively. Libraries of candidate molecules or compounds previously aπanged on a substrate are incubated in the presence of labeled cDNA or protein. After incubation under conditions for either a nucleic acid or amino acid sequence, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed, and the Ugand is identified. 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. XIV Two-Hybrid Screen
A yeast two-hybrid system, MATCHMAKER LexA Two-Hybrid system (Clontech Laboratories, Palo Alto CA), is used to screen for peptides that bind the protein of the invention. A cDNA encoding the protein is inserted into the multiple cloning site of a pLexA vector, Ugated, and transformed into E. coU. cDNA, prepared from mRNA, is inserted into the multiple cloning site of a pB42AD vector, Ugated, and transformed into E. coU to construct a cDNA Ubrary. The pLexA plasmid and pB42AD-cDNA Ubrary constructs are isolated from E. coU and used in a 2:1 ratio to co- transform competent yeast EGY48[p8op-lacZ] ceUs using a polyethylene glycol/Uthium acetate protocol. Transformed yeast ceUs are plated on synthetic dropout (SD) media lacking histidine (-His), tryptophan (-Trp), and uracil (-Ura), and incubated at 30C until the colonies have grown up and are counted. The colonies are pooled in a minimal volume of lx TE (pH 7.5), replated on SD/-His/-Leu/- TrpAUra media supplemented with 2% galactose (Gal), 1% raffinose (Raf), and 80 mg/ml 5-bromo-4- chloro-3-indolyl β-d-galactopyranoside (X-Gal), and subsequently examined for growth of blue colonies. Interaction between expressed protein and cDNA fusion proteins activates expression of a LEU2 reporter gene in EGY48 and produces colony growth on media lacking leucine (-Leu).
Interaction also activates expression of β-galactosidase from the p8op-lacZ reporter construct that produces blue color in colonies grown on X-Gal.
Positive interactions between expressed protein and cDNA fusion proteins are verified by isolating individual positive colonies and growing them in SD/-Trp/-Ura Uquid medium for 1 to 2 days at 30C. A sample of the culture is plated on SD/-Trp/-Ura media and incubated at 30C until colonies appear. The sample is repUca-plated on SD/-Trp/-Ura and SD/-His/-Trp/-Ura plates. Colonies that grow on SD containing histidine but not on media lacking histidine have lost the pI_«xA plasmid. Histidine-requiring colonies are grown on SD/Gal/Raf/X-Gal/-Trp/-Ura, and white colonies are isolated and propagated. The pB42AD-cDNA plasmid, which contains a cDNA encoding a protein that physicaUy interacts with the protein, is isolated from the yeast ceUs and characterized.
XV RRP Assay
RRP activity is deteπnined in assays for PKR binding. RRP is labeled with 125I Bolton-Hunter reagent (Bolton and Hunter (1973) Biochem J 133:529-539). PKR, previously aπayed in the weUs of a multi-weU plate, is incubated with the 125I-labeled RRP, washed, and any weUs with labeled RRP-PKR complex are assayed. Data obtained using different concentrations of RRP are used to calculate values for the number, affinity, and association of RRP with PKR.
AU patents and pubUcations mentioned in the specification are incorporated by reference herein. Various modifications and variations of the described method and system of the invention wiU 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 prefeπed 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 skiUed in the field of molecular biology or related fields are intended to be within the scope of the following claims. Clone Abs Pet
Tissue Category Count Found in Abund Abund
Cardiovascular System 266190 5/68 6 0.0023
Connective Tissue 144645 5/47 7 0.0048
Digestive System 501101 13/148 15 0.0030
Embryonic Structures 106713 4/21 6 0.0056
Endocrine System 225386 9/53 13 0.0058
Exocrine Glands 254635 9/64 12 0.0047
Reproductive, Female 427284 17/106 22 0.0051
Reproductive, Male 448207 15/114 27 0.0060
Germ Cells 38282 2/5 3 0.0078
Hemic and Immune System 680277 10/159 20 0.0029
Liver 109378 1/35 1 0.0009
Musculoskeletal System 159280 7/47 11 0.0069
Nervous System 955753 36/198 46 0.0048
Pancreas 110207 2/24 2 0.0018
Respiratory System 390086 11/93 13 0.0033
Sense Organs 19256 0/8 0 0.0000
Skin 72292 2/15 3 0.0041
Stomatognathic System 12923 0/10 0 0.0000
Unclassified/Mixed 120926 3/13 7 0.0058
Urinary Tract 279062 15/64 23 0.0082
Totals 5321883 166/1292 237 0.0000
TABLE 1
TABLE 2
Found in:
Clone Abs Pet
Library ID Count Library Description Abund Abund
CONFTUP01 1234 adipose tumor, lipoSAR, 3' CGAP 1 0.0810
CONUTUT01 7670 mesentery tumor, sigmoid, mets mixed-mullerian tumor, 61F 2 0.0261
CONNTUT04 4208 soft tissue tumor, spinal schwannoma, 35M 1 0.0238
GBLATUT01 4137 gallbladder tumor, squamous cell CA, 78F 2 0.0483
COLNCRT01 2271 colon, Crohn's, mw/benign carcinoid, 40M, m/COLNNOT05 1 0.0440
PROETMP01 3287 prostate, epithelium, mw/cancer, PIN, 45M, m/PROETUP02, CGAP 1 0.0304
PROSNOT28 3822 prostate, AH, mw/adenoCA, 55M, m/PROSTUT16 1 0.0262
PROSTMT03 3821 prostate, mw/adenoCA, 68M, m/PROSTUT18 1 0.0262
BRAFTUE03 1638 brain tumor, frontal, astrocytoma, 40F, 5RP, m/BRAINOT14 1 0.0611
LUNLTUT04 2810 lung tumor, squamous cell CA, 65F 1 0.0356
NOSETUE01 3483 nasal/cribriform tumor, olfactory neuroblastoma, 45M, 5RP 1 0.0287
LUNGTUT09 3972 lung tumor, squamous cell CA, 68M 1 0.0252
LUNGTUT08 5651 lung tumor, adenoCA, 63M 1 0.0177
LUNGTUT03 6268 lung tumor, squamous cell CA, 69M, m/LUNGNOTl5 1 0.0160
BLADTUT05 3734 bladder tumor, TC CA, 66M, m/BLADNOT06 4 0.1071
Not found in:
Clone
Library ID Count Library Description
COLNNO 05 3560 colon, sigmoid, mw/Crohn's, carcinoid, 40M, m/COLNCRTOl
PROETUP02 3615 prostate tumor, cancer, 45M, m/PROETMP01/02, CGAP
PROSTUT16 3943 prostate tumor, adenoCA, M, m/PROSNOT28/PROSTMC01/PROSTMT05
PROSTUT18 2201 prostate tumor, adenoCA, 68M, m/PROSTMT03
BRAINOT14 5199 brain, frontal, mw/astrocytoma, 40F, m/BRAITUT12/ BRAFTUE03
LUNGNOT15 3545 lung, pneumonitis, mw/squamous cell CA, 69M, m/LUNGTUT03
BLADNOT06 3739 bladder, mw/TC CA, aw/prostate TC CA, 66M, m/BLADTUT05

Claims

What is claimed is:
1. An isolated cDNA comprising a nucleic acid sequence encoding a protein having the amino acid sequence of SEQ ED NO:l, or the complement thereof.
2. An isolated cDNA comprising a nucleic acid sequence selected from: a) SEQ ED NO:2 or the complement thereof; b) a fragment of SEQ ED NO:2 selected from SEQ ID NOs:3-12 or the complement thereof; and c) a variant of SEQ ED NO:2 selected from SEQ ED NOs:13-15 or the complement thereof.
3. A composition comprising the cDNA or the complement of the cDNA of claim 1 and a labeling moiety.
4. A vector comprising the cDNA of claim 1.
5. A host ceU comprising the vector of claim 4.
6. A method for using a cDNA to produce a protein, the method comprising: a) culturing the host ceU of claim 5 under conditions for protein expression; and b) recovering the protein from the host ceU culture.
7. A method for using a cDNA to detect expression of a nucleic acid in a sample comprising: a) hybridizing the composition of claim 3 to nucleic acids of the sample, thereby forming hybridization complexes; and b) comparing hybridization complex formation with a standard, wherein the comparison indicates expression of the cDNA in the sample.
8. The method of claim 7 further comprising ampUfying the nucleic acids of the sample prior to hybridization.
9. The method of claim 7 wherein the composition is attached to a substrate.
10. The method of claim 7 wherein the cDNA is differentially expressed when compared with a standard and is diagnostic of a cancer, particularly Uposarcoma, mesentery mixed-muUerian tumor, spinal schwannoma, gaUbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer.
11. A method of using a cDNA to screen a pluraUty of molecules or compounds, the method comprising: a) combining the cDNA of claim 1 with a pluraUty of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding, thereby identifying a molecule or compound which specifically binds the cDNA.
12. The method of claim 11 wherein the molecules or compounds are selected from DNA molecules, RNA molecules, peptide nucleic acids, artificial chromosome constructions, peptides, transcription factors, repressors, and regulatory molecules.
13. A purified protein or a portion thereof produced by the method of claim 6 and selected from: a) an amino acid sequence of SEQ ED NO:l; b) an antigenic epitope of SEQ ED NO:l; and c) a biologicaUy active portion of SEQ ED NO: 1.
14. A composition comprising the protein of claim 13 and a pharmaceutical caπier.
15. A method for using a protein to screen a pluraUty of molecules or compounds to identify at least one Ugand, the method comprising: a) combining the protein of claim 13 with the molecules or compounds under conditions to allow specific binding; and b) detecting specific binding, thereby identifying a Ugand which specificaUy binds the protein.
16. The method of claim 15 wherein the molecules or compounds are selected from DNA molecules, RNA molecules, peptide nucleic acids, peptides, proteins, mimetics, agonists, antagonists, antibodies, immunoglobuUns, inhibitors, and drugs.
17. A method of using a protein to prepare and purify antibodies comprising: a) immunizing a animal with the protein of claim 15 under conditions to eUcit an antibody response; b) isolating animal antibodies; c) attaching the protein to a substrate; d) contacting the substrate with isolated antibodies under conditions to aUow specific binding to the protein; e) dissociating the antibodies from the protein, thereby obtaining purified antibodies.
18. An antibody produced by the method of claim 17.
19. A method for using an antibody to diagnose a condition or disease associated with expression of a protein, the method comprising: a) combining the antibody of claim 18 with a sample, thereby forming antibody:protein complexes; and b) comparing complex formation with a standard, wherein the comparison indicates expression of the protein in the sample.
20. The method of claim 19 wherein expression is diagnostic of cancer, particularly Uposarcoma, mesentery mixed-mullerian tumor, spinal schwannoma, gaUbladder cancer, prostate cancer, brain astrocytoma, lung cancer, and bladder cancer, and Crohn's disease.
21. A method for preparing a monoclonal antibody with the specificity of the antibody of claim 18 comprising: a) immunizing a animal with a protein of SEQ ED NO:l under conditions to eUcit an antibody response; b) isolating antibody-producing ceUs from the animal; c) fusing the antibody-producing ceUs with immortaUzed ceUs in culture to form monoclonal antibody producing hybridoma ceUs; d) culturing the hybridoma ceUs; and e) isolating monoclonal antibodies from culture.
22. A monoclonal antibody produced by the method of claim 21.
23. A method for using an antibody to immunopurify a protein comprising: a) attaching an antibody of claim 18 to a substrate, b) exposing the antibody to a sample containing protein under conditions to aUow antibody:protein complexes to form, c) dissociating the protein from the complex, and d) coUecting the purified protein.
24. A composition comprising an antibody of claim 18 and a labeling moiety.
25. A composition comprising an antibody of claim 18 and a pharmaceutical agent.
PCT/US2002/006179 2001-02-26 2002-02-26 Rax-related protein WO2002068621A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002248517A AU2002248517A1 (en) 2001-02-26 2002-02-26 Rax-related protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79384001A 2001-02-26 2001-02-26
US09/793,840 2001-02-26

Publications (2)

Publication Number Publication Date
WO2002068621A2 true WO2002068621A2 (en) 2002-09-06
WO2002068621A3 WO2002068621A3 (en) 2003-12-18

Family

ID=25160944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/006179 WO2002068621A2 (en) 2001-02-26 2002-02-26 Rax-related protein

Country Status (2)

Country Link
AU (1) AU2002248517A1 (en)
WO (1) WO2002068621A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5858675A (en) * 1997-05-13 1999-01-12 Incyte Pharmaceuticals, Inc. Double-stranded RNA-binding protein
WO1999015658A2 (en) * 1997-09-23 1999-04-01 Incyte Pharmaceuticals, Inc. Human regulatory molecules

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5858675A (en) * 1997-05-13 1999-01-12 Incyte Pharmaceuticals, Inc. Double-stranded RNA-binding protein
WO1999015658A2 (en) * 1997-09-23 1999-04-01 Incyte Pharmaceuticals, Inc. Human regulatory molecules
US6132973A (en) * 1997-09-23 2000-10-17 Incyte Pharmaceuticals, Inc. Human regulatory molecules

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE [Online] 13 June 1997 (1997-06-13) HILLIER ET AL: "WashU-Merck EST project 1997" retrieved from EBI, accession no. HS1256257 Database accession no. AA461277 XP002237671 *
DATABASE [Online] 14 December 2000 (2000-12-14) BONALDO ET AL: "Normalization and subtraction: two approaches to facilitate gene discovery" retrieved from EBI, accession no. BF549034 Database accession no. BF549034 XP002237673 *
DATABASE [Online] 2 February 2001 (2001-02-02) STRAUSBERG: "National Institutes of Health, Mammalian Gene Collection" retrieved from EBI, accession no. BG107958 Database accession no. BG107958 XP002237672 *
RUVOLO ET AL: "Ceramide regulates protein synthesis by a novel mechanism involving the cellular PKR activator RAX" JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 276, 8 January 2001 (2001-01-08), pages 11754-11758, XP002236908 *
WANG ET AL: "Large-scale identification, mapping, and genotyping of single-nucleotide polymorphisms in the human genome" SCIENCE, vol. 280, 1998, pages 1077-1082, XP002089398 ISSN: 0036-8075 *

Also Published As

Publication number Publication date
AU2002248517A1 (en) 2002-09-12
WO2002068621A3 (en) 2003-12-18

Similar Documents

Publication Publication Date Title
US20020187472A1 (en) Steap-related protein
US20020102569A1 (en) Diagnostic marker for cancers
US20140155288A1 (en) Methods for diagnosing cancer
US6566066B1 (en) Aquaporin-8 variant
US6485910B1 (en) Ras association domain containing protein
US6444430B1 (en) Ndr2-related proteins
US6590089B1 (en) RVP-1 variant differentially expressed in Crohn&#39;s disease
US20030211515A1 (en) Novel compounds
US20020123054A1 (en) Human angiopoietin
US6783955B2 (en) Polynucleotides encoding human presenilin variant
US6632617B1 (en) Tumor-associated antigen
WO2002068621A2 (en) Rax-related protein
US20030175787A1 (en) Vesicle membrane proteins
US20020025555A1 (en) GPCR diagnostic for brain cancer
US20020127636A1 (en) Ankyrin repeat domain 2 protein variant
US20020137166A1 (en) ASIP-related proteins
US20030166041A1 (en) TIMM8b-related protein
US20030166501A1 (en) Mucin-related tumor marker
EP1255834A2 (en) Gtpase activating proteins
US20040110937A1 (en) Xin-related proteins
WO2002024742A2 (en) Atp-binding cassette protein
US20020132238A1 (en) Progesterone receptor complex p23-like protein
US20020137038A1 (en) Intestinal proteins
WO2002046420A2 (en) Nebulin-related protein
EP1311665A2 (en) Kidney-specific protein

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP