US20030181706A1 - Inflammation-associated polynucleotides - Google Patents

Inflammation-associated polynucleotides Download PDF

Info

Publication number
US20030181706A1
US20030181706A1 US10/440,366 US44036603A US2003181706A1 US 20030181706 A1 US20030181706 A1 US 20030181706A1 US 44036603 A US44036603 A US 44036603A US 2003181706 A1 US2003181706 A1 US 2003181706A1
Authority
US
United States
Prior art keywords
polypeptide
polynucleotide
antibody
seq
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/440,366
Inventor
Michael Walker
Wayne Volkmuth
Tod Klingler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Corp
Original Assignee
Incyte Corp
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 Corp filed Critical Incyte Corp
Priority to US10/440,366 priority Critical patent/US20030181706A1/en
Publication of US20030181706A1 publication Critical patent/US20030181706A1/en
Abandoned legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates to eleven isolated polynucleotides and their encoded proteins that are highly co-expressed with genes known to be diagnostic markers of inflammation and useful for diagnosis, prognosis, treatment of inflammatory disorders.
  • Inflammation is the body's immediate, general response to wounding or infection by a pathogen. There are many complex phenomena that occur during an inflammation response. Initiation of the complement cascade, leukocyte recruitment and leukocyte activation are three key events. In the complement cascade a set of serum proteins collectively called complement non-specifically coat foreign matter. The coating proceeds in a cascade of steps using particular subsets of factors called complement components. The coated particles are then phagocytosed by macrophages or neutrophils recruited to the inflammation site. Leukocyte recruitment of monocytes and neutrophils is mediated by cytokines, which are proteins secreted by tissue at the inflammation site.
  • Interleukin-8 is the primary chemoattractant cytokine responsible for recruitment in the initial stage of inflammation.
  • monocytes and neutrophils are activated.
  • An immediate response to activation is the expression of L-selectin and the integrins.
  • L-selectin is a surface molecule that facilitates leukocyte binding (with relatively low affinity) to the endothelial cells lining blood vessels in the vicinity of the inflammation site.
  • the integrins also cell surface molecules, have stronger binding and mediate the actual extravasation of leukoctyes from the blood vessel.
  • receptors to the complement factors coating foreign particles are expressed on the leukocytes leading to phagocytosis and enzymatic degradation.
  • the present invention provides new compositions that are highly co-expressed with genes known to be diagnostic of inflammatory disorders and useful for diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders.
  • the invention provides for a plurality of polynucleotides having the nucleic acid sequences of SEQ ID NOs: 1-11 or the complements thereof that are co-expressed with genes such as CD16; L-selectin; Src-like adapter protein (SLAP); IP-30; superoxidase homoenzyme subunits, p67phox, p47phox, and p40phox; alpha-1-antitrypsin (AAT); Clq-A; 5-lipoxygenase activating protein (FLAP); and SRC family tyrosine kinase (HCK) known to be highly expressed during inflammation.
  • genes such as CD16; L-selectin; Src-like adapter protein (SLAP); IP-30; superoxidase homoenzyme subunits, p67phox, p47phox, and p40phox; alpha-1-antitrypsin (AAT); Clq-A; 5-lipoxygenase activating protein (
  • the invention also provides an isolated polynucleotide comprising a nucleic acid sequence selected from SEQ ID NOs: 1-11 or the complement thereof.
  • the polynucleotide is used as a surrogate marker, as a probe, in an expression vector, and in the diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders.
  • the invention further provides a composition comprising either a plurality of polynucleotides or a polynucleotide and a labeling moiety.
  • the invention provides a method for using a composition or a polynucleotide of the invention to screen a plurality of molecules and compounds to identify ligands which bind the polynucleotide(s).
  • the molecules are selected from DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, transcription factors, enhancers, and repressors.
  • the invention also provides a method of using a composition or a polynucleotide to purify a ligand.
  • the invention provides a method for using a composition or an isolated polynucleotide to detect gene expression in a sample by hybridizing the composition or polynucleotide to nucleic acids of the sample under conditions for formation of one or more hybridization complexes and detecting hybridization complex formation, wherein complex formation indicates gene expression in the sample.
  • the composition or polynucleotide is attached to a substrate.
  • the nucleic acids of the sample are amplified prior to hybridization.
  • complex formation is compared with at least one standard and indicates the presence of an inflammatory disorder.
  • the invention provides a purified protein or a portion thereof, SEQ ID NOs: 12-17, which is encoded by a polynucleotide that is co-expressed with genes that are diagnostic markers of inflammation or inflammatory disorders.
  • the invention also provides a method for using a protein to screen a plurality of molecules to identify at least one ligand which specifically binds the protein.
  • the molecules are selected from aptamers, DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, proteins, antibodies, agonists, antagonists, immunoglobulins, inhibitors, pharmaceutical agents or drug compounds.
  • the invention further provides a method of using a protein to purify a ligand.
  • the invention provides a method of using a protein to make an antibody that specifically binds to the protein of the invention, and methods for using the antibody to diagnose or treat an inflammatory disorder.
  • the invention further provides a composition comprising a polynucleotide, a protein, or an antibody that specifically binds a protein and a pharmaceutical carrier.
  • Sequence Listing provides exemplary polynucleotides comprising the nucleic acid sequences of SEQ ID NOs: 1-1I some of which encode the proteins comprising the amino acid sequences of SEQ ID NOs: 12-17. Each sequence is identified by a sequence identification number (SEQ ID NO) and by the Incyte clone number with which the sequence was first identified.
  • Markers for inflammatory disorders refers to polynucleotides, proteins, and antibodies which are useful in the diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. Typically, this means that the marker gene is differentially expressed in samples from subjects predisposed to, manifesting, or diagnosed with an inflammatory disorder.
  • “Differential expression” refers to an increased or upregulated or a decreased or downregulated expression as detected by presence, absence or at least about a two-fold change in the amount of transcribed messenger RNA or protein in a sample.
  • “Inflammatory disorders” specifically include, but are not limited to, the following conditions, diseases, and disorders: adult respiratory distress syndrome, allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes mellitus, emphysema, gastritis, hypereosinophilia, irritable bowel syndrome, lung cancer of complications thereof, lymphoma, meningitis, multiple sclerosis, osteoarthritis, psoriasis, rheumatoid arthritis, and toxic shock syndrome.
  • adult respiratory distress syndrome allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes mellitus, emphysema, gastritis, hypereos
  • isolated or purified refers to a polynucleotide or protein that is removed from its natural environment and that is separated from other components with which it is naturally present.
  • Genes known to be highly expressed during inflammation included CD16; L-selectin; Src-like adapter protein (SLAP); IP-30; superoxidase homoenzyme subunits, p67phox, p47phox, and p40phox; alpha-1-antitrypsin (AAT); Clq-A; 5-lipoxygenase activating protein (FLAP); and SRC family tyrosine kinase (HCK).
  • Polynucleotide refers to an isolated cDNA. It can be of genomic or synthetic origin, double-stranded or single-stranded, and combined with vitamins, minerals, carbohydrates, lipids, proteins, or other nucleic acids to perform a particular activity or form a useful composition.
  • Protein refers to a purified polypeptide whether naturally occurring or synthetic.
  • sample is used in its broadest sense.
  • a sample containing nucleic acids can comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; genomic DNA, RNA, or cDNA in solution or bound to a substrate; a cell; a tissue; a tissue print; and the like.
  • Substrate refers to any rigid or semi-rigid support to which polynucleotides 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.
  • a “transcript image” is a profile of gene transcription activity in a particular tissue at a particular time.
  • a “variant” refers to a polynucleotide or protein whose sequence diverges from about 5% to about 30% from the nucleic acid or amino acid sequences of the Sequence Listing.
  • the present invention employed “guilt by association or GBA”, a method for using marker genes known to be associated with a particular condition, disease or disorder to identify surrogate markers, polynucleotides and their encoded proteins, that are similarly associated or co-expressed in the same condition, disease, or disorder (Walker and Volkmuth (1999) Prediction of gene function by genome-scale expression analysis: prostate-associated genes. Genome Res 9:1198-1203, incorporated herein by reference).
  • the method identifies cDNAs cloned from mRNA transcripts which are active in tissues known to have been removed from subjects with inflammatory disorders.
  • the polynucleotides are useful for diagnosis, prognosis, evaluation of therapies, and treatment of inflammatory disorders.
  • Guilt by association provides for the identification of polynucleotides that are expressed in a plurality of libraries.
  • the polynucleotides represent genes of unknown function which are expressed in a specific signaling pathway, disease process, subcellular compartment, cell type, tissue, or species.
  • the expression patterns of the genes known to be highly expressed during inflammation, CD16, L-selectin, SLAP, IP-30, p67phox, p47phox, p40phox, AAT, Clq-A; FLAP, and HCK are compared with those of polynucleotides with unknown function to determine whether a specified co-expression probability threshold is met. Through this comparison, a subset of the polynucleotides having a high co-expression probability with the known marker genes can be identified.
  • the polynucleotides originate from human cDNA libraries. These polynucleotides can also be selected from a variety of sequence types including, but not limited to, expressed sequence tags (ESTs), assembled polynucleotides, full length coding regions, and 3′ untranslated regions. To be considered in the co-expression analysis, the polynucleotides have been expressed in at least five cDNA libraries. In this application, GBA was applied to a total of 41,419 assembled gene bins to identify eleven novel polynucleotides.
  • ESTs expressed sequence tags
  • assembled polynucleotides full length coding regions
  • 3′ untranslated regions To be considered in the co-expression analysis, the polynucleotides have been expressed in at least five cDNA libraries. In this application, GBA was applied to a total of 41,419 assembled gene bins to identify eleven novel polynucleotides.
  • the cDNA libraries used in the co-expression analysis were obtained from adrenal gland, biliary tract, bladder, blood cells, blood vessels, bone marrow, brain, bronchus, cartilage, chromaffin system, colon, connective tissue, cultured cells, embryonic stem cells, endocrine glands, epithelium, esophagus, fetus, ganglia, heart, hypothalamus, hemic/immune system, intestine, islets of Langerhans, kidney, larynx, liver, lung, lymph, muscles, neurons, ovary, pancreas, penis, phagocytes, pituitary, placenta, pleura, prostate, salivary glands, seminal vesicles, skeleton, spleen, stomach, testis, thymus, tongue, ureter, uterus, and the like.
  • the number of cDNA libraries selected can range from as few as three to greater than 10,000 and preferably, the number of the number of
  • the polynucleotides are assembled from related sequences, such as sequence fragments derived from a single transcript. Assembly of the polynucleotide can be performed using sequences of various types including, but not limited to, ESTs, extension of the EST, shotgun sequences from a cloned insert, or full length cDNAs. In a most preferred embodiment, the polynucleotides are derived from human sequences that have been assembled using the algorithm disclosed in U.S. Ser. No. 9,276,534, filed Mar. 25, 1999, and used in U.S. Ser. No. 09/195,292, filed 18 November 1998, both incorporated herein by reference.
  • differential expression of the polynucleotides can be evaluated by methods including, but not limited to, differential display by spatial immobilization or by gel electrophoresis, genome mismatch scanning, representational difference analysis, and transcript imaging.
  • the results of transcript imaging for SEQ ID NOs: 1, 2, 3, and 6 shows significant expression of these sequences in BPH, hypereosinophilia, lung cancer, and rheumatoid arthritis, respectively.
  • the transcript images provided direct confirmation of the strength of co-expression analysis using known genes to identify unknown polynucleotides and their encoded proteins which are highly significantly associated with inflammatory disorders. Additionally, differential expression can be assessed by microarray technology. These methods can be used alone or in combination.
  • Genes known to be highly expressed during inflammation can be selected based on research in which the genes are found to be key elements of immune response pathways or on the known use of the genes as diagnostic or prognostic markers or therapeutic targets for inflammatory disorders.
  • the known genes are CD 16, L-selectin, SLAP, IP-30, p67phox, p47phox, p40phox, AAT, Clq-A, FLAP, and HCK.
  • the procedure for identifying novel polynucleotides that exhibit a statistically significant co-expression pattern with known genes is as follows. First, the presence or absence of a polynucleotide in a cDNA library is defined: a polynucleotide is present in a cDNA library when at least one cDNA fragment corresponding to the polynucleotide is detected in a cDNA from that library, and a polynucleotide is absent from a library when no corresponding cDNA fragment is detected.
  • the significance of co-expression is evaluated using a probability method to measure a due-to-chance probability of the co-expression.
  • the probability method can be the Fisher exact test, the chi-squared test, or the kappa test. These tests and examples of their applications are well known in the art and can be found in standard statistics texts (Agresti (1990) Categorical Data Analysis, John Wiley & Sons, New York N.Y.; Rice (1988) Mathematical Statistics and Data Analysis, Duxbury Press, Pacific Grove Calif.).
  • a Bonferroni correction (Rice, supra, p. 384) can also be applied in combination with one of the probability methods for correcting statistical results of one polynucleotide versus multiple other polynucleotides.
  • the due-to-chance probability is measured by a Fisher exact test, and the threshold of the due-to-chance probability is set preferably to less than 0.001, more preferably to less than 0.00001.
  • occurrence data vectors can be generated as illustrated in the table below. The presence of a gene occurring at least once in a library is indicated by a one, and its absence from the library, by a zero.
  • Library 1 Library 2 Library 3 ... Library N Gene A 1 1 0 ... 0 Gene B 1 0 1 ... 0
  • the second table summarizes and presents: 1) the number of times gene A and B are both present in a library; 2) the number of times gene A and B are both absent in a library; 3) the number of times gene A is present, and gene B is absent; and 4) the number of times gene B is present, and gene A is absent.
  • the upper left entry is the number of times the two genes co-occur in a library, and the middle right entry is the number of times neither gene occurs in a library.
  • the off diagonal entries are the number of times one gene occurs, and the other does not.
  • Both A and B are present eight times and absent 18 times. Gene A is present, and gene B is absent, two times; and gene B is present, and gene A is absent, two times.
  • the probability (“p-value”) that the above association occurs due to chance as calculated using a Fisher exact test is 0.0003. Associations are generally considered significant if a p-value is less than 0.01 (Agresti, supra; Rice, supra).
  • This method of estimating the probability for co-expression makes several assumptions. The method assumes that the libraries are independent and are identically sampled. However, in practical situations, the selected cDNA libraries are not entirely independent, because more than one library can be obtained from a single subject or tissue. Nor are they entirely identically sampled, because different numbers of cDNAs can have been sequenced from each library. The number of cDNAs sequenced typically ranges from 5,000 to 10,000 cDNAs per library. After the Fisher exact co-expression probability is calculated for each polynucleotide versus all other assembled polynucleotides that occur, a Bonferroni correction for multiple statistical tests is applied.
  • polynucleotides SEQ ID NOs: 1-11 and their encoded proteins, SEQ ID NOs: 12-17, that exhibit highly significant co-expression probability with known marker genes for inflammatory disorders.
  • the results presented in Example VI show the direct (known gene to unknown polynucleotide) or indirect (known gene to unknown polynucleotide to a second unknown polynucleotide) associations among the novel polynucleotides and the known marker genes for inflammatory disorders. Therefore, by these associations, the novel polynucleotides are useful as surrogate markers for the co-expressed known marker genes in diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. Further, the proteins or peptides expressed from the novel polynucleotides are either potential therapeutics or targets for the identification and/or development of therapeutics.
  • the present invention encompasses a composition comprising a plurality of polynucleotides having the nucleic acid sequences of SEQ ID NOs: 1-11 or the complements thereof. These eleven polynucleotides are shown by the method of the present invention to have significant co-expression with known genes associated with inflammatory disorders.
  • the invention also provides a polynucleotide, its complement, a probe comprising the polynucleotide or the complement thereof selected from SEQ ID NOs: 1-11 and variants of the polynucleotides.
  • the polynucleotide can be used to search against the GenBank primate (pri), rodent (rod), mammalian (mam), vertebrate (vrtp), and eukaryote (eukp) databases; the encoded protein, against GenPept, SwissProt, BLOCKS (Bairoch et al. (1997) Nucleic Acids Res 25:217-221), PFAM, and other databases that contain previously identified and annotated protein sequences, motifs, and gene functions. Methods that search for primary sequence patterns with secondary structure gap penalties (Smith et al.
  • polynucleotides that are capable of hybridizing to SEQ ID NOs: 1-11, and fragments thereof, under highly stringent conditions.
  • Stringency can be defined by salt concentration, temperature, and other chemicals and conditions well known in the art. Conditions can be selected, for example, by varying the concentrations of salt in the prehybridization, hybridization, and wash solutions or by varying the hybridization and wash temperatures. With some substrates, the temperature can be decreased by adding a solvent such as formamide to the prehybridization and hybridization solutions.
  • Hybridization can be performed at low stringency, with buffers such as 5 ⁇ SSC (saline sodium citrate) with 1% sodium dodecyl sulfate (SDS) at 60° C., which permits complex formation between two nucleic acid sequences that contain some mismatches. Subsequent washes are performed at higher stringency with buffers such as 0.2 ⁇ SSC with 0.1% SDS at either 45° C. (medium stringency) or 68° C. (high stringency), to maintain hybridization of only those complexes that contain completely complementary sequences. Background signals can be reduced by the use of detergents such as SDS, sarcosyl, or TRITON X-100 (Sigma-Aldrich, St.
  • a polynucleotide can be extended utilizing primers and employing various PCR-based methods known in the art to detect upstream sequences such as promoters and other regulatory elements.
  • upstream sequences such as promoters and other regulatory elements.
  • PCR-based methods See, e.g., Dieffenbach and Dveksler (1995) PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview N.Y.).
  • kits such as XL-PCR (Applied Biosystems, Foster City Calif.), cDNA libraries (Life Technologies, Rockville Md.) or genomic libraries (Clontech, Palo Alto Calif.) and nested primers can be used to extend the sequence.
  • primers can be designed using commercially available software (LASERGENE software, DNASTAR, Madison Wis.) or another program, to be about 15 to 30 nucleotides in length, to have a GC content of about 50%, and to form a hybridization complex at temperatures of about 68° C. to 72° C.
  • the polynucleotide in another aspect of the invention, can be cloned into a recombinant vector that directs the expression of the protein, or structural or functional portions thereof, in host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode functionally equivalent amino acid sequence can be produced and used to express the protein encoded by the polynucleotide.
  • the nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter the nucleotide sequences for a variety of purposes including, but not limited to, modification of the cloning, processing, and/or expression of the gene product. DNA shuffling by random fragmentation, as described in U.S. Pat. No.
  • oligonucleotide-mediated site-directed mutagenesis can be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.
  • the polynucleotide or derivatives thereof can be inserted into an expression vector with elements for transcriptional and translational control of the inserted coding sequence in a particular host.
  • elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5′ and 3′ untranslated regions.
  • Methods which are well known to those skilled in the art can be used to construct such expression vectors. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination (Ausubel, supra, unit 16).
  • a variety of expression vector/host cell systems can be utilized to express the polynucleotide. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with baculovirus vectors; plant cell systems transformed with viral or bacterial expression vectors; or animal cell systems. For long term production of recombinant proteins in mammalian systems, stable expression in cell lines is preferred.
  • the polynucleotide can be transformed into cell lines using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable or visible marker gene on the same or on a separate vector.
  • expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable or visible marker gene on the same or on a separate vector.
  • the invention is not to be limited by the vector or host cell employed.
  • host cells that contain the polynucleotide and that express the protein can be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations, PCR amplification, and protein bioassay or immunoassay techniques which include membrane, solution, or chip-based technologies for the detection and/or quantification of nucleic acid or amino acid sequences. Immunological methods for detecting and measuring the expression of the protein using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS).
  • ELISAs enzyme-linked immunosorbent assays
  • RIAs radioimmunoassays
  • FACS fluorescence activated cell sorting
  • Host cells transformed with the polynucleotide can be cultured under conditions for the expression and recovery of the protein from cell culture.
  • the protein produced by a transgenic cell can be secreted or retained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing the polynucleotide can be designed to contain signal sequences which direct secretion of the protein through a prokaryotic cell wall or eukaryotic cell membrane.
  • a host cell strain can be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion.
  • modifications of the protein include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
  • Post-translational processing which cleaves a “prepro” form of the protein can also be used to specify protein targeting, folding, and/or activity.
  • Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and W138) are available from the ATCC (Manassas Va.) and can be chosen to ensure the correct modification and processing of the expressed protein.
  • natural, modified, or recombinant polynucleotides are ligated to a heterologous sequence resulting in translation of a fusion protein containing heterologous protein moieties in any of the aforementioned host systems.
  • heterologous protein moieties facilitate purification of fusion proteins using commercially available affinity matrices.
  • moieties include, but are not limited to, glutathione S-transferase, maltose binding protein, thioredoxin, calmodulin binding peptide, 6-His, FLAG, c-myc, hemaglutinin, and monoclonal antibody epitopes.
  • the polynucleotides are synthesized using chemical or enzymatic methods well known in the art (Caruthers et al. (1980) Nucl Acids Symp Ser (7) 215-233; Ausubel, supra, units 10.4 and 10.16). Peptide synthesis can be performed using various solid-phase techniques (Roberge et al. (1995) Science 269:202-204), and machines such as the ABI 431A peptide synthesizer (Applied Biosystems) can be used to automate synthesis. If desired, the amino acid sequence can be altered during synthesis to produce a more stable variant for therapeutic use.
  • the polynucleotides can be used as surrogate markers in diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders including but not limited to adult respiratory distress syndrome, allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes mellitus, emphysema, gastritis, hypereosinophilia, irritable bowel syndrome, lung cancer of complications thereof, lymphoma, meningitis, multiple sclerosis, osteoarthritis, psoriasis, rheumatoid arthritis, and toxic shock syndrome.
  • inflammatory disorders including but not limited to adult respiratory distress syndrome, allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes
  • the polynucleotide can be used to screen a plurality or library of molecules and compounds for specific binding affinity.
  • the assay can be used to screen DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, or proteins including transcription factors, enhancers, repressors, and the like which regulate the activity of the polynucleotide in the biological system.
  • the assay involves providing a plurality of molecules and compounds, combining a polynucleotide or a composition of the invention with the plurality of molecules and compounds under conditions to allow specific binding, and detecting specific binding to identify at least one molecule or compound which specifically binds at least one polynucleotides of the invention.
  • the proteins, or portions thereof can be used to screen a plurality or library of molecules or compounds in any of a variety of screening assays to identify a ligand.
  • the protein employed in such screening can be free in solution, affixed to an abiotic substrate or expressed on the external, or a particular internal surface, of a bacterial, or other, cell. Specific binding between the protein and the ligand can be measured.
  • the assay can be used to screen aptamers, DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, proteins, antibodies, agonists, antagonists, immunoglobulins, inhibitors, pharmaceutical agents or drug compounds and the like, which specifically bind the protein.
  • One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in Burbaum et al. U.S. Pat. No. 5,876,946, incorporated herein by reference, which screens large numbers of molecules for enzyme inhibition or receptor binding.
  • the polynucleotides are used for diagnostic purposes to determine the differential expression of a gene in a sample.
  • the polynucleotide consists of complementary RNA and DNA molecules, branched nucleic acids, and/or PNAs.
  • the polynucleotides are used to detect and quantify gene expression in biopsied samples in which differential expression of the polynucleotide indicates the presence of a disorder.
  • the polynucleotide can be used to detect genetic polymorphisms associated with a disease. In a preferred embodiment, these polymorphisms are detected in an mRNA transcribed from an endogenous gene.
  • the polynucleotide is used as a probe. Specificity of the probe is determined by whether it is made from a unique region, a regulatory region, or from a region encoding a conserved motif. Both probe specificity and the stringency of the diagnostic hybridization or amplification will determine whether the probe identifies only naturally occurring, exactly complementary sequences, allelic variants, or related sequences. Probes designed to detect related sequences should preferably have at least 50% sequence identity to at least a fragment of a polynucleotide of the invention.
  • Methods for producing hybridization probes include the cloning of nucleic acid sequences into vectors for the production of RNA probes.
  • Such vectors are known in the art, are commercially available, and can be used to synthesize RNA probes in vitro by adding RNA polymerases and labeled nucleotides.
  • Probes can incorporate nucleotides labeled by a variety of reporter groups including, but not limited to, radionuclides such as 32p or 35S, enzymatic labels such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, fluorescent labels such as Cy3 and Cy5, and the like.
  • the labeled polynucleotides can be used in Southern or northern analysis, dot blot, or other membrane-based technologies, on chips or other substrates, and in PCR technologies. Hybridization probes are also useful in mapping the naturally occurring genomic sequence. Fluorescent in situ hybridization (FISH) can be correlated with other physical chromosome mapping techniques and genetic map data as described in Heinz-Ulrich et al. (In: Meyers, supra, pp. 965-968). In many cases, genomic context helps identify genes that are encode a particular protein family. (See, e.g., Kirschning et al. (1997) Genomics 46:416-25.)
  • the polynucleotide can be labeled using standard methods and added to a sample from a subject under conditions for the formation and detection of hybridization complexes. After incubation the sample is washed, and the signal associated with complex formation is quantitated and compared with at least one standard value. Standard values are derived from any control sample, typically one that is free of the suspect disorder and from diseased samples, preferably from samples each of which represents a single, specific and preferably, staged disorder. If the amount of signal in the subject sample is altered in comparison to the standard values, then differential expression in the biopsied sample indicates the presence of the disorder. Qualitative and quantitative methods for comparing complex formation in subject samples with previously established standards are well known in the art.
  • Such assays can also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or to monitor the treatment of an individual subject. Once the presence of disease is established and a treatment protocol is initiated, hybridization or amplification assays can be repeated on a regular basis to determine gene expression in the patient begins to approximate that which is observed in a healthy subject. The results obtained from successive assays can be used to show the efficacy of treatment over a period ranging from several hours, e.g. in the case of toxic shock, to many years ,e.g. in the case of osteoarthritis.
  • the polynucleotides can be used on a substrate such as a microarray to monitor gene expression, to identify splice variants, mutations, and polymorphisms. Information derived from analyses of expression patterns can be used to determine gene function, to understand the genetic basis of a disease, to diagnose a disorder, and to develop and monitor the activities of therapeutic agents used to treat a disorder. Microarrays can also be used to detect genetic diversity, single nucleotide polymorphisms, which may characterize a particular population, at the genomic level.
  • antibodies or Fabs comprising an antigen binding site that specifically binds the protein can be used for the diagnosis of diseases characterized by the differential expression of the protein.
  • a variety of protocols for measuring protein expression including ELISAs, RIAs, and FACS, are well known in the art and provide a basis for diagnosing altered or abnormal levels of expression.
  • Standard values for protein expression parallel those reviewed above for nucleotide expression.
  • the amount of complex formation can be quantitated by various methods, preferably by photometric means. Quantities of the protein expressed in subject samples are compared with standard values. Deviation between standard and subject values establishes the parameters for diagnosing or monitoring a particular disorder.
  • antibodies of the present invention can be used for treatment, delivery of therapeutics, or monitoring therapy for inflammatory disorders.
  • the polynucleotide, or its complement can be used therapeutically for the purpose of expressing mRNA and protein, or conversely to block transcription or translation of the mRNA.
  • Expression vectors can be constructed using elements from retroviruses, adenoviruses, herpes or vaccinia viruses, or bacterial plasmids, and the like. These vectors can be used for delivery of nucleotide sequences to a particular target cell population, tissue, or organ. Methods well known to those skilled in the art can be used to construct vectors to express the polynucleotides or their complements. (See, e.g., Maulik et al.
  • the polynucleotide or its complement can be used for somatic cell or stem cell gene therapy.
  • Vectors can be introduced in vivo, in vitro, and ex vivo.
  • vectors are introduced into stem cells taken from the subject, and the resulting transgenic cells are clonally propagated for autologous transplant back into that same subject.
  • Delivery of the polynucleotide by transfection, liposome injections, or polycationic amino polymers can be achieved using methods which are well known in the art. (See, e.g., Goldman et al.
  • endogenous gene expression can be inactivated using homologous recombination methods which insert an inactive gene sequence into the coding region or other targeted region of the genome (See, e.g. Thomas et al. (1987) Cell 51: 503-512.)
  • Vectors containing the polynucleotide can be transformed into a cell or tissue to express a missing protein or to replace a nonfunctional protein.
  • a vector constructed to express the complement of the polynucleotide can be transformed into a cell to downregulate protein expression.
  • Complementary or antisense sequences can consist of an oligonucleotide derived from the transcription initiation site; nucleotides between about positions ⁇ 10 and +10 from the ATG are preferred.
  • inhibition can be achieved using triple helix base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules.
  • Ribozymes enzymatic RNA molecules
  • Ribozymes can also be used to catalyze the cleavage of mRNA and decrease the levels of particular mRNAs, such as those comprising the polynucleotides of the invention.
  • Ribozymes can cleave mRNA at specific cleavage sites.
  • ribozymes can cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The construction and production of ribozymes is well known in the art and is described in Meyers (supra).
  • RNA molecules can be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends of the molecule, or the use of phosphorothioate or 2′O-methyl rather than phosphodiester linkages within the backbone of the molecule.
  • nontraditional bases such as inosine, queosine, and wybutosine, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases, can be included.
  • an antagonist, or an antibody that binds specifically to the protein can be administered to a subject to treat an inflammatory disorder.
  • the antagonist, antibody, or fragment can be used directly to inhibit the activity of the protein or indirectly to deliver a therapeutic agent to cells or tissues which express the protein.
  • the therapeutic agent can be a cytotoxic agent selected from a group including, but not limited to, abrin, ricin, doxorubicin, daunorubicin, taxol, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin D, diphteria toxin, Pseudomonas exotoxin A and 40, radioisotopes, and glucocorticoid.
  • a cytotoxic agent selected from a group including, but not limited to, abrin, ricin, doxorubicin, daunorubicin, taxol, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin D, diphteria toxin, Pseudom
  • Antibodies to the protein can be generated using methods that are well known in the art. Such antibodies can include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies, such as those which inhibit dimer formation, are especially preferred for therapeutic use. Monoclonal antibodies to the protein can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma, the human B-cell hybridoma, and the EBV-hybridoma techniques. In addition, techniques developed for the production of chimeric antibodies can be used.
  • an agonist of the protein can be administered to a subject to treat or prevent a disease associated with decreased expression, longevity or activity of the protein.
  • An additional aspect of the invention relates to the administration of a pharmaceutical or sterile composition, in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic applications discussed above.
  • Such pharmaceutical compositions can consist of the protein or antibodies, mimetics, agonists, antagonists, or inhibitors of the protein.
  • the compositions can be administered alone or in combination with at least one other agent, such as a stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier including, but not limited to, saline, buffered saline, dextrose, and water.
  • the compositions can be administered to a subject alone or in combination with other agents, drugs, or hormones.
  • compositions utilized in this invention can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
  • these pharmaceutical compositions can contain pharmaceutically-acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration can be found in the latest edition of Remington's Pharmaceutical Sciences (Mack Publishing, Easton Pa.).
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, or pigs.
  • animal models such as mice, rats, rabbits, dogs, or pigs.
  • An animal model can also be used to determine the concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • a therapeutically effective dose refers to that amount of active ingredient which ameliorates the symptoms or condition.
  • Therapeutic efficacy and toxicity can be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating and contrasting the ED 50 (the dose therapeutically effective in 50% of the population) and LD 50 (the dose lethal to 50% of the population) statistics. Any of the therapeutic compositions described above can be applied to any subject in need of such therapy, including, but not limited to, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans.
  • SEQ ID NOs: 1-11 can be useful in the differentiation of stem cells.
  • Eukaryotic stem cells are able to differentiate into the multiple cell types of various tissues and organs and to play roles in embryogenesis and adult tissue regeneration (Gearhart (1998) Science 282:1061-1062; Watt and Hogan (2000) Science 287:1427-1430).
  • stem cells can be totipotent with the potential to create every cell type in an organism and to generate a new organism, pluripotent with the potential to give rise to most cell types and tissues, but not a whole organism; or multipotent cells with the potential to differentiate into a limited number of cell types.
  • Stem cells can be transfected with polynucleotides which can be transiently expressed or can be integrated within the cell as transgenes.
  • Embryonic stem (ES) cell lines are derived from the inner cell masses of human blastocysts and are pluripotent (Thomson et al. (1998) Science 282:1145-1147). They have normal karyotypes and express high levels of telomerase which prevents senescence and allows the cells to replicate indefinitely. ES cells produce derivatives that give rise to embryonic epidermal, mesodermal and endodermal cells. Embryonic germ (EG) cell lines, which are produced from primordial germ cells isolated from gonadal ridges and mesenteries, also show stem cell behavior (Shamblott et al. (1998) Proc Natl Acad Sci 95:13726-13731). EG cells have normal karyotypes and appear to be pluripotent.
  • Organ-specific adult stem cells differentiate into the cell types of the tissues from which they were isolated. They maintain their original tissues by replacing cells destroyed from disease or injury.
  • Adult stem cells are multipotent and under proper stimulation can be used to generate cell types of various other tissues (Vogel (2000) Science 287:1418-1419).
  • Hematopoietic stem cells from bone marrow provide not only blood and immune cells, but can also be induced to transdifferentiate to form brain, liver, heart, skeletal muscle and smooth muscle cells.
  • mesenchymal stem cells can be used to produce bone marrow, cartilage, muscle cells, and some neuron-like cells, and stem cells from muscle have the ability to differentiate into muscle and blood cells (Jackson et al.
  • Neural stem cells which produce neurons and glia, can also be induced to differentiate into heart, muscle, liver, intestine, and blood cells (Kuhn and Svendsen (1999) BioEssays 21:625-630); Clarke et al. (2000) Science 288:1660-1663; Gage (2000) Science 287:1433-1438; and Galli et al. (2000) Nature Neurosci 3:986-991).
  • Neural stem cells can be used to treat neurological disorders such as Alzheimer disease, Parkinson disease, and multiple sclerosis and to repair tissue damaged by strokes and spinal cord injuries.
  • Hematopoietic stem cells can be used to restore immune function in immunodeficient patients or to treat autoimmune disorders by replacing autoreactive immune cells with normal cells to treat diseases such as multiple sclerosis, scleroderma, rheumatoid arthritis, and systemic lupus erythematosus.
  • Mesenchymal stem cells can be used to repair tendons or to regenerate cartilage to treat arthritis.
  • Liver stem cells can be used to repair liver damage.
  • Pancreatic stem cells can be used to replace islet cells to treat diabetes.
  • Muscle stem cells can be used to regenerate muscle to treat muscular dystrophies (Fontes and Thomson (1999) BMJ 319:1-3; Weissman (2000) Science 287:1442-1446 Marshall (2000) Science 287:1419-1421; Marmont (2000) Ann Rev Med 51:115-134).
  • the cDNA library was selected as an example to demonstrate the construction of the cDNA libraries from which the sequences used to identify genes associated with inflammatory disorders were derived.
  • the OVARTUT05 library was constructed from tumorous ovary tissue obtained from a 62 year-old Caucasian female. Pathology indicated a grade 4 endometrioid carcinoma with extensive squamous differentiation forming a solid mass in the right ovary. The cervix showed mild chronic cervicitis, and the posterior uterine serosa showed focal endometriosis. Prior pathology indicated weakly proliferative endometrium with excessive stromal breakdown in the uterus and a mild chronic cervicitis with prominent nabothian cyst in the cervix.
  • the frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments, Westbury N.Y.) in guanidinium isothiocyanate solution.
  • the lysate was centrifuged over a 5.7 M CsCl cushion using an SW28 rotor in a L8-70M ultracentrifuge (Beckman Coulter, Fullerton Calif.) 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 DNAse treated at 37C.
  • the RNA extraction was repeated.
  • the MRNA was isolated with the OLIGOTEX kit (Qiagen, Chatsworth Calif.) and used to construct the cDNA library.
  • the mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies).
  • the cDNAs were fractionated on a SEPHAROSE CL4B column (Amersham Pharmacia Biotech (APB), Piscataway N.J.), and those cDNAs exceeding 400 bp were ligated into pINCY plasmid (Incyte Genomics, Palo Alto Calif.).
  • the plasmid was then transformed into DH5 ⁇ competent cells (Life Technologies).
  • Plasmid DNA was released from the bacterial cells and purified using the REAL PREP 96 plasmid kit (Qiagen). This kit enabled the simultaneous purification of 96 samples in a 96-well block using multi-channel reagent dispensers.
  • the recommended protocol was employed except for the following changes: 1) the bacteria were cultured in 1 ml of sterile TERRIFIC BROTH (BD Biosciences, San Jose Calif.) with carbenicillin at 25 mg/L and glycerol at 0.4%; 2) after inoculation and incubation for 19 hours, the cells were lysed with 0.3 ml of lysis buffer; and 3) following isopropanol precipitation, the plasmid DNA pellet was resuspended in 0.1 ml of distilled water. After the last step in the protocol, samples were transferred to a 96-well block for storage at 4C.
  • the cDNAs were prepared using a MICROLAB 2200 system (Hamilton, Reno Nev.) in combination with DNA ENGINE thermal cyclers (MJ Research, Watertown Mass.).
  • the cDNAs were sequenced by the method of Sanger and Coulson (1975; J Mol Biol 94:441448) using ABI PRISM 377 DNA sequencing systems (Applied Biosystems). Most of the sequences were sequenced using standard ABI protocols and kits at solution volumes of 0.25x ⁇ 1.0x. In the alternative, some of the sequences were sequenced using solutions and dyes from APB.
  • sequences used for co-expression analysis were assembled from EST sequences, 5′ and 3′ long read sequences, and full length coding sequences. Of the 41,419 assembled sequences used in the analysis, each was expressed in at least five cDNA libraries.
  • Bins were annotated by screening the consensus sequence in each bin against public databases, such as GBpri and GenPept from NCBI.
  • the annotation process involved a FASTn screen against the GBpri database in GenBank. Those hits with a percent identity of greater than or equal to 75% and an alignment length of greater than or equal to 100 base pairs were recorded as homolog hits.
  • the residual unannotated sequences were screened by FASTx against GenPept. Those hits with an E value of less than or equal to 10 ⁇ 8 were recorded as homolog hits.
  • Sequences were then reclustered using BLASTn and Cross-Match, a program for rapid amino acid and nucleic acid sequence comparison and database search (Green, supra), sequentially. Any BLAST alignment between a sequence and a consensus sequence with a score greater than 150 was realigned using cross-match. The sequence was added to the bin whose consensus sequence gave the highest Smith-Waterman score (Smith et al. (1992) Protein Engineering 5:35-51) amongst local alignments with at least 82% identity. Non-matching sequences were moved into new bins, and assembly processes were repeated.
  • polynucleotides of the Sequence Listing or their encoded proteins 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 BLAST 2 (Altschul et al. supra; Altschul, supra) to produce alignments and to determine which sequences were exact matches or homologs. The alignments 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. All of the sequences disclosed in this application have lengths of at least 49 nucleotides, and no more than 12% uncalled bases (where N is recorded rather than A, C, G, or T).
  • BLAST matches between a query sequence and a database sequence were evaluated statistically 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 follows: the % nucleotide or amino acid identity [between the query and reference sequences] in BLAST is multiplied 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 electronic stringency for an exact match was set at 70, and the conservative lower limit for an exact match was set at approximately 40 (with 1-2% error due to uncalled bases).
  • the BLAST software suite includes various sequence analysis programs including “blastn” that is used to align nucleic acid molecules and BLAST 2 that is used for direct pairwise comparison of either nucleic or amino acid molecules.
  • 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.
  • polynucleotides of this application were compared with assembled consensus sequences or templates found in the LIFESEQ GOLD database.
  • Component sequences from cDNA, extension, full length, and shotgun sequencing projects were subjected to PHRED analysis and assigned a quality score. All sequences with an acceptable quality score were subjected to various pre-processing and editing pathways to remove low quality 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 quality score greater than or equal to 150 and an alignment 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 split. Templates were subjected to analysis by STITCHER/EXON MAPPER algorithms that analyze the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced genes across tissue types or disease states, and the like. Assembly procedures were repeated periodically, and templates were annotated using BLAST against GenBank databases such as GBpri.
  • templates were subjected to BLAST, motif, and other functional analyses and categorized in protein hierarchies using methods described in U.S. Ser. No. 08/812,290 and U.S. Ser. No. 08/811,758, both filed Mar. 6, 1997; in U.S. Ser. No. 08/947,845, filed Oct. 9, 1997; and in U.S. Ser. No. 09/034,807, filed Mar. 4, 1998.
  • templates were analyzed by translating each template in all three forward reading frames and searching each translation against the PFAM database of hidden Markov model-based protein families and domains using the HMMER software package (Washington University School of Medicine, St. Louis Mo.; http://pfam.wustl.edu/).
  • polynucleotide was further analyzed using MACDNASIS PRO software (Hitachi Software Engineering), and LASERGENE software (DNASTAR) and queried against public databases such as the GenBank rodent, mammalian, vertebrate, prokaryote, and eukaryote databases, SwissProt, BLOCKS, PRINTS, PFAM, and Prosite.
  • FLAP 5-lipoxygenase activating protein lipoxygenase enzyme catalyzes formation of leukotrienes which are potent inflammatory mediators.
  • FLAP is an anti- inflammatory therapeutic target (Byrum et al.
  • the method also identified polynucleotides from among a total of 41,419 assembled sequences that show strong association with the known genes. The process was reiterated until the number of polynucleotides was reduced to the final eleven polynucleotides shown below. The following tabular entries show the negative log of the p-value ( ⁇ log p) for the co-expression among the known marker genes and the novel polynucleotides.
  • the novel polynucleotides are identified in the table below by their Incyte clone numbers and the known genes their abbreviated names as shown in Example IV above.
  • the p-value is the probability that the observed co-expression is due to chance, using the Fisher Exact Test.
  • Gene or SEQ ⁇ Gene or clone L-sel- ID NO (clone) CD16 ectin SLAP IP-30 p67-phox AA p47-phox ClqA p40-phox FLAP HC 1221361 3055142 402234 CD16 L-selectin 4 SLAP 5 6 IP-30 5 3 4 P67-phox 3 5 7 13 AAT 6 2 4 10 5 P47-phox 5 6 5 14 7 3 ClqA 7 2 6 11 3 7 6 P40-phox 5 5 3 4 7 4 6 1 FLAP 7 5 3 11 5 3 5 7 3 HCK 9 8 4 5 8 5 7 3 6 3 4 (1221361) 3 5 7 4 3 1 4 2 4 2 4 10 (3055142) 6 5 9 4 5 3 8 1 4 3 8 4 1 (402234) 3 6 4 4 2 0 6 1 3 4 2 3 1 1
  • the highest co-expression value is obtained when the highest ( ⁇ log p) value found along the horizontal line following each SEQ ID NO (clone number) are correlated with a known marker gene (abbreviation along the top line of the chart).
  • a known marker gene abbreviation along the top line of the chart.
  • polynucleotides were found to be associated with known genes that are diagnostic markers for inflammation and inflammatory disorders.
  • the polynucleotides comprising the nucleic acid sequences of SEQ ID NOs:1-11 were first identified as Incyte Clones 402234, 569989, 706377, 1221361, 1335016, 2349263, 2471716, 2726173, 3054032, 3055142, and 3507924, respectively. These sequences were assembled according to the procedures described in Example IV. BLAST and motif searches were performed for SEQ ID NOs: 1-11 and SEQ ID NOs: 12-17 according to Example V. Proteins or peptides comprising the amino acid sequences of SEQ ID NOs: 12-17 were encoded by the nucleic acids of SEQ ID NO: 1, 2, 6, 7, 8, and 11, respectively.
  • SEQ ID NO:3 is 1229 nucleotides in length and shares about 99% sequence identity from about nucleotide 250 to about nucleotide 1216 with a human basement membrane-induced gene identified in a human endometrial adenocarcinoma cell line (g3132521).
  • SEQ ID NO:4 is about 1261 nucleotides in length and has about 34% sequence identity from nucleotide 23 to nucleotide 994 with a sequence similar to a RNA recognition motif (g2645068).
  • SEQ ID NO:5 is 1340 nucleotides in length and has about 60% sequence identity from about nucleotide 21 to about nucleotide 925 with a human prostaglandin transporter hPGT mRNA (g3006201).
  • the protein encoded by SEQ ID NO:5 exhibits several potential transmembrane domains identified by HMM analysis.
  • SEQ ID NO:9 is 2309 nucleotides in length and shows sequence identity from about nucleotide 104 to about nucleotide 785 with a human polycystic kidney disease-associated protein gene (g790818).
  • SEQ ID NO:12 is 127 amino acid residues in length and shows about 50% sequence identity from about residue 37 to about residue 106 with a tobacco LIM-domain-containing protein (g181464).
  • the LIM domain is a cysteine-rich, zinc-binding motif of about 60 amino-acid residues that plays a potential role in DNA binding and regulation (Perez-Alvarado et al. (1994) Nat Struct Biol 1:388-398).
  • PFAM analysis shows that residues 40 to 97 of SEQ ID NO: 12 align with and encompass the LIM domain.
  • SEQ ID NO: 13 is 93 amino acid residues in length and has a potential signal peptide from residue 1 to residue 18. SEQ ID NO: 13 also exhibits a potential transmembrane domain from about residues 47 to residue 69.
  • SEQ ID NO: 14 is 225 amino acid residues in length and has about 32% sequence identity from about residue 5 to about residue 135 with a mouse high affinity IgE receptor beta subunit (g309225).
  • SEQ ID NO: 15 is 547 amino acid residues in length and has about 35% sequence identity from about residue 413 to about 546 with a rat beta-chimaerin, a GTPase-activating protein expressed exclusively in the testis at the onset of sexual maturation (g203117).
  • PFAM analysis shows that SEQ ID NO: 15 has sequence homology from about residue 353 to about residue 523 with the GTPase-activator protein for Rho-like GTPases.
  • SEQ ID NO: 16 is 265 amino acid residues in length and shows about 93% sequence identity from about residue 39 to about residues 265 with Maxp1, a rat protein which interacts with Mss4, a guanine nucleotide exchange factor (g2459833), and about 91% sequence identity from about residue 38 to about residue 265 with Nore1, a mouse putative Ras effector that plays a role in transmitting growth and differentiation signals received from Ras proteins (g2997698).
  • PFAM analysis confirms that SEQ ID NO: 16 from about residue 119 to about residue 211 matches a Ras association domain which interacts directly with the Ras proteins.
  • SEQ ID NO: 17 is 394 amino acid residues in length and exhibits a potential signal peptide sequence from about residue 1 to residue 19 and a potential transmembrane domain from about residues 273 to residue 295.
  • the polynucleotides are applied to a substrate by one of the following methods.
  • a mixture of polynucleotides is fractionated by gel electrophoresis and transferred to a nylon membrane by capillary transfer.
  • the polynucleotides are individually ligated to a vector and inserted into bacterial host cells to form a library.
  • the polynucleotides are then arranged on a substrate by one of the following methods.
  • bacterial cells containing individual clones are robotically picked and arranged on a nylon membrane.
  • the membrane is placed on LB agar containing selective agent (carbenicillin, kanamycin, ampicillin, or chloramphenicol depending on the vector used) and incubated at 37C.
  • the membrane is removed from the agar and consecutively placed colony side up in 10% SDS, denaturing solution (1.5 M NaCl, 0.5 M NaOH), neutralizing solution (1.5 M NaCl, 1 M Tris-HCl, pH 8.0), and twice in 2 ⁇ SSC for 10 min each.
  • the membrane is then UV irradiated in a STRATALINKER UV-crosslinker (Stratagene).
  • polynucleotides are amplified from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR amplification increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 ⁇ g.
  • Amplified nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are arranged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobilized by denaturation, neutralization, and UV irradiation as described above.
  • Purified nucleic acids are robotically arranged and immobilized on polymer-coated glass slides using the procedure described in U.S. Pat. No. 5,807,522.
  • Polymer-coated slides are prepared by cleaning glass microscope slides (Corning, Acton Mass.) 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 slides are washed extensively with distilled water between and after treatments.
  • the nucleic acids are arranged on the slide and then immobilized by exposing the array to UV irradiation using a STRATALINKER UV-crosslinker (Stratagene). Arrays are then washed at room temperature in 0.2% SDS and rinsed three times in distilled water. Non-specific binding sites are blocked by incubation of arrays in 0.2% casein in phosphate buffered saline (PBS; Tropix, Bedford Mass.) for 30 min at 60C.; then the arrays are washed in 0.2% SDS and rinsed in distilled water as before.
  • PBS phosphate buffered saline
  • Hybridization probes derived from the polynucleotides of the Sequence Listing are employed for screening cDNAs, mRNAs, or genomic DNA in membrane-based hybridizations. Probes are prepared by diluting the polynucleotides 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 polynucleotide is then added to a REDIPRIME tube (APB), gently mixed until blue color is evenly distributed, and briefly centrifuged. Five ⁇ l of [ 32 P]dCTP is added to the tube, and the contents are incubated at 37C. for 10 min.
  • APB REDIPRIME tube
  • the labeling reaction is stopped by adding 5 ⁇ l 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.
  • Hybridization probes derived from mRNA isolated from samples are employed for screening polynucleotides of the Sequence Listing in array-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 5 ⁇ buffer, 1 ⁇ l 0.1 M DTT, 3 ⁇ l Cy3 or Cy5 labeling mix, 1 ⁇ l RNAse inhibitor, 1 ⁇ l reverse transcriptase, and 5 ⁇ l 1 ⁇ yeast control mRNAs.
  • Yeast control mRNAs are synthesized by in vitro transcription from noncoding yeast genomic DNA (W. Lei, unpublished).
  • 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.
  • 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.
  • probes are purified using two successive CHROMA SPIN+TE 30 columns (Clontech, Palo Alto Calif.). Purified probe is ethanol precipitated by diluting probe to 90 ⁇ l in DEPC-treated water, adding 2 ⁇ l 1 mg/ml glycogen, 60 ⁇ l 5 M sodium acetate, and 300 ⁇ l 100% ethanol. The probe is centrifuged for 20 min at 20,800 ⁇ g, and the pellet is resuspended in 12 ⁇ l 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 array-based hybridizations as described below.
  • Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and 1 ⁇ high 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.
  • the membrane is washed for 15 min at 25C. in 1 mM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 min each at 25C. in 1 mM Tris (pH 8.0).
  • XOMAT-AR film Eastman Kodak, Rochester N.Y.
  • XOMAT-AR film Eastman Kodak, Rochester N.Y.
  • Probe is heated to 65C. for five min, centrifuged five min at 9400 rpm in a 5415C microcentrifuge (Eppendorf Scientific, Westbury N.Y.), and then 18 ⁇ l are aliquoted onto the array surface and covered with a coverslip.
  • the arrays are transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide.
  • the chamber is kept at 100% humidity internally by the addition of 140 ⁇ l of 5 ⁇ SSC in a corner of the chamber.
  • the chamber containing the arrays is incubated for about 6.5 hr at 60C.
  • the arrays are washed for 10 min at 45C. in 1 ⁇ SSC, 0.1% SDS, and three times for 10 min each at 45C. in 0.1 ⁇ SSC, 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 correlates 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 array elements, and signals are examined under conditions in which the emissions from the two different labels are individually detectable. Elements on the array that are hybridized to 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 Calif.) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of Cy5.
  • the excitation laser light is focused on the array using a 20 ⁇ microscope objective (Nikon, Melville N.Y.).
  • the slide containing the array is placed on a computer-controlled X-Y stage on the microscope and raster-scanned past the objective with a resolution of 20 micrometers.
  • the two fluorophores are sequentially excited by the laser.
  • Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477, Hamamatsu Photonics Systems, Bridgewater N.J.) corresponding to the two fluorophores.
  • PMT R1477 Hamamatsu Photonics Systems, Bridgewater N.J.
  • Appropriate filters positioned between the array and the photomultiplier 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 calibrated using the signal intensity generated by the yeast control mRNAs added to the probe mix.
  • a specific location on the array contains a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000.
  • the output of the photomultiplier tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood Mass.) installed in an IBM-compatible PC computer.
  • the digitized data are displayed as an image where the signal intensity is mapped using a linear 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 corrected 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).
  • transcript images for SEQ ID NOs: 1, 2, 3, and 6 were performed using the LIFESEQ GOLD database (Incyte Genomics).
  • the product score for sequences was calculated as follows: the BLAST score is multiplied 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% alignment over the length of the shorter sequence gives a product score of 100.
  • the four transcript images below show independent confirmation of the results of the co-expression analysis.
  • the transcript images demonstrate differential expression of SEQ ID NOs: 1, 2, 3 and 6, each in a different category, as produced using the LIFESEQ GOLD database (Incyte Genomics).
  • SEQ ID NO:1 Category: Male Reproductive
  • Library cDNAs Description Abundance % Abundance PROSDIP01 487 prostate, stroma, BPH, M, 3′ CGAP 3 0.6160 PROSTUP02 869 prostate tumor, N, 3′ CGAP 1 0.1151 PROSTUT01 3224 prostate tumor, adenoCA, 50 M 1 0.0310 PROSNOT26 3695 prostate, mw/adenoCA, 65 M 1 0.0271
  • SEQ ID NO: I is differentially expressed in benign prostate hyperplasia. Expression exceeds that of any other prostate library, including tumor and cytologically normal tissue, by greater than five-fold.
  • SEQ ID NO:2 (Category: Hemic/Immune) Library cDNAs Description Abundance % Abundance EOSIHET02 9261 periph blood, hypereosinophilia, 48 M 2 0.0216 EOSITXT01 8976 periph blood, eosinophils, t/IL-5 1 0.0111
  • SEQ ID NO:2 is differentially expressed in hypereosinophilia, even exceeding IL-5 activated expression by approximately two-fold.
  • SEQ ID NO:3 (Category: Respiratory System) Library cDNAs Description Abundance % Abundance LUNGTUT13 3990 lung tumor, adenoCA, 47 M 2 0.0501 LUNGNOT38 3447 lung, asthma, 15 M 1 0.0290 LUNGNOT03 4959 lung, mw/mets thyroid CA, 79 M 1 0.0202 LUNLTMT01 6668 lung, mw/adenoCA, aw/node mets, 63 F 1 0.0150
  • SEQ ID NO:3 is differentially expressed in adenocarcinoma of the lung. Expression exceeds that seen in asthmatic and cytologically normal lung by approximately two-fold.
  • SEQ ID NO:6 Category: Musculoskeletal System
  • Library cDNAs Description Abundance % Abundance SYNWDIT01 3232 synovium, wrist, dorsal, rheuA, 64 F 4 0.1238 SYNORAB01 5140 synovium, hip, rheuA, 68 F 6 0.1167 SYNORAT03 5814 synovium, wrist, rheuA, 56 F 6 0.1032 SYNORAT04 5665 synovium, wrist, rheuA, 62 F 3 0.0530
  • SEQ ID NO:6 was differentially expressed in the synovium afflicted with rheumatoid arthritis. The sequence was not highly expressed in association with libraries with designated osteoarthritis or in cultured chrondrocytes.
  • the complement of the novel polynucleotide from about 5 bp (e.g., a PNA) to about 5000 bp (e.g., the complement of a cDNA insert), are used to detect or inhibit gene expression. These molecules are selected using LASERGENE software (DNASTAR). Detection is described in Example VIII. 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 helix” base pairing to compromise the ability of the double helix 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 cell, zygote, or other reproducing lineage for long term or stable gene therapy.
  • Transient expression lasts for a month or more with a non-replicating vector and for three months or more if appropriate elements for inducing vector replication are used in the transformation/expression system.
  • Expression and purification of the protein are achieved using either a cell expression system or an insect cell expression system.
  • the pUB6/V5-His vector system (Invitrogen, Carlsbad Calif.) is used to express protein 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 (6 ⁇ His) sequence for rapid purification on PROBOND resin (Invitrogen). Transformed cells are selected on media containing blasticidin.
  • Spodoptera frugiperda (Sf9) insect cells are infected with recombinant Autographica californica nuclear polyhedrosis virus (baculovirus).
  • the polyhedrin gene is replaced with the polynucleotide by homologous recombination and the polyhedrin promoter drives transcription.
  • the protein is synthesized as a fusion protein with 6 ⁇ his which enables purification as described above. Purified protein is used in the following activity and to make antibodies.
  • the protein 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 the expressed protein is analyzed using LASERGENE software (DNASTAR) to determine regions of high antigenicity. An antigenic epitope, usually found near the C-terminus or in a hydrophilic region is selected, synthesized, and used to raise antibodies.
  • epitopes of about 15 residues in length are produced using an ABI 431A peptide synthesizer (Applied Biosystems) using Fmoc-chemistry and coupled to KLH (Sigma-Aldrich) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester to increase antigenicity.
  • ABI 431A peptide synthesizer Applied 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 well known in the art are used to determine antibody titer and the amount of complex formation.
  • Naturally occurring or recombinant protein is purified by immunoaffinity chromatography using antibodies which specifically bind the protein.
  • An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected.
  • APB CNBr-activated SEPHAROSE resin
  • the polynucleotide, 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 FITC (Molecular Probes, Eugene Oreg.), respectively.
  • Libraries of candidate molecules or compounds previously arranged on a substrate are incubated in the presence of composition, a labeled polynucleotide 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 ligand 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 Calif.), is used to screen for peptides that bind the protein of the invention.
  • a polynucleotide encoding the protein is inserted into the multiple cloning site of a pLexA vector, ligated, and transformed into E. coli.
  • cDNA prepared from mRNA, is inserted into the multiple cloning site of a pB42AD vector, ligated, and transformed into E. coli to construct a cDNA library.
  • the pLexA plasmid and pB42AD-cDNA library constructs are isolated from E.
  • Transformed yeast cells 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 1x TE (pH 7.5), replated on SD/-His/-Leu/-Trp/-Ura 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.
  • 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 polynucleotide encoding a protein that physically interacts with the protein, is isolated from the yeast cells and characterized.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention provides compositions and novel polynucleotides and proteins that co-express with known marker genes for inflammatory disorders. The invention also provides expression vectors, host cells, proteins encoded by the polynucleotides and antibodies which specifically bind the proteins. The invention also provides methods for the diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders.

Description

  • This application is a divisional application of U.S. Ser. No. 09/855,323, filed May 14, 2001, which is a continuation-in-part of U.S. Ser. No. 09/195,292, filed 18 Nov. 1998, both of which applications are hereby incorporated by reference herein.[0001]
  • FIELD OF THE INVENTION
  • The invention relates to eleven isolated polynucleotides and their encoded proteins that are highly co-expressed with genes known to be diagnostic markers of inflammation and useful for diagnosis, prognosis, treatment of inflammatory disorders. [0002]
  • BACKGROUND OF THE INVENTION
  • Inflammation is the body's immediate, general response to wounding or infection by a pathogen. There are many complex phenomena that occur during an inflammation response. Initiation of the complement cascade, leukocyte recruitment and leukocyte activation are three key events. In the complement cascade a set of serum proteins collectively called complement non-specifically coat foreign matter. The coating proceeds in a cascade of steps using particular subsets of factors called complement components. The coated particles are then phagocytosed by macrophages or neutrophils recruited to the inflammation site. Leukocyte recruitment of monocytes and neutrophils is mediated by cytokines, which are proteins secreted by tissue at the inflammation site. Interleukin-8 (IL-8) is the primary chemoattractant cytokine responsible for recruitment in the initial stage of inflammation. In response to IL-8, monocytes and neutrophils are activated. An immediate response to activation is the expression of L-selectin and the integrins. L-selectin is a surface molecule that facilitates leukocyte binding (with relatively low affinity) to the endothelial cells lining blood vessels in the vicinity of the inflammation site. The integrins, also cell surface molecules, have stronger binding and mediate the actual extravasation of leukoctyes from the blood vessel. Upon reaching the site of inflammation, receptors to the complement factors coating foreign particles are expressed on the leukocytes leading to phagocytosis and enzymatic degradation. [0003]
  • Many genes that participate in and regulate the inflammation response are known, but many remain to be identified. Identification of currently unknown genes will provide new diagnostic markers and therapeutic targets for control of the inflammation response and treatment of inflammatory disorders. [0004]
  • The present invention provides new compositions that are highly co-expressed with genes known to be diagnostic of inflammatory disorders and useful for diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. [0005]
  • SUMMARY OF THE INVENTION
  • The invention provides for a plurality of polynucleotides having the nucleic acid sequences of SEQ ID NOs: 1-11 or the complements thereof that are co-expressed with genes such as CD16; L-selectin; Src-like adapter protein (SLAP); IP-30; superoxidase homoenzyme subunits, p67phox, p47phox, and p40phox; alpha-1-antitrypsin (AAT); Clq-A; 5-lipoxygenase activating protein (FLAP); and SRC family tyrosine kinase (HCK) known to be highly expressed during inflammation. The invention also provides an isolated polynucleotide comprising a nucleic acid sequence selected from SEQ ID NOs: 1-11 or the complement thereof. In different aspects, the polynucleotide is used as a surrogate marker, as a probe, in an expression vector, and in the diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. The invention further provides a composition comprising either a plurality of polynucleotides or a polynucleotide and a labeling moiety. [0006]
  • The invention provides a method for using a composition or a polynucleotide of the invention to screen a plurality of molecules and compounds to identify ligands which bind the polynucleotide(s). The molecules are selected from DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, transcription factors, enhancers, and repressors. The invention also provides a method of using a composition or a polynucleotide to purify a ligand. [0007]
  • The invention provides a method for using a composition or an isolated polynucleotide to detect gene expression in a sample by hybridizing the composition or polynucleotide to nucleic acids of the sample under conditions for formation of one or more hybridization complexes and detecting hybridization complex formation, wherein complex formation indicates gene expression in the sample. In one aspect, the composition or polynucleotide is attached to a substrate. In another aspect, the nucleic acids of the sample are amplified prior to hybridization. In yet another aspect, complex formation is compared with at least one standard and indicates the presence of an inflammatory disorder. [0008]
  • The invention provides a purified protein or a portion thereof, SEQ ID NOs: 12-17, which is encoded by a polynucleotide that is co-expressed with genes that are diagnostic markers of inflammation or inflammatory disorders. The invention also provides a method for using a protein to screen a plurality of molecules to identify at least one ligand which specifically binds the protein. The molecules are selected from aptamers, DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, proteins, antibodies, agonists, antagonists, immunoglobulins, inhibitors, pharmaceutical agents or drug compounds. The invention further provides a method of using a protein to purify a ligand. [0009]
  • The invention provides a method of using a protein to make an antibody that specifically binds to the protein of the invention, and methods for using the antibody to diagnose or treat an inflammatory disorder. The invention further provides a composition comprising a polynucleotide, a protein, or an antibody that specifically binds a protein and a pharmaceutical carrier. [0010]
  • BRIEF DESCRIPTION OF THE SEQUENCE LISTING
  • The Sequence Listing provides exemplary polynucleotides comprising the nucleic acid sequences of SEQ ID NOs: 1-1I some of which encode the proteins comprising the amino acid sequences of SEQ ID NOs: 12-17. Each sequence is identified by a sequence identification number (SEQ ID NO) and by the Incyte clone number with which the sequence was first identified. [0011]
  • DESCRIPTION OF THE INVENTION
  • It must be noted that as used herein and in the appended claims, the singular forms “a”, “an”, and “the” include the plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to “a host cell” includes a plurality of such host cells, and a reference to “an antibody” is a reference to one or more antibodies and equivalents thereof known to those skilled in the art, and so forth. [0012]
  • Definitions
  • “Markers for inflammatory disorders” refers to polynucleotides, proteins, and antibodies which are useful in the diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. Typically, this means that the marker gene is differentially expressed in samples from subjects predisposed to, manifesting, or diagnosed with an inflammatory disorder. [0013]
  • “Differential expression” refers to an increased or upregulated or a decreased or downregulated expression as detected by presence, absence or at least about a two-fold change in the amount of transcribed messenger RNA or protein in a sample. [0014]
  • “Inflammatory disorders” specifically include, but are not limited to, the following conditions, diseases, and disorders: adult respiratory distress syndrome, allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes mellitus, emphysema, gastritis, hypereosinophilia, irritable bowel syndrome, lung cancer of complications thereof, lymphoma, meningitis, multiple sclerosis, osteoarthritis, psoriasis, rheumatoid arthritis, and toxic shock syndrome. [0015]
  • “Isolated or purified” refers to a polynucleotide or protein that is removed from its natural environment and that is separated from other components with which it is naturally present. [0016]
  • “Genes known to be highly expressed during inflammation” and used in the co-expression analysis included CD16; L-selectin; Src-like adapter protein (SLAP); IP-30; superoxidase homoenzyme subunits, p67phox, p47phox, and p40phox; alpha-1-antitrypsin (AAT); Clq-A; 5-lipoxygenase activating protein (FLAP); and SRC family tyrosine kinase (HCK). [0017]
  • “Polynucleotide” refers to an isolated cDNA. It can be of genomic or synthetic origin, double-stranded or single-stranded, and combined with vitamins, minerals, carbohydrates, lipids, proteins, or other nucleic acids to perform a particular activity or form a useful composition. [0018]
  • “Protein” refers to a purified polypeptide whether naturally occurring or synthetic. [0019]
  • “Sample” is used in its broadest sense. A sample containing nucleic acids can comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; genomic DNA, RNA, or cDNA in solution or bound to a substrate; a cell; a tissue; a tissue print; and the like. [0020]
  • “Substrate” refers to any rigid or semi-rigid support to which polynucleotides 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. [0021]
  • A “transcript image” is a profile of gene transcription activity in a particular tissue at a particular time. [0022]
  • A “variant” refers to a polynucleotide or protein whose sequence diverges from about 5% to about 30% from the nucleic acid or amino acid sequences of the Sequence Listing. [0023]
  • The Invention
  • The present invention employed “guilt by association or GBA”, a method for using marker genes known to be associated with a particular condition, disease or disorder to identify surrogate markers, polynucleotides and their encoded proteins, that are similarly associated or co-expressed in the same condition, disease, or disorder (Walker and Volkmuth (1999) Prediction of gene function by genome-scale expression analysis: prostate-associated genes. Genome Res 9:1198-1203, incorporated herein by reference). In particular, the method identifies cDNAs cloned from mRNA transcripts which are active in tissues known to have been removed from subjects with inflammatory disorders. The polynucleotides are useful for diagnosis, prognosis, evaluation of therapies, and treatment of inflammatory disorders. [0024]
  • Guilt by association provides for the identification of polynucleotides that are expressed in a plurality of libraries. The polynucleotides represent genes of unknown function which are expressed in a specific signaling pathway, disease process, subcellular compartment, cell type, tissue, or species. The expression patterns of the genes known to be highly expressed during inflammation, CD16, L-selectin, SLAP, IP-30, p67phox, p47phox, p40phox, AAT, Clq-A; FLAP, and HCK, are compared with those of polynucleotides with unknown function to determine whether a specified co-expression probability threshold is met. Through this comparison, a subset of the polynucleotides having a high co-expression probability with the known marker genes can be identified. [0025]
  • The polynucleotides originate from human cDNA libraries. These polynucleotides can also be selected from a variety of sequence types including, but not limited to, expressed sequence tags (ESTs), assembled polynucleotides, full length coding regions, and 3′ untranslated regions. To be considered in the co-expression analysis, the polynucleotides have been expressed in at least five cDNA libraries. In this application, GBA was applied to a total of 41,419 assembled gene bins to identify eleven novel polynucleotides. [0026]
  • The cDNA libraries used in the co-expression analysis were obtained from adrenal gland, biliary tract, bladder, blood cells, blood vessels, bone marrow, brain, bronchus, cartilage, chromaffin system, colon, connective tissue, cultured cells, embryonic stem cells, endocrine glands, epithelium, esophagus, fetus, ganglia, heart, hypothalamus, hemic/immune system, intestine, islets of Langerhans, kidney, larynx, liver, lung, lymph, muscles, neurons, ovary, pancreas, penis, phagocytes, pituitary, placenta, pleura, prostate, salivary glands, seminal vesicles, skeleton, spleen, stomach, testis, thymus, tongue, ureter, uterus, and the like. The number of cDNA libraries selected can range from as few as three to greater than 10,000 and preferably, the number of the cDNA libraries is greater than 500. [0027]
  • In a preferred embodiment, the polynucleotides are assembled from related sequences, such as sequence fragments derived from a single transcript. Assembly of the polynucleotide can be performed using sequences of various types including, but not limited to, ESTs, extension of the EST, shotgun sequences from a cloned insert, or full length cDNAs. In a most preferred embodiment, the polynucleotides are derived from human sequences that have been assembled using the algorithm disclosed in U.S. Ser. No. 9,276,534, filed Mar. 25, 1999, and used in U.S. Ser. No. 09/195,292, filed 18 November 1998, both incorporated herein by reference. [0028]
  • Experimentally, differential expression of the polynucleotides can be evaluated by methods including, but not limited to, differential display by spatial immobilization or by gel electrophoresis, genome mismatch scanning, representational difference analysis, and transcript imaging. The results of transcript imaging for SEQ ID NOs: 1, 2, 3, and 6 shows significant expression of these sequences in BPH, hypereosinophilia, lung cancer, and rheumatoid arthritis, respectively. The transcript images provided direct confirmation of the strength of co-expression analysis using known genes to identify unknown polynucleotides and their encoded proteins which are highly significantly associated with inflammatory disorders. Additionally, differential expression can be assessed by microarray technology. These methods can be used alone or in combination. [0029]
  • Genes known to be highly expressed during inflammation can be selected based on research in which the genes are found to be key elements of immune response pathways or on the known use of the genes as diagnostic or prognostic markers or therapeutic targets for inflammatory disorders. Preferably, the known genes are CD 16, L-selectin, SLAP, IP-30, p67phox, p47phox, p40phox, AAT, Clq-A, FLAP, and HCK. [0030]
  • The procedure for identifying novel polynucleotides that exhibit a statistically significant co-expression pattern with known genes is as follows. First, the presence or absence of a polynucleotide in a cDNA library is defined: a polynucleotide is present in a cDNA library when at least one cDNA fragment corresponding to the polynucleotide is detected in a cDNA from that library, and a polynucleotide is absent from a library when no corresponding cDNA fragment is detected. [0031]
  • Second, the significance of co-expression is evaluated using a probability method to measure a due-to-chance probability of the co-expression. The probability method can be the Fisher exact test, the chi-squared test, or the kappa test. These tests and examples of their applications are well known in the art and can be found in standard statistics texts (Agresti (1990) [0032] Categorical Data Analysis, John Wiley & Sons, New York N.Y.; Rice (1988) Mathematical Statistics and Data Analysis, Duxbury Press, Pacific Grove Calif.). A Bonferroni correction (Rice, supra, p. 384) can also be applied in combination with one of the probability methods for correcting statistical results of one polynucleotide versus multiple other polynucleotides. In a preferred embodiment, the due-to-chance probability is measured by a Fisher exact test, and the threshold of the due-to-chance probability is set preferably to less than 0.001, more preferably to less than 0.00001.
  • For example, to determine whether two genes, A and B, have similar co-expression patterns, occurrence data vectors can be generated as illustrated in the table below. The presence of a gene occurring at least once in a library is indicated by a one, and its absence from the library, by a zero. [0033]
    Library 1 Library 2 Library 3 ... Library N
    Gene A 1 1 0 ... 0
    Gene B 1 0 1 ... 0
  • For a given pair of genes, the occurrence data in the table above can be summarized in a 2×2 contingency table. The second table (below) presents co-occurrence data for gene A and gene B in a total of 30 libraries. Both gene A and gene B occur 10 times in the libraries. [0034]
    Gene A Present Gene A Absent Total
    Gene B Present 8 2 10
    Gene B Absent 2 18 20
    Total 10 20 30
  • The second table summarizes and presents: 1) the number of times gene A and B are both present in a library; 2) the number of times gene A and B are both absent in a library; 3) the number of times gene A is present, and gene B is absent; and 4) the number of times gene B is present, and gene A is absent. The upper left entry is the number of times the two genes co-occur in a library, and the middle right entry is the number of times neither gene occurs in a library. The off diagonal entries are the number of times one gene occurs, and the other does not. Both A and B are present eight times and absent 18 times. Gene A is present, and gene B is absent, two times; and gene B is present, and gene A is absent, two times. The probability (“p-value”) that the above association occurs due to chance as calculated using a Fisher exact test is 0.0003. Associations are generally considered significant if a p-value is less than 0.01 (Agresti, supra; Rice, supra). [0035]
  • This method of estimating the probability for co-expression makes several assumptions. The method assumes that the libraries are independent and are identically sampled. However, in practical situations, the selected cDNA libraries are not entirely independent, because more than one library can be obtained from a single subject or tissue. Nor are they entirely identically sampled, because different numbers of cDNAs can have been sequenced from each library. The number of cDNAs sequenced typically ranges from 5,000 to 10,000 cDNAs per library. After the Fisher exact co-expression probability is calculated for each polynucleotide versus all other assembled polynucleotides that occur, a Bonferroni correction for multiple statistical tests is applied. [0036]
  • Using the method of the present invention, we have identified polynucleotides, SEQ ID NOs: 1-11 and their encoded proteins, SEQ ID NOs: 12-17, that exhibit highly significant co-expression probability with known marker genes for inflammatory disorders. The results presented in Example VI show the direct (known gene to unknown polynucleotide) or indirect (known gene to unknown polynucleotide to a second unknown polynucleotide) associations among the novel polynucleotides and the known marker genes for inflammatory disorders. Therefore, by these associations, the novel polynucleotides are useful as surrogate markers for the co-expressed known marker genes in diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders. Further, the proteins or peptides expressed from the novel polynucleotides are either potential therapeutics or targets for the identification and/or development of therapeutics. [0037]
  • In one embodiment, the present invention encompasses a composition comprising a plurality of polynucleotides having the nucleic acid sequences of SEQ ID NOs: 1-11 or the complements thereof. These eleven polynucleotides are shown by the method of the present invention to have significant co-expression with known genes associated with inflammatory disorders. The invention also provides a polynucleotide, its complement, a probe comprising the polynucleotide or the complement thereof selected from SEQ ID NOs: 1-11 and variants of the polynucleotides. [0038]
  • The polynucleotide can be used to search against the GenBank primate (pri), rodent (rod), mammalian (mam), vertebrate (vrtp), and eukaryote (eukp) databases; the encoded protein, against GenPept, SwissProt, BLOCKS (Bairoch et al. (1997) Nucleic Acids Res 25:217-221), PFAM, and other databases that contain previously identified and annotated protein sequences, motifs, and gene functions. Methods that search for primary sequence patterns with secondary structure gap penalties (Smith et al. (1992) Protein Engineering 5:35-51) as well as algorithms such as Basic Local Alignment Search Tool (BLAST; Altschul (1993) J Mol Evol 36:290-300; Altschul et al. (1990) J Mol Biol 215:403410), BLOCKS (Henikoff and Henikoff (1991) Nucleic Acids Res 19:6565-6572), Hidden Markov Models (HMM; Eddy (1996) Cur Opin Str Biol 6:361-365; Sonnhammer et al. (1997) Proteins 28:405-420), and the like, can be used to manipulate and analyze nucleotide and amino acid sequences. These databases, algorithms and other methods are well known in the art and are described in Ausubel et al. (1997; [0039] Short Protocols in Molecular Biology, John Wiley & Sons, New York N.Y., unit 7.7) and in Meyers (1995; Molecular Biology and Biotechnology, Wiley VCH, New York N.Y., p 856-853).
  • Also encompassed by the invention are polynucleotides that are capable of hybridizing to SEQ ID NOs: 1-11, and fragments thereof, under highly stringent conditions. Stringency can be defined by salt concentration, temperature, and other chemicals and conditions well known in the art. Conditions can be selected, for example, by varying the concentrations of salt in the prehybridization, hybridization, and wash solutions or by varying the hybridization and wash temperatures. With some substrates, the temperature can be decreased by adding a solvent such as formamide to the prehybridization and hybridization solutions. [0040]
  • Hybridization can be performed at low stringency, with buffers such as 5×SSC (saline sodium citrate) with 1% sodium dodecyl sulfate (SDS) at 60° C., which permits complex formation between two nucleic acid sequences that contain some mismatches. Subsequent washes are performed at higher stringency with buffers such as 0.2×SSC with 0.1% SDS at either 45° C. (medium stringency) or 68° C. (high stringency), to maintain hybridization of only those complexes that contain completely complementary sequences. Background signals can be reduced by the use of detergents such as SDS, sarcosyl, or TRITON X-100 (Sigma-Aldrich, St. Louis Mo.), and/or a blocking agent, such as salmon sperm DNA. Hybridization methods are described in detail in Ausubel (supra, units 2.8-2.11, 3.18-3.19 and 4-6-4.9) and Sambrook et al. (1989; [0041] Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview N.Y.)
  • A polynucleotide can be extended utilizing primers and employing various PCR-based methods known in the art to detect upstream sequences such as promoters and other regulatory elements. (See, e.g., Dieffenbach and Dveksler (1995) [0042] PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview N.Y.). Commercially available kits such as XL-PCR (Applied Biosystems, Foster City Calif.), cDNA libraries (Life Technologies, Rockville Md.) or genomic libraries (Clontech, Palo Alto Calif.) and nested primers can be used to extend the sequence. For all PCR-based methods, primers can be designed using commercially available software (LASERGENE software, DNASTAR, Madison Wis.) or another program, to be about 15 to 30 nucleotides in length, to have a GC content of about 50%, and to form a hybridization complex at temperatures of about 68° C. to 72° C.
  • In another aspect of the invention, the polynucleotide can be cloned into a recombinant vector that directs the expression of the protein, or structural or functional portions thereof, in host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode functionally equivalent amino acid sequence can be produced and used to express the protein encoded by the polynucleotide. The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter the nucleotide sequences for a variety of purposes including, but not limited to, modification of the cloning, processing, and/or expression of the gene product. DNA shuffling by random fragmentation, as described in U.S. Pat. No. 5,830,721, and PCR reassembly of gene fragments and synthetic oligonucleotides can be used to engineer the nucleotide sequences. For example. oligonucleotide-mediated site-directed mutagenesis can be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth. [0043]
  • In order to express a biologically active protein, the polynucleotide or derivatives thereof, can be inserted into an expression vector with elements for transcriptional and translational control of the inserted coding sequence in a particular host. These elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5′ and 3′ untranslated regions. Methods which are well known to those skilled in the art can be used to construct such expression vectors. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination (Ausubel, supra, unit 16). [0044]
  • A variety of expression vector/host cell systems can be utilized to express the polynucleotide. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with baculovirus vectors; plant cell systems transformed with viral or bacterial expression vectors; or animal cell systems. For long term production of recombinant proteins in mammalian systems, stable expression in cell lines is preferred. For example, the polynucleotide can be transformed into cell lines using expression vectors which can contain viral origins of replication and/or endogenous expression elements and a selectable or visible marker gene on the same or on a separate vector. The invention is not to be limited by the vector or host cell employed. [0045]
  • In general, host cells that contain the polynucleotide and that express the protein can be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations, PCR amplification, and protein bioassay or immunoassay techniques which include membrane, solution, or chip-based technologies for the detection and/or quantification of nucleic acid or amino acid sequences. Immunological methods for detecting and measuring the expression of the protein using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). [0046]
  • Host cells transformed with the polynucleotide can be cultured under conditions for the expression and recovery of the protein from cell culture. The protein produced by a transgenic cell can be secreted or retained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing the polynucleotide can be designed to contain signal sequences which direct secretion of the protein through a prokaryotic cell wall or eukaryotic cell membrane. [0047]
  • In addition, a host cell strain can be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the protein include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a “prepro” form of the protein can also be used to specify protein targeting, folding, and/or activity. Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDCK, HEK293, and W138) are available from the ATCC (Manassas Va.) and can be chosen to ensure the correct modification and processing of the expressed protein. [0048]
  • In another embodiment of the invention, natural, modified, or recombinant polynucleotides are ligated to a heterologous sequence resulting in translation of a fusion protein containing heterologous protein moieties in any of the aforementioned host systems. Such heterologous protein moieties facilitate purification of fusion proteins using commercially available affinity matrices. Such moieties include, but are not limited to, glutathione S-transferase, maltose binding protein, thioredoxin, calmodulin binding peptide, 6-His, FLAG, c-myc, hemaglutinin, and monoclonal antibody epitopes. [0049]
  • In another embodiment, the polynucleotides, wholly or in part, are synthesized using chemical or enzymatic methods well known in the art (Caruthers et al. (1980) Nucl Acids Symp Ser (7) 215-233; Ausubel, supra, units 10.4 and 10.16). Peptide synthesis can be performed using various solid-phase techniques (Roberge et al. (1995) Science 269:202-204), and machines such as the ABI 431A peptide synthesizer (Applied Biosystems) can be used to automate synthesis. If desired, the amino acid sequence can be altered during synthesis to produce a more stable variant for therapeutic use. [0050]
  • Screening, Diagnostics and Therapeutics
  • The polynucleotides can be used as surrogate markers in diagnosis, prognosis, evaluation of therapies and treatment of inflammatory disorders including but not limited to adult respiratory distress syndrome, allergy, anemia, asthma, atherosclerosis, bacterial infection, benign prostatic hyperplasia (BPH), cholecystitis, chronic heart failure (CHF), chronic ulcerative colitis, Crohn's disease, diabetes mellitus, emphysema, gastritis, hypereosinophilia, irritable bowel syndrome, lung cancer of complications thereof, lymphoma, meningitis, multiple sclerosis, osteoarthritis, psoriasis, rheumatoid arthritis, and toxic shock syndrome. [0051]
  • The polynucleotide can be used to screen a plurality or library of molecules and compounds for specific binding affinity. The assay can be used to screen DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, or proteins including transcription factors, enhancers, repressors, and the like which regulate the activity of the polynucleotide in the biological system. The assay involves providing a plurality of molecules and compounds, combining a polynucleotide or a composition of the invention with the plurality of molecules and compounds under conditions to allow specific binding, and detecting specific binding to identify at least one molecule or compound which specifically binds at least one polynucleotides of the invention. [0052]
  • Similarly the proteins, or portions thereof, can be used to screen a plurality or library of molecules or compounds in any of a variety of screening assays to identify a ligand. The protein employed in such screening can be free in solution, affixed to an abiotic substrate or expressed on the external, or a particular internal surface, of a bacterial, or other, cell. Specific binding between the protein and the ligand can be measured. The assay can be used to screen aptamers, DNA molecules, RNA molecules, peptide nucleic acids, peptides, mimetics, ribozymes, proteins, antibodies, agonists, antagonists, immunoglobulins, inhibitors, pharmaceutical agents or drug compounds and the like, which specifically bind the protein. One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in Burbaum et al. U.S. Pat. No. 5,876,946, incorporated herein by reference, which screens large numbers of molecules for enzyme inhibition or receptor binding. [0053]
  • In one preferred embodiment, the polynucleotides are used for diagnostic purposes to determine the differential expression of a gene in a sample. The polynucleotide consists of complementary RNA and DNA molecules, branched nucleic acids, and/or PNAs. In one alternative, the polynucleotides are used to detect and quantify gene expression in biopsied samples in which differential expression of the polynucleotide indicates the presence of a disorder. In another alternative, the polynucleotide can be used to detect genetic polymorphisms associated with a disease. In a preferred embodiment, these polymorphisms are detected in an mRNA transcribed from an endogenous gene. [0054]
  • In another preferred embodiment, the polynucleotide is used as a probe. Specificity of the probe is determined by whether it is made from a unique region, a regulatory region, or from a region encoding a conserved motif. Both probe specificity and the stringency of the diagnostic hybridization or amplification will determine whether the probe identifies only naturally occurring, exactly complementary sequences, allelic variants, or related sequences. Probes designed to detect related sequences should preferably have at least 50% sequence identity to at least a fragment of a polynucleotide of the invention. [0055]
  • Methods for producing hybridization probes include the cloning of nucleic acid sequences into vectors for the production of RNA probes. Such vectors are known in the art, are commercially available, and can be used to synthesize RNA probes in vitro by adding RNA polymerases and labeled nucleotides. Probes can incorporate nucleotides labeled by a variety of reporter groups including, but not limited to, radionuclides such as 32p or 35S, enzymatic labels such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, fluorescent labels such as Cy3 and Cy5, and the like. The labeled polynucleotides can be used in Southern or northern analysis, dot blot, or other membrane-based technologies, on chips or other substrates, and in PCR technologies. Hybridization probes are also useful in mapping the naturally occurring genomic sequence. Fluorescent in situ hybridization (FISH) can be correlated with other physical chromosome mapping techniques and genetic map data as described in Heinz-Ulrich et al. (In: Meyers, supra, pp. 965-968). In many cases, genomic context helps identify genes that are encode a particular protein family. (See, e.g., Kirschning et al. (1997) Genomics 46:416-25.) [0056]
  • The polynucleotide can be labeled using standard methods and added to a sample from a subject under conditions for the formation and detection of hybridization complexes. After incubation the sample is washed, and the signal associated with complex formation is quantitated and compared with at least one standard value. Standard values are derived from any control sample, typically one that is free of the suspect disorder and from diseased samples, preferably from samples each of which represents a single, specific and preferably, staged disorder. If the amount of signal in the subject sample is altered in comparison to the standard values, then differential expression in the biopsied sample indicates the presence of the disorder. Qualitative and quantitative methods for comparing complex formation in subject samples with previously established standards are well known in the art. [0057]
  • Such assays can also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or to monitor the treatment of an individual subject. Once the presence of disease is established and a treatment protocol is initiated, hybridization or amplification assays can be repeated on a regular basis to determine gene expression in the patient begins to approximate that which is observed in a healthy subject. The results obtained from successive assays can be used to show the efficacy of treatment over a period ranging from several hours, e.g. in the case of toxic shock, to many years ,e.g. in the case of osteoarthritis. [0058]
  • The polynucleotides can be used on a substrate such as a microarray to monitor gene expression, to identify splice variants, mutations, and polymorphisms. Information derived from analyses of expression patterns can be used to determine gene function, to understand the genetic basis of a disease, to diagnose a disorder, and to develop and monitor the activities of therapeutic agents used to treat a disorder. Microarrays can also be used to detect genetic diversity, single nucleotide polymorphisms, which may characterize a particular population, at the genomic level. [0059]
  • In another embodiment, antibodies or Fabs comprising an antigen binding site that specifically binds the protein can be used for the diagnosis of diseases characterized by the differential expression of the protein. A variety of protocols for measuring protein expression, including ELISAs, RIAs, and FACS, are well known in the art and provide a basis for diagnosing altered or abnormal levels of expression. Standard values for protein expression parallel those reviewed above for nucleotide expression. The amount of complex formation can be quantitated by various methods, preferably by photometric means. Quantities of the protein expressed in subject samples are compared with standard values. Deviation between standard and subject values establishes the parameters for diagnosing or monitoring a particular disorder. Alternatively, one can use competitive drug screening assays in which neutralizing antibodies capable of binding specifically with the protein compete with a test compound. Antibodies can be used to detect the presence of any peptide which shares one or more epitopes or antigenic determinants with the protein. In one aspect, the antibodies of the present invention can be used for treatment, delivery of therapeutics, or monitoring therapy for inflammatory disorders. [0060]
  • In another aspect, the polynucleotide, or its complement, can be used therapeutically for the purpose of expressing mRNA and protein, or conversely to block transcription or translation of the mRNA. Expression vectors can be constructed using elements from retroviruses, adenoviruses, herpes or vaccinia viruses, or bacterial plasmids, and the like. These vectors can be used for delivery of nucleotide sequences to a particular target cell population, tissue, or organ. Methods well known to those skilled in the art can be used to construct vectors to express the polynucleotides or their complements. (See, e.g., Maulik et al. (1997) [0061] Molecular Biotechnology, Therapeutic Applications and Strategies, Wiley-Liss, New York NY.) Alternatively, the polynucleotide or its complement, can be used for somatic cell or stem cell gene therapy. Vectors can be introduced in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors are introduced into stem cells taken from the subject, and the resulting transgenic cells are clonally propagated for autologous transplant back into that same subject. Delivery of the polynucleotide by transfection, liposome injections, or polycationic amino polymers can be achieved using methods which are well known in the art. (See, e.g., Goldman et al. (1997) Nature Biotechnology 15:462-466.) Additionally, endogenous gene expression can be inactivated using homologous recombination methods which insert an inactive gene sequence into the coding region or other targeted region of the genome (See, e.g. Thomas et al. (1987) Cell 51: 503-512.)
  • Vectors containing the polynucleotide can be transformed into a cell or tissue to express a missing protein or to replace a nonfunctional protein. Similarly a vector constructed to express the complement of the polynucleotide can be transformed into a cell to downregulate protein expression. Complementary or antisense sequences can consist of an oligonucleotide derived from the transcription initiation site; nucleotides between about positions −10 and +10 from the ATG are preferred. Similarly, inhibition can be achieved using triple helix base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature. (See, e.g., Gee et al. In: Huber and Carr (1994) [0062] Molecular and Immunologic Approaches, Futura Publishing, Mt. Kisco N.Y., pp. 163-177.)
  • Ribozymes, enzymatic RNA molecules, can also be used to catalyze the cleavage of mRNA and decrease the levels of particular mRNAs, such as those comprising the polynucleotides of the invention. (See, e.g., Rossi (1994) Current Biology 4: 469-471.) Ribozymes can cleave mRNA at specific cleavage sites. Alternatively, ribozymes can cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The construction and production of ribozymes is well known in the art and is described in Meyers (supra). [0063]
  • RNA molecules can be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends of the molecule, or the use of phosphorothioate or 2′O-methyl rather than phosphodiester linkages within the backbone of the molecule. Alternatively, nontraditional bases such as inosine, queosine, and wybutosine, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases, can be included. [0064]
  • Further, an antagonist, or an antibody that binds specifically to the protein can be administered to a subject to treat an inflammatory disorder. The antagonist, antibody, or fragment can be used directly to inhibit the activity of the protein or indirectly to deliver a therapeutic agent to cells or tissues which express the protein. The therapeutic agent can be a cytotoxic agent selected from a group including, but not limited to, abrin, ricin, doxorubicin, daunorubicin, taxol, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin D, diphteria toxin, Pseudomonas exotoxin A and 40, radioisotopes, and glucocorticoid. [0065]
  • Antibodies to the protein can be generated using methods that are well known in the art. Such antibodies can include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies, such as those which inhibit dimer formation, are especially preferred for therapeutic use. Monoclonal antibodies to the protein can be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma, the human B-cell hybridoma, and the EBV-hybridoma techniques. In addition, techniques developed for the production of chimeric antibodies can be used. (See, e.g., Pound (1998) [0066] Immunochemical Protocols, Methods Mol Biol Vol. 80). Alternatively, techniques described for the production of single chain antibodies can be employed. Fabs which contain specific binding sites for the protein can also be generated. Various immunoassays can be used to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art.
  • Yet further, an agonist of the protein can be administered to a subject to treat or prevent a disease associated with decreased expression, longevity or activity of the protein. [0067]
  • An additional aspect of the invention relates to the administration of a pharmaceutical or sterile composition, in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic applications discussed above. Such pharmaceutical compositions can consist of the protein or antibodies, mimetics, agonists, antagonists, or inhibitors of the protein. The compositions can be administered alone or in combination with at least one other agent, such as a stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier including, but not limited to, saline, buffered saline, dextrose, and water. The compositions can be administered to a subject alone or in combination with other agents, drugs, or hormones. [0068]
  • The pharmaceutical compositions utilized in this invention can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means. [0069]
  • In addition to the active ingredients, these pharmaceutical compositions can contain pharmaceutically-acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration can be found in the latest edition of [0070] Remington's Pharmaceutical Sciences (Mack Publishing, Easton Pa.).
  • For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, or pigs. An animal model can also be used to determine the concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. [0071]
  • A therapeutically effective dose refers to that amount of active ingredient which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity can be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating and contrasting the ED[0072] 50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population) statistics. Any of the therapeutic compositions described above can be applied to any subject in need of such therapy, including, but not limited to, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans.
  • Stem Cells and Their Use [0073]
  • SEQ ID NOs: 1-11 can be useful in the differentiation of stem cells. Eukaryotic stem cells are able to differentiate into the multiple cell types of various tissues and organs and to play roles in embryogenesis and adult tissue regeneration (Gearhart (1998) Science 282:1061-1062; Watt and Hogan (2000) Science 287:1427-1430). Depending on their source and developmental stage, stem cells can be totipotent with the potential to create every cell type in an organism and to generate a new organism, pluripotent with the potential to give rise to most cell types and tissues, but not a whole organism; or multipotent cells with the potential to differentiate into a limited number of cell types. Stem cells can be transfected with polynucleotides which can be transiently expressed or can be integrated within the cell as transgenes. [0074]
  • Embryonic stem (ES) cell lines are derived from the inner cell masses of human blastocysts and are pluripotent (Thomson et al. (1998) Science 282:1145-1147). They have normal karyotypes and express high levels of telomerase which prevents senescence and allows the cells to replicate indefinitely. ES cells produce derivatives that give rise to embryonic epidermal, mesodermal and endodermal cells. Embryonic germ (EG) cell lines, which are produced from primordial germ cells isolated from gonadal ridges and mesenteries, also show stem cell behavior (Shamblott et al. (1998) Proc Natl Acad Sci 95:13726-13731). EG cells have normal karyotypes and appear to be pluripotent. [0075]
  • Organ-specific adult stem cells differentiate into the cell types of the tissues from which they were isolated. They maintain their original tissues by replacing cells destroyed from disease or injury. Adult stem cells are multipotent and under proper stimulation can be used to generate cell types of various other tissues (Vogel (2000) Science 287:1418-1419). Hematopoietic stem cells from bone marrow provide not only blood and immune cells, but can also be induced to transdifferentiate to form brain, liver, heart, skeletal muscle and smooth muscle cells. Similarly mesenchymal stem cells can be used to produce bone marrow, cartilage, muscle cells, and some neuron-like cells, and stem cells from muscle have the ability to differentiate into muscle and blood cells (Jackson et al. (1999) Proc Natl Acad Sci 96:14482-14486). Neural stem cells, which produce neurons and glia, can also be induced to differentiate into heart, muscle, liver, intestine, and blood cells (Kuhn and Svendsen (1999) BioEssays 21:625-630); Clarke et al. (2000) Science 288:1660-1663; Gage (2000) Science 287:1433-1438; and Galli et al. (2000) Nature Neurosci 3:986-991). [0076]
  • Neural stem cells can be used to treat neurological disorders such as Alzheimer disease, Parkinson disease, and multiple sclerosis and to repair tissue damaged by strokes and spinal cord injuries. Hematopoietic stem cells can be used to restore immune function in immunodeficient patients or to treat autoimmune disorders by replacing autoreactive immune cells with normal cells to treat diseases such as multiple sclerosis, scleroderma, rheumatoid arthritis, and systemic lupus erythematosus. Mesenchymal stem cells can be used to repair tendons or to regenerate cartilage to treat arthritis. Liver stem cells can be used to repair liver damage. Pancreatic stem cells can be used to replace islet cells to treat diabetes. Muscle stem cells can be used to regenerate muscle to treat muscular dystrophies (Fontes and Thomson (1999) BMJ 319:1-3; Weissman (2000) Science 287:1442-1446 Marshall (2000) Science 287:1419-1421; Marmont (2000) Ann Rev Med 51:115-134).[0077]
  • EXAMPLES
  • It is to be understood that this invention is not limited to the particular devices, machines, materials and methods described. Although particular embodiments are described, equivalent embodiments can be used to practice the invention. The described embodiments are provided to illustrate the invention and are not intended to limit the scope of the invention which is limited only by the appended claims. [0078]
  • I cDNA Library Construction [0079]
  • The cDNA library, OVARTUT05, was selected as an example to demonstrate the construction of the cDNA libraries from which the sequences used to identify genes associated with inflammatory disorders were derived. The OVARTUT05 library was constructed from tumorous ovary tissue obtained from a 62 year-old Caucasian female. Pathology indicated a grade 4 endometrioid carcinoma with extensive squamous differentiation forming a solid mass in the right ovary. The cervix showed mild chronic cervicitis, and the posterior uterine serosa showed focal endometriosis. Prior pathology indicated weakly proliferative endometrium with excessive stromal breakdown in the uterus and a mild chronic cervicitis with prominent nabothian cyst in the cervix. [0080]
  • The frozen tissue was homogenized and lysed using a POLYTRON homogenizer (Brinkmann Instruments, Westbury N.Y.) in guanidinium isothiocyanate solution. The lysate was centrifuged over a 5.7 M CsCl cushion using an SW28 rotor in a L8-70M ultracentrifuge (Beckman Coulter, Fullerton Calif.) 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 DNAse treated at 37C. The RNA extraction was repeated. The MRNA was isolated with the OLIGOTEX kit (Qiagen, Chatsworth Calif.) and used to construct the cDNA library. [0081]
  • The mRNA was handled according to the recommended protocols in the SUPERSCRIPT plasmid system (Life Technologies). The cDNAs were fractionated on a SEPHAROSE CL4B column (Amersham Pharmacia Biotech (APB), Piscataway N.J.), and those cDNAs exceeding 400 bp were ligated into pINCY plasmid (Incyte Genomics, Palo Alto Calif.). The plasmid was then transformed into DH5α competent cells (Life Technologies). [0082]
  • II Isolation and Sequencing of cDNA Clones [0083]
  • Plasmid DNA was released from the bacterial cells and purified using the REAL PREP 96 plasmid kit (Qiagen). This kit enabled the simultaneous purification of 96 samples in a 96-well block using multi-channel reagent dispensers. The recommended protocol was employed except for the following changes: 1) the bacteria were cultured in 1 ml of sterile TERRIFIC BROTH (BD Biosciences, San Jose Calif.) with carbenicillin at 25 mg/L and glycerol at 0.4%; 2) after inoculation and incubation for 19 hours, the cells were lysed with 0.3 ml of lysis buffer; and 3) following isopropanol precipitation, the plasmid DNA pellet was resuspended in 0.1 ml of distilled water. After the last step in the protocol, samples were transferred to a 96-well block for storage at 4C. [0084]
  • The cDNAs were prepared using a MICROLAB 2200 system (Hamilton, Reno Nev.) in combination with DNA ENGINE thermal cyclers (MJ Research, Watertown Mass.). The cDNAs were sequenced by the method of Sanger and Coulson (1975; J Mol Biol 94:441448) using ABI PRISM 377 DNA sequencing systems (Applied Biosystems). Most of the sequences were sequenced using standard ABI protocols and kits at solution volumes of 0.25x−1.0x. In the alternative, some of the sequences were sequenced using solutions and dyes from APB. [0085]
  • III Selection, Assembly, and Characterization of Sequences [0086]
  • The sequences used for co-expression analysis were assembled from EST sequences, 5′ and 3′ long read sequences, and full length coding sequences. Of the 41,419 assembled sequences used in the analysis, each was expressed in at least five cDNA libraries. [0087]
  • The assembly process is described as follows. EST sequence chromatograms were processed and verified. Quality scores were obtained using PHRED (Ewing et al. (1998) Genome Res 8:175-185; Ewing and Green (1998) Genome Res 8:186-194), and edited sequences were loaded into a relational database management system (RDBMS). The sequences were clustered using BLAST with a product score of 50. All clusters of two or more sequences created a bin which represents one transcribed gene. [0088]
  • Assembly of the component sequences within each bin was performed using a modification of Phrap, a publicly available program for assembling DNA fragments (Green, P. University of Washington, Seattle Wash.). Bins that showed 82% identity from a local pair-wise alignment between any of the consensus sequences were merged. [0089]
  • Bins were annotated by screening the consensus sequence in each bin against public databases, such as GBpri and GenPept from NCBI. The annotation process involved a FASTn screen against the GBpri database in GenBank. Those hits with a percent identity of greater than or equal to 75% and an alignment length of greater than or equal to 100 base pairs were recorded as homolog hits. The residual unannotated sequences were screened by FASTx against GenPept. Those hits with an E value of less than or equal to 10[0090] −8 were recorded as homolog hits.
  • Sequences were then reclustered using BLASTn and Cross-Match, a program for rapid amino acid and nucleic acid sequence comparison and database search (Green, supra), sequentially. Any BLAST alignment between a sequence and a consensus sequence with a score greater than 150 was realigned using cross-match. The sequence was added to the bin whose consensus sequence gave the highest Smith-Waterman score (Smith et al. (1992) Protein Engineering 5:35-51) amongst local alignments with at least 82% identity. Non-matching sequences were moved into new bins, and assembly processes were repeated. [0091]
  • IV Homology Searching of Polynucleotides and their Encoded Proteins [0092]
  • The polynucleotides of the Sequence Listing or their encoded proteins 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 BLAST 2 (Altschul et al. supra; Altschul, supra) to produce alignments and to determine which sequences were exact matches or homologs. The alignments 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. All of the sequences disclosed in this application have lengths of at least 49 nucleotides, and no more than 12% uncalled bases (where N is recorded rather than A, C, G, or T). [0093]
  • As detailed in Karlin (supra), BLAST matches between a query sequence and a database sequence were evaluated statistically and only reported when they satisfied the threshold of 10[0094] −25 for nucleotides and 10−14 for peptides. Homology was also evaluated by product score calculated as follows: the % nucleotide or amino acid identity [between the query and reference sequences] in BLAST is multiplied 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 electronic stringency for an exact match was set at 70, and the conservative lower limit for an exact match was set at approximately 40 (with 1-2% error due to uncalled bases).
  • The BLAST software suite, freely available sequence comparison algorithms (NCBI, Bethesda Md.; http://www.ncbi.nlm.nih.gov/gorf/bl2.html), includes various sequence analysis programs including “blastn” that is used to align nucleic acid molecules and BLAST 2 that is used for direct pairwise comparison of either nucleic or amino acid molecules. 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 or similarity is measured over the entire length of a sequence or some smaller portion thereof. Brenner et al. (1998; Proc Natl Acad Sci 95:6073-6078, incorporated herein by reference) analyzed the BLAST for its ability to identify structural homologs by sequence identity and found 30% identity is a reliable threshold for sequence alignments of at least 150 residues and 40%, for alignments of at least 70 residues. [0095]
  • The polynucleotides of this application were compared with assembled consensus sequences or templates found in the LIFESEQ GOLD database. Component sequences from cDNA, extension, full length, and shotgun sequencing projects were subjected to PHRED analysis and assigned a quality score. All sequences with an acceptable quality score were subjected to various pre-processing and editing pathways to remove low quality 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. [0096]
  • 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 quality score greater than or equal to 150 and an alignment 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. [0097]
  • 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 split. Templates were subjected to analysis by STITCHER/EXON MAPPER algorithms that analyze the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced genes across tissue types or disease states, and the like. Assembly procedures were repeated periodically, 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 probability score) of ≦1×10[0098] −8. The templates were also subjected to frameshift FASTx against GENPEPT, and homolog match was defined as having an E-value of ≦1×10−8. Template analysis and assembly was described in U.S. Ser. No. 09/276,534, filed Mar. 25, 1999.
  • Following assembly, templates were subjected to BLAST, motif, and other functional analyses and categorized in protein hierarchies using methods described in U.S. Ser. No. 08/812,290 and U.S. Ser. No. 08/811,758, both filed Mar. 6, 1997; in U.S. Ser. No. 08/947,845, filed Oct. 9, 1997; and in U.S. Ser. No. 09/034,807, filed Mar. 4, 1998. Then templates were analyzed by translating each template in all three forward reading frames and searching each translation against the PFAM database of hidden Markov model-based protein families and domains using the HMMER software package (Washington University School of Medicine, St. Louis Mo.; http://pfam.wustl.edu/). [0099]
  • The polynucleotide was further analyzed using MACDNASIS PRO software (Hitachi Software Engineering), and LASERGENE software (DNASTAR) and queried against public databases such as the GenBank rodent, mammalian, vertebrate, prokaryote, and eukaryote databases, SwissProt, BLOCKS, PRINTS, PFAM, and Prosite. [0100]
  • V Description of Genes Known to be Associated with Inflammatory Disorders [0101]
  • Eleven known inflammation genes were selected to identify novel genes that are closely associated with inflammation. These known genes, CD16, L-selectin, SLAP, IP-30, p67phox, p47phox, p40phox, AAT, Clq-A, FLAP, and HCK are described below. [0102]
    Gene Description & References
    CD16 receptor for IgG a.k.a. FcgammaRIII; phagocytosis of complement-generated immune
    complexes occurs through CD16 (Tamm et al. (1996) J Biol Chem 271:3659-66;
    Marsh et al. (1998) J Immunol 160:3942-8)
    L-selectin Leukocyte adhesion molecule, binds carbohydrate ligand on endothelial cell
    glycoprotein Adhesion is required for extravasation near inflammation site; auxiliary function in
    neutrophil activation during inflammation (Frohlich et al. (1998) Blood 91:2558-64;
    Girard and Amalric (1998) Adv Exp Med Biol 435:55-62)
    SLAP Src-like adapter protein; associated with Eck RPTK transduction pathway; Eck
    RPTK autocrine loop implicated in inflammation (Pandey et al. (1995) J Biol Chem
    270: 19201- 4; Pandey et al. (1995) Science 268:567-9)
    IP-30 Functions in MHC Class II processing of peptides, implicated in inflammation and α-
    interferon inducible (Luster et al. (1988) J Biol Chem 263:12036-43; Arunachalam et
    al. (1998) J Immunol 160:5797-806; Schuelke et al. (1998) Biochem Biophys Res
    Commun 245:599-606)
    p67phox superoxidase holoenzyme subunits; macrophages utilize reactive superoxide in
    p47phox degradation of phagocytosed matter; phox subunits induced by complement during
    p40phox inflammation (Ratnam and Mookerjea (1998) Immunology 94:560-568)
    AAT alpha-1-antitrypsin inhibits trypsin, a protease; differentially expressed in
    inflammation Several alleles linked to chronic inflammatory disorders; (Breit and Penny (1980)
    Aust N ZJ Med 10:449-53; Takeuchi et al. (1984) Int J Tissue React 6:1-8)
    C1q-A First complement component, subcomponent q, subunit A (Alberts et al. (1994)
    Molecular Biology of the Cell, Garland Publishing, New York NY, p. 1214)
    FLAP 5-lipoxygenase activating protein; lipoxygenase enzyme catalyzes formation of
    leukotrienes which are potent inflammatory mediators. FLAP is an anti-
    inflammatory therapeutic target (Byrum et al. (1997) J Exp Med 185:1065-75; Muller-
    Peddinghaus (1997) J Physiol Pharmacol 48:529-36)
    HCK Src-family tyrosine kinase specific to hematopoietic cells; functions in integrin
    signaling Mouse knockouts have impaired inflammation response (Lowell and Berton (1998)
    Proc Natl Acad Sci 95:7580-84)
  • VI Co-Expression Among Known Marker Genes and Novel Polynucleotides [0103]
  • The co-expression of the 11 known genes with each other is shown below. The entries are the negative log of the p-value (−log p) for the co-expression of the two genes. As shown, co-expression analysis successfully identified the strong association among the known genes which indicates that co-expression analysis was effective in identifying genes that are highly associated with inflammation and inflammatory disorders. The degree of association was measured by probability values and the cutoff p-value used in this analysis was less than 0.00001. [0104]
  • Using the LIFESEQ GOLD database (Incyte Genomics), the method also identified polynucleotides from among a total of 41,419 assembled sequences that show strong association with the known genes. The process was reiterated until the number of polynucleotides was reduced to the final eleven polynucleotides shown below. The following tabular entries show the negative log of the p-value (−log p) for the co-expression among the known marker genes and the novel polynucleotides. The novel polynucleotides are identified in the table below by their Incyte clone numbers and the known genes their abbreviated names as shown in Example IV above. For each polynucleotide, the p-value is the probability that the observed co-expression is due to chance, using the Fisher Exact Test. [0105]
    Gene or SEQ \Gene or clone
    L-sel-
    ID NO (clone) CD16 ectin SLAP IP-30 p67-phox AA p47-phox ClqA p40-phox FLAP HC 1221361 3055142 402234
    CD16
    L-selectin 4
    SLAP 5 6
    IP-30 5 3 4
    P67-phox 3 5 7 13
    AAT 6 2 4 10 5
    P47-phox 5 6 5 14 7 3
    ClqA 7 2 6 11 3 7 6
    P40-phox 5 5 3 4 7 4 6 1
    FLAP 7 5 3 11 5 3 5 7 3
    HCK 9 8 4 5 8 5 7 3 6 3
     4 (1221361) 3 5 7 4 3 1 4 2 4 2 4
    10 (3055142) 6 5 9 4 5 3 8 1 4 3 8 4
     1 (402234) 3 6 4 4 2 0 6 1 3 4 2 3 1
    11 (3507924) 2 5 3 4 4 8 4 1 7 3 2 3 2
     5 (1335016) 7 3 3 8 2 2 5 6 3 4 3 4 5
     9 (3054032) 5 11 9 5 4 4 7 7 7 8 7 5 7
     2 (569989) 4 4 4 6 9 1 8 2 4 5 4 4 4
     6 (2349263) 8 1 2 6 4 4 3 8 5 5 6 3 3
     7 (2471716) 3 5 3 7 5 1 7 4 5 2 4 3 5
     8 (2726173) 3 8 5 4 6 2 7 0 2 3 5 4 3
     3 (706377) 4 5 5 8 6 5 10 3 6 3 3 4 4
    Gene or SEQ \Gene or clone
    ID NO (clone) 3507924 1335016 3054032 569989 2349263 2471716 2726173 706377
    CD16
    L-selectin
    SLAP
    IP-30
    P67-phox
    AAT
    p47-phox
    ClqA
    P40-phox
    FLAP
    HCK
     4 (1221361)
    10 (3055142)
     1 (402234)
    11 (3507924) 4
     5 (1335016) 2 2
     9 (3054032) 5 4 4
     2 (569989) 7 2 2 5
     6 (2349263) 1 3 9 5 4
     7 (2471716) 4 5 4 8 3 7
     8 (2726173) 4 4 2 3 3 2 5
     3 (706377) 9 4 5 5 7 3 6 3
  • The highest co-expression value is obtained when the highest (−log p) value found along the horizontal line following each SEQ ID NO (clone number) are correlated with a known marker gene (abbreviation along the top line of the chart). For example Another look at the data above simplified by reducing it to a single highest co-expression (−log p) and naming at least one inflammatory disorder for each polynucleotide is shown below: [0106]
    Gene SEQ ID NO p-value* Inflammatory Disorder
    CD16 6 8 rheumatoid arthritis
    L-selectin 1 6 BPH
    L-selectin 8 8 asthma
    L-selectin 9 11 cholecystitis
    SLAP 4 7 chronic heart failure (CHF)
    SLAP 10 9 bacterial infection, toxic shock
    IP-30 5 8 chronic inflammation of bowel
    IP-30 7 7 immune response
    P67-phox 2 9 hypereosinophilia
    AAT 11 8 asthma
    P47-phox 3 10 lung cancer and complications
  • VII Novel Polynucleotides Associated with Inflammatory Disorders [0107]
  • Eleven polynucleotides were found to be associated with known genes that are diagnostic markers for inflammation and inflammatory disorders. The polynucleotides comprising the nucleic acid sequences of SEQ ID NOs:1-11 were first identified as Incyte Clones 402234, 569989, 706377, 1221361, 1335016, 2349263, 2471716, 2726173, 3054032, 3055142, and 3507924, respectively. These sequences were assembled according to the procedures described in Example IV. BLAST and motif searches were performed for SEQ ID NOs: 1-11 and SEQ ID NOs: 12-17 according to Example V. Proteins or peptides comprising the amino acid sequences of SEQ ID NOs: 12-17 were encoded by the nucleic acids of SEQ ID NO: 1, 2, 6, 7, 8, and 11, respectively. [0108]
  • SEQ ID NO:3 is 1229 nucleotides in length and shares about 99% sequence identity from about nucleotide 250 to about nucleotide 1216 with a human basement membrane-induced gene identified in a human endometrial adenocarcinoma cell line (g3132521). [0109]
  • SEQ ID NO:4 is about 1261 nucleotides in length and has about 34% sequence identity from nucleotide 23 to nucleotide 994 with a sequence similar to a RNA recognition motif (g2645068). [0110]
  • SEQ ID NO:5 is 1340 nucleotides in length and has about 60% sequence identity from about nucleotide 21 to about nucleotide 925 with a human prostaglandin transporter hPGT mRNA (g3006201). The protein encoded by SEQ ID NO:5 exhibits several potential transmembrane domains identified by HMM analysis. [0111]
  • SEQ ID NO:9 is 2309 nucleotides in length and shows sequence identity from about nucleotide 104 to about nucleotide 785 with a human polycystic kidney disease-associated protein gene (g790818). [0112]
  • SEQ ID NO:12 is 127 amino acid residues in length and shows about 50% sequence identity from about residue 37 to about residue 106 with a tobacco LIM-domain-containing protein (g181464). The LIM domain is a cysteine-rich, zinc-binding motif of about 60 amino-acid residues that plays a potential role in DNA binding and regulation (Perez-Alvarado et al. (1994) Nat Struct Biol 1:388-398). PFAM analysis shows that residues 40 to 97 of SEQ ID NO: 12 align with and encompass the LIM domain. [0113]
  • SEQ ID NO: 13 is 93 amino acid residues in length and has a potential signal peptide from residue 1 to residue 18. SEQ ID NO: 13 also exhibits a potential transmembrane domain from about residues 47 to residue 69. [0114]
  • SEQ ID NO: 14 is 225 amino acid residues in length and has about 32% sequence identity from about residue 5 to about residue 135 with a mouse high affinity IgE receptor beta subunit (g309225). [0115]
  • SEQ ID NO: 15 is 547 amino acid residues in length and has about 35% sequence identity from about residue 413 to about 546 with a rat beta-chimaerin, a GTPase-activating protein expressed exclusively in the testis at the onset of sexual maturation (g203117). PFAM analysis shows that SEQ ID NO: 15 has sequence homology from about residue 353 to about residue 523 with the GTPase-activator protein for Rho-like GTPases. [0116]
  • SEQ ID NO: 16 is 265 amino acid residues in length and shows about 93% sequence identity from about residue 39 to about residues 265 with Maxp1, a rat protein which interacts with Mss4, a guanine nucleotide exchange factor (g2459833), and about 91% sequence identity from about residue 38 to about residue 265 with Nore1, a mouse putative Ras effector that plays a role in transmitting growth and differentiation signals received from Ras proteins (g2997698). PFAM analysis confirms that SEQ ID NO: 16 from about residue 119 to about residue 211 matches a Ras association domain which interacts directly with the Ras proteins. [0117]
  • SEQ ID NO: 17 is 394 amino acid residues in length and exhibits a potential signal peptide sequence from about residue 1 to residue 19 and a potential transmembrane domain from about residues 273 to residue 295. [0118]
  • VIII Hybridization Technologies and Analyses [0119]
  • Immobilization of Polynucleotides on a Substrate [0120]
  • The polynucleotides are applied to a substrate by one of the following methods. A mixture of polynucleotides is fractionated by gel electrophoresis and transferred to a nylon membrane by capillary transfer. Alternatively, the polynucleotides are individually ligated to a vector and inserted into bacterial host cells to form a library. The polynucleotides are then arranged on a substrate by one of the following methods. In the first method, bacterial cells containing individual clones are robotically picked and arranged on a nylon membrane. The membrane is placed on LB agar containing selective agent (carbenicillin, kanamycin, ampicillin, or chloramphenicol depending on the vector used) and incubated at 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), neutralizing solution (1.5 M NaCl, 1 M Tris-HCl, pH 8.0), and twice in 2×SSC for 10 min each. The membrane is then UV irradiated in a STRATALINKER UV-crosslinker (Stratagene). [0121]
  • In the second method, polynucleotides are amplified from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR amplification increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 μg. Amplified nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are arranged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobilized by denaturation, neutralization, and UV irradiation as described above. Purified nucleic acids are robotically arranged and immobilized on polymer-coated glass slides using the procedure described in U.S. Pat. No. 5,807,522. Polymer-coated slides are prepared by cleaning glass microscope slides (Corning, Acton Mass.) 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 slides are washed extensively with distilled water between and after treatments. The nucleic acids are arranged on the slide and then immobilized by exposing the array to UV irradiation using a STRATALINKER UV-crosslinker (Stratagene). Arrays are then washed at room temperature in 0.2% SDS and rinsed three times in distilled water. Non-specific binding sites are blocked by incubation of arrays in 0.2% casein in phosphate buffered saline (PBS; Tropix, Bedford Mass.) for 30 min at 60C.; then the arrays are washed in 0.2% SDS and rinsed in distilled water as before. [0122]
  • Probe Preparation for Membrane Hybridization [0123]
  • Hybridization probes derived from the polynucleotides of the Sequence Listing are employed for screening cDNAs, mRNAs, or genomic DNA in membrane-based hybridizations. Probes are prepared by diluting the polynucleotides 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 polynucleotide is then added to a REDIPRIME tube (APB), gently mixed until blue color is evenly distributed, and briefly centrifuged. Five μl of [[0124] 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 Slide Hybridization [0125]
  • Hybridization probes derived from mRNA isolated from samples are employed for screening polynucleotides of the Sequence Listing in array-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 5×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 1×yeast control mRNAs. Yeast control mRNAs are synthesized by in vitro transcription from noncoding yeast genomic DNA (W. Lei, unpublished). 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 nin at 85C., and probes are purified using two successive CHROMA SPIN+TE 30 columns (Clontech, Palo Alto Calif.). Purified probe is ethanol precipitated by diluting probe to 90 μl in DEPC-treated water, adding 2 μl 1 mg/ml glycogen, 60 μl 5 M sodium acetate, and 300 μl 100% ethanol. The probe is centrifuged for 20 min at 20,800×g, and the pellet 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 array-based hybridizations as described below. [0126]
  • Membrane-Based Hybridization [0127]
  • Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and 1×high phosphate buffer (0.5 M NaCl, 0.1 M Na[0128] 2HPO4, 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. Following hybridization, the membrane is washed for 15 min at 25C. in 1 mM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 min each at 25C. in 1 mM Tris (pH 8.0). To detect hybridization complexes, XOMAT-AR film (Eastman Kodak, Rochester N.Y.) is exposed to the membrane overnight at −70C., developed, and examined visually.
  • Polymer Coated Slide-Based Hybridization [0129]
  • Probe is heated to 65C. for five min, centrifuged five min at 9400 rpm in a 5415C microcentrifuge (Eppendorf Scientific, Westbury N.Y.), and then 18 μl are aliquoted onto the array surface and covered with a coverslip. The arrays are transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber is kept at 100% humidity internally by the addition of 140 μl of 5×SSC in a corner of the chamber. The chamber containing the arrays is incubated for about 6.5 hr at 60C. The arrays are washed for 10 min at 45C. in 1×SSC, 0.1% SDS, and three times for 10 min each at 45C. in 0.1×SSC, and dried. [0130]
  • 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 correlates 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 array elements, and signals are examined under conditions in which the emissions from the two different labels are individually detectable. Elements on the array that are hybridized to equal numbers of probes derived from both biological samples give a distinct combined fluorescence (Shalon WO95/35505). [0131]
  • Hybridization complexes are detected with a microscope equipped with an INNOVA 70 mixed gas 10 W laser (Coherent, Santa Clara Calif.) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of Cy5. The excitation laser light is focused on the array using a 20× microscope objective (Nikon, Melville N.Y.). The slide containing the array is placed on a computer-controlled 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 sequentially excited by the laser. Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477, Hamamatsu Photonics Systems, Bridgewater N.J.) corresponding to the two fluorophores. Appropriate filters positioned between the array and the photomultiplier 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 calibrated using the signal intensity generated by the yeast control mRNAs added to the probe mix. A specific location on the array contains a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000. [0132]
  • The output of the photomultiplier tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood Mass.) installed in an IBM-compatible PC computer. The digitized data are displayed as an image where the signal intensity is mapped using a linear 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 corrected 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). [0133]
  • IX Transcript Imaging [0134]
  • The transcript images for SEQ ID NOs: 1, 2, 3, and 6 were performed using the LIFESEQ GOLD database (Incyte Genomics). The product score for sequences was calculated as follows: the BLAST score is multiplied 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% alignment over the length of the shorter sequence gives a product score of 100. A product score of 70, which assures an exact match, was the cut-off score for the transcript images. [0135]
  • All sequences and cDNA libraries in the LIFESEQ database were categorized by system, organ/tissue and cell type. The categories included cardiovascular system, connective tissue, digestive system, embryonic structures, endocrine system, exocrine glands, female and male reproductive, germ cells, hemic/immune system, liver, musculoskeletal system, nervous system, pancreas, respiratory system, sense organs, skin, stomatognathic system, unclassified/mixed, and the urinary tract. For each category, the number of libraries in which the sequence was expressed were counted and shown over the total number of libraries in that category. All normalized or pooled libraries, which have high copy number sequences removed prior to processing. All mixed or pooled tissues, which are considered non-specific in that they contain more than one tissue type or more than one subject's tissue, were excluded from this analysis. Cell lines and fetal tissues were generally not considered. [0136]
  • For purposes of example, the four transcript images below show independent confirmation of the results of the co-expression analysis. The transcript images demonstrate differential expression of SEQ ID NOs: 1, 2, 3 and 6, each in a different category, as produced using the LIFESEQ GOLD database (Incyte Genomics). [0137]
    SEQ ID NO:1 (Category: Male Reproductive)
    Library cDNAs Description Abundance % Abundance
    PROSDIP01 487 prostate, stroma, BPH, M, 3′ CGAP 3 0.6160
    PROSTUP02 869 prostate tumor, N, 3′ CGAP 1 0.1151
    PROSTUT01 3224 prostate tumor, adenoCA, 50 M 1 0.0310
    PROSNOT26 3695 prostate, mw/adenoCA, 65 M 1 0.0271
  • SEQ ID NO: I is differentially expressed in benign prostate hyperplasia. Expression exceeds that of any other prostate library, including tumor and cytologically normal tissue, by greater than five-fold. [0138]
    SEQ ID NO:2 (Category: Hemic/Immune)
    Library cDNAs Description Abundance % Abundance
    EOSIHET02 9261 periph blood, hypereosinophilia, 48 M 2 0.0216
    EOSITXT01 8976 periph blood, eosinophils, t/IL-5 1 0.0111
  • SEQ ID NO:2 is differentially expressed in hypereosinophilia, even exceeding IL-5 activated expression by approximately two-fold. [0139]
    SEQ ID NO:3 (Category: Respiratory System)
    Library cDNAs Description Abundance % Abundance
    LUNGTUT13 3990 lung tumor, adenoCA, 47 M 2 0.0501
    LUNGNOT38 3447 lung, asthma, 15 M 1 0.0290
    LUNGNOT03 4959 lung, mw/mets thyroid CA, 79 M 1 0.0202
    LUNLTMT01 6668 lung, mw/adenoCA, aw/node mets, 63 F 1 0.0150
  • SEQ ID NO:3 is differentially expressed in adenocarcinoma of the lung. Expression exceeds that seen in asthmatic and cytologically normal lung by approximately two-fold. [0140]
    SEQ ID NO:6 (Category: Musculoskeletal System)
    Library cDNAs Description Abundance % Abundance
    SYNWDIT01 3232 synovium, wrist, dorsal, rheuA, 64 F 4 0.1238
    SYNORAB01 5140 synovium, hip, rheuA, 68 F 6 0.1167
    SYNORAT03 5814 synovium, wrist, rheuA, 56 F 6 0.1032
    SYNORAT04 5665 synovium, wrist, rheuA, 62 F 3 0.0530
  • SEQ ID NO:6 was differentially expressed in the synovium afflicted with rheumatoid arthritis. The sequence was not highly expressed in association with libraries with designated osteoarthritis or in cultured chrondrocytes. [0141]
  • X Complementary Molecules [0142]
  • The complement of the novel polynucleotide, from about 5 bp (e.g., a PNA) to about 5000 bp (e.g., the complement of a cDNA insert), are used to detect or inhibit gene expression. These molecules are selected using LASERGENE software (DNASTAR). Detection is described in Example VIII. 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 helix” base pairing to compromise the ability of the double helix 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. [0143]
  • 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 cell, zygote, or other reproducing lineage for long term or stable gene therapy. Transient expression lasts for a month or more with a non-replicating vector and for three months or more if appropriate elements for inducing vector replication are used in the transformation/expression system. [0144]
  • Stable transformation of appropriate dividing cells with a vector encoding the complementary molecule produces a transgenic cell line, tissue, or organism (U.S. Pat. No. 4,736,866). Those cells that assimilate and replicate sufficient quantities of the vector to allow stable integration also produce enough complementary molecules to compromise or entirely eliminate activity of the polynucleotide encoding the protein. [0145]
  • XI Protein Expression [0146]
  • Expression and purification of the protein are achieved using either a cell expression system or an insect cell expression system. The pUB6/V5-His vector system (Invitrogen, Carlsbad Calif.) is used to express protein 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 (6×His) sequence for rapid purification on PROBOND resin (Invitrogen). Transformed cells are selected on media containing blasticidin. [0147]
  • [0148] Spodoptera frugiperda (Sf9) insect cells are infected with recombinant Autographica californica nuclear polyhedrosis virus (baculovirus). The polyhedrin gene is replaced with the polynucleotide by homologous recombination and the polyhedrin promoter drives transcription. The protein is synthesized as a fusion protein with 6×his which enables purification as described above. Purified protein is used in the following activity and to make antibodies.
  • XII Production of Antibodies [0149]
  • The protein 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 the expressed protein is analyzed using LASERGENE software (DNASTAR) to determine regions of high antigenicity. An antigenic epitope, usually found near the C-terminus or in a hydrophilic region is selected, synthesized, and used to raise antibodies. Typically, epitopes of about 15 residues in length are produced using an ABI 431A peptide synthesizer (Applied Biosystems) using Fmoc-chemistry and coupled to KLH (Sigma-Aldrich) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester to increase antigenicity. [0150]
  • 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 well known in the art are used to determine antibody titer and the amount of complex formation. [0151]
  • XIII Purification of Naturally Occurring Protein Using Specific Antibodies [0152]
  • Naturally occurring or recombinant protein is purified by immunoaffinity chromatography using antibodies which specifically bind the protein. An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected. [0153]
  • XIV Screening Molecules for Specific Binding Using Polynucleotide or Protein [0154]
  • The polynucleotide, or fragments thereof, or the protein, or portions thereof, are labeled with [0155] 32P-dCTP, Cy3-dCTP, or Cy5-dCTP (APB), or with BIODIPY or FITC (Molecular Probes, Eugene Oreg.), respectively. Libraries of candidate molecules or compounds previously arranged on a substrate are incubated in the presence of composition, a labeled polynucleotide 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 ligand 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.
  • XV Two-Hybrid Screen [0156]
  • A yeast two-hybrid system, MATCHMAKER LexA Two-Hybrid system (Clontech Laboratories, Palo Alto Calif.), is used to screen for peptides that bind the protein of the invention. A polynucleotide encoding the protein is inserted into the multiple cloning site of a pLexA vector, ligated, and transformed into [0157] E. coli. cDNA, prepared from mRNA, is inserted into the multiple cloning site of a pB42AD vector, ligated, and transformed into E. coli to construct a cDNA library. The pLexA plasmid and pB42AD-cDNA library constructs are isolated from E. coli and used in a 2:1 ratio to co-transform competent yeast EGY48[p8op-lacZ] cells using a polyethylene glycol/lithium acetate protocol. Transformed yeast cells 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 1x TE (pH 7.5), replated on SD/-His/-Leu/-Trp/-Ura 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 liquid 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 replica-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 pLexA 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 polynucleotide encoding a protein that physically interacts with the protein, is isolated from the yeast cells and characterized. [0158]
  • All patents and publications mentioned in the specification are incorporated by reference herein. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the field of molecular biology or related fields are intended to be within the scope of the following claims. [0159]
  • 1 17 1 1298 DNA Homo sapiens 402234CB1 1 gggacatgac ttagggaaag tcccaaccgg aatcccccca gcccctgcct gtcaacaccc 60 cccaccctgc aggctggggc cgggctggcg gggccctccc gactgacttc cccttgcaga 120 acccagcggg tgccgcttct ccacccgagg cttccacctc caacgagcca tgttccaggc 180 tgcaggagcc gcccaggcca ccccctctca tgacgccaaa ggcggcggca gcagcacggt 240 gcagcgctcc aagtccttca gcctgcgggc ccaggtgaag gagacctgcg ccgcctgcca 300 gaagaccgtg taccccatgg agcggctggt ggccgacaag ctcattttcc acaactcttg 360 cttctgctgc aagcactgtc acaccaagct cagcctgggc agctacgccg cgctgcacgg 420 ggagttctac tgcaaacccc acttccagca gctgtttaag agcaaaggca actacgacga 480 ggggtttggc cgcaagcagc acaaggagct ctgggcccac aaggaggtgg accccggcac 540 caagacggcc tgaggcctct gtaaccttcc accccctctg cggaaggcct ggagccggca 600 gggggaaggt gggaaggagg tcgagctggg cttgcgtggg ggccaggtgg gaaggggatg 660 aggcttgctc aggcgtaggg gaccagggca gggctctgct ccaggactcc ttccttcttc 720 cttctcccgc agccggtgag ggtttggaaa ccaggattgg ggtctgccca ccaccctgct 780 tcctgcttcg ttcagcctcc ctccccacct caccccagga ccccctggga ggcccccaag 840 cccagctccc ctatctaggt gccttttctc cagcaaggag tcagcatgcc cccctcaggg 900 tcccaagctc cctcactgcc accggagact gtgtggcccc cacgtctccc catctacctc 960 tacccttaac ctgtttctga gccacggaga cagggaggaa ggagcgcgac agtgccacct 1020 gttgggcatc ataaatgccc ctgcagccca tgggggagga gatggggaag tggagccacc 1080 ctgcctctgc agggcaaggc agggcctgcc ccagtggggc ttgggaccat ctcgaaccac 1140 cagcgtggag aagcagaagc aaaagcactc gccaggctgc agcctcaggc actggcaggg 1200 gctggtgcgg ccccactccc ctcccccgct cccatttgtg cccatcctgt tgtgaccaac 1260 cccgttttaa acatgtttca atagatccaa aaaaaaaa 1298 2 532 DNA Homo sapiens 569989CB1 2 cccacgcgtc cgccttgaca ccagcagggt gacatccgct attgctactt ctctgctccc 60 ccacagttcc tctggacttc tctggaccac agtcctctgc cagacccctg ccagacccca 120 gtccaccatg atccatctgg gtcacatcct cttcctgctt ttgctcccag tggctgcagc 180 tcagacgact ccaggagaga gatcatcact ccctgccttt taccctggca cttcaggctc 240 ttgttccgga tgtgggtccc tctctctgcc gctcctggca ggcctcgtgg ctgctgatgc 300 ggtggcatcg ctgctcatcg tgggggcggt gttcctgtgc gcacgcccac gccgcagccc 360 cgcccaagaa gatggcaaag tctacatcaa catgccaggc aggggctgac cctcctgcag 420 cttggacctt tgacttctga ccctctcatc ctggatggtg tgtggtggca caggaacccc 480 cgccccaact tttggattgt aataaaacaa ttgaaacacc aaaaaaaaaa aa 532 3 1229 DNA Homo sapiens 706377CB1 3 cagagagccg cggggaccat ggagccggtg ccgctgcagg acttcgtgcg cgccttggac 60 cccgcctccc tcccgcgcgt gctgcgggtc tgctcggggg tctacttcga gggctccatc 120 tatgagatct ctgggaatga gtgctgcctc tccacggggg acctgatcaa ggtcacccag 180 gtccgcctcc agaaggtggt ctgtgagaac ccgaagacca gccagaccat ggagctcgcc 240 cccaacttcc aggtcttctc aagtcttagg attgcagcaa cacgctcggc tgcccaaacc 300 caaggcgaag accttgccag agttcatcaa ggatggctcc agtacgtaca gcaagattcc 360 tgcccacagg aagggccaca ggccgctaag ccccaaaggc aggatctaga tgatgatgaa 420 catgattatg aagaaatact tgagcaattt cagaaaacca tctaagtgct ggaggaacca 480 cgcttcctaa ctgctgcttc tcagggaatc cgacaccagc caaccatttt aagcctctaa 540 aagacctcgg gcaagtctca cagaaactga gctgcagacg gggagtagct ttgtggaaac 600 tgatttgatg gacactgcac cagcttcctt caggttctag attcttgcta cttagggcgg 660 gctggtttgg acctaacatc tcgcacgtga ctccctcagc ctcagagcct tgggatgcag 720 agcagctggc agggttcctc tcaatcctgc aaccccagct gtcccaccgg tggatgcaga 780 ggggaatccg aggccatcaa ccttggtgac agcagcgcag tgccaatgct gatcacactg 840 catgggagat tttgttaacg tctgccaccc ccactctcac ccccaagctc taagcccccg 900 ggaggcctgg actgtcttcc tcatctctgt agcaccaagc ctgatagatc tgtatatggt 960 aaacaggggt ttaaccacat gtggttaaca tggattaatg tgggaacttg gcttcaagaa 1020 cacaacctta ggaccttggg ccccaaaagc tggtggtgaa atgaggagga gccaatttaa 1080 gaagaccctt atggagacct gaggctgcag aaactggtag gtttcatcag gtggttaaag 1140 tcgtcaaagt tgtaagtgac taaccaagat tatttcattt taaaaccata gaataaaaat 1200 gacacctgag cttctctaaa aaaaaaaaa 1229 4 1261 DNA Homo sapiens 1221361CB1 4 cggacgcgtg ggcggacgcg tggcggacgc gtgggcggac gcgtgggcgc gatgggcctc 60 ttggaacatt ggtgtgttca tctgcattcg atgtgctgga atccacagga atctgggggt 120 gcacatatcc agggtaaagt cagttaacct cgaccagtgg actcaagaac agattcagtg 180 catgcaagag atgggaaatg gaaaggcaaa ccgactttat gaagcctatc ttcctgagac 240 ctttcggcga cctcagatag acccagctgt tgaaggattt attcgagaca aatatgagaa 300 gaagaaatac atggaccgaa gtctggacat caatgccttt aggaaagaaa aagatgacaa 360 gtggaaaaga gggagcgaac cagttccaga aaaaaaattg gaacctgttg tttttgagaa 420 ggtgaaaatg ccacagaaaa aagaagaccc acagctacct cggaaaagct ccccgaaatc 480 cacagcgcct gtcatggatt tgttgggcct tgatgctcct gtggcctgct ccattgcaaa 540 tagtaagacc agcaataccc tagagaagga tttagatctg ttggcctctg ttccatcccc 600 ttcttcttcg ggttccagaa aggttgtagg ttccatgcca actgcaggga gtgccggctc 660 tgttcctgaa aatctgaacc tgtttccgga gccagggagc aaatcagaag aaataggcaa 720 gaaacagctc tctaaagact ccattctttc actgtatgga tcccagacgc ctcaaatgcc 780 tactcaagca atgttcatgg ctcccgctca gatggcatat cccacagcct accccagctt 840 ccccggggtt acacctccta acagcataat ggggagcatg atgcctccac cagtaggcat 900 ggttgctcag ccaggagctt ctgggatggt tgcccccatg gccatgcctg caggctatat 960 gggtggcatg caggcatcaa tgatgggtgt gccgaatgga atgatgacca cccagcaggc 1020 tggctacatg gcaggcatgg cagctatgcc ccagactgtg tatggggtcc agccagctca 1080 gcagctgcaa tggaacctta ctcagatgac ccagcagatg gctgggatga acttctatgg 1140 agccaatggc atgatgaact atggacagtc aatgagtggc ggaaatggac aggcagcaaa 1200 tcagactctc agtcctcaga tgtggaaata aaaacaaaac accttgtata aaaaaaaaaa 1260 a 1261 5 1340 DNA Homo sapiens 1335016CB1 5 ccctcggaat tcggctcgag cagcaactcg cccctctacc tcgggatcct gtttgcagtg 60 accatgatgg ggccaggcct ggcctttggg ctgggcagcc tcatgctgcg cctttatgtg 120 gacattaacc agatgccaga aggtggtatc agcctgacca taaaggaccc ccgatgggtg 180 ggtgcctggt ggctgggttt cctcatcgct gccggtgcag tggccctggc tgccatcccc 240 tacttcttct tccccaagga aatgcccaag gaaaaacgtg agcttcagtt tcggcgaaag 300 gtcttagcag tcacagactc acctgccagg aagggcaagg actctccctc taagcagagc 360 cctggggagt ccacgaagaa gcaggatggc ctagtccaga ttgcaccaaa cctgactgtg 420 atccagttca ttaaagtctt ccccagggtg ctgctgcaga ccctacgcca ccccatcttc 480 ctgctggtgg tcctgtccca ggtatgcttg tcatccatgg ctgcgggcat ggccaccttc 540 ctgcccaagt tcctggagcg ccagttttcc atcacagcct cctacgccaa cctgctcatc 600 ggctgcctct ccttcccttc ggtcatcgtg ggcatcgtgg tgggtggcgt cctggtcaag 660 cggctccacc tgggccctgt gggatgcggt gccctttgcc tgctggggat gctgctgtgc 720 ctcttcttca gcctgccgct cttctttatc ggctgctcca gccaccagat tgcgggcatc 780 acacaccaga ccagtgccca ccctgggctg gagctgtctc caagctgcat ggaggcctgc 840 tcctgcccat tggacggctt taaccctgtc tgcgacccca gcactcgtgt ggaatacatc 900 acaccctgcc acgcaggctg ctcaagctgg gtggtccagg atgctctgga caacagccag 960 agtcctccca cctcccaccc tcatgctggg catcagcatc taaacctgag gctcctccag 1020 ggagagacct gggctgcact ggctggtgca gaagaacctg ttgatggtgc atagtccttc 1080 agaagccagc caggcaccac ctgggcctga gagcccttcc agagaccccc aggccttggc 1140 aggtggagca gtgaactcct gtggatatgg gaaccgattc aaatccttct taggcctcta 1200 actgactctg ttaccttagg caaattattt aactagtgcc tcagtttctt ggtctgtaaa 1260 ataggggaga tattattaag tgcctactac agagcaggaa tgtgctgaat aaatgcttta 1320 cctggatgaa aaaaaaaaaa 1340 6 2192 DNA Homo sapiens 2349263CB1 6 ttctttctct atattatgat tactagcact attactgtta ttagttacat gttattgaaa 60 gcttcaaagc agcataggct ttttataaat atttttgctc atctttatga caattctcca 120 gtgttggtat tgctcctcta tttaacagat tagaaaactg aagcttcaag aacagacttg 180 cctaacaaca ggaaacttgt atgtctcgaa gtggcaattc acacataagg ctccatgact 240 cctgaactct cacaaatatt agttggctct tttcatggtt ttactgaagt tgctagaagt 300 ttacagaaaa ggaagtgcag gaacatttca caaatctaca atctgtgagt atcacatcct 360 gtatagctgt aaacactgga ataaggaagg gctgatgact ttcagaagat gaaggtaagt 420 agaaaccgtt gatgggactg agaaaccaga gttaaaacct ctttggagct tctgaggact 480 cagctggaac caacgggcac agttggcaac accatcatga catcacaacc tgttcccaat 540 gagaccatca tagtgctccc atcaaatgtc atcaacttct cccaagcaga gaaacccgaa 600 cccaccaacc aggggcagga tagcctgaag aaacatctac acgcagaaat caaagttatt 660 gggactatcc agatcttgtg tggcatgatg gtattgagct tggggatcat tttggcatct 720 gcttccttct ctccaaattt tacccaagtg acttctacac tgttgaactc tgcttaccca 780 ttcataggac cctttttttt tatcatctct ggctctctat caatcgccac agagaaaagg 840 ttaaccaagc ttttggtgca tagcagcctg gttggaagca ttctgagtgc tctgtctgcc 900 ctggtgggtt tcattatcct gtctgtcaaa caggccacct taaatcctgc ctcactgcag 960 tgtgagttgg acaaaaataa tataccaaca agaagttatg tttcttactt ttatcatgat 1020 tcactttata ccacggactg ctatacagcc aaagccagtc tggctggaac tctctctctg 1080 atgctgattt gcactctgct ggaattctgc ctagctgtgc tcactgctgt gctgcggtgg 1140 aaacaggctt actctgactt ccctggggtg agtgtgctgg ccggcttcac ttaaccttgc 1200 ctagtgtatc ttatccctgc actgtgttga gtatgtcacc aagagtggta gaaggaacaa 1260 ccagccaatc acgagataca catgggaggg catttgcatt gtgatggaag acagagaaga 1320 aaagcagatg gcaattgagt agctgataag ctgaaaattc actggatatg aaaatagtta 1380 atcatgagaa atcaactgat tcaatcttcc tattttgtca gcgaagggaa tgagactctg 1440 ggaagttaaa tgactggcct ggcattatgc tatgagtttg tgcctttgct gaggacacta 1500 gaacctggct tgcctccctt ataagcagaa acaatttctg ccacaaccac tagtctcttt 1560 aatagtattg acttggtaaa gggcatttac acacgtaact ggatccagtg aatgtcttat 1620 gctctgcatt tgcccctggt gatcttaaaa ttcgtttgcc tttttaaagc tatattaaaa 1680 atgtattgtt gaatcaaacc cctatggact tattgcttta tttaactgaa ttaaaaagcc 1740 ttgatttatc caaaattgta ttatagagtg tagaatgaat actagggtga taaattgcaa 1800 ttatttgaag aacctggtga tatgctctac ttatcttgga ttagctaaga attctatgta 1860 tacagttgga aaaatggcat atatacatct atcttgaacc tgattgaagt ctgaagacct 1920 aacatatttt gtttcttcta gagtgtactt ttcctgcctc acagttacat tggtaattct 1980 ggcatgtcct caaaaatgac tcatgactgt ggatatgaag aactattgac ttcttaagaa 2040 aaaagggaga aatattaatc agaaagttga ttcttatgat aatatggaaa agttaaccat 2100 tatagaaaag caaagcttga gtttcctaaa tgtaagcttt taaagtaatg aacattaaaa 2160 aaaaccatta tttcactgtc aaaaaaaaaa aa 2192 7 1992 DNA Homo sapiens 2471716CB1 7 agaaaactgt gagagagaga atttttaaaa agcagctggg gcctgaggtt tctcccccag 60 taccctgggt cacctcagcc cagagctggc ggcaggcccc cagcccctca tgtcagagcc 120 ccctgtgtac tgtaacctgg tggaccttcg ccgctgtcct cggtccccac ccccaggccc 180 tgcatgcccc ctgctgcaga ggctggatgc ctgggagcag cacctggacc ccaactctgg 240 acgctgcttc tacataaatt cactgactgg ctgcaagtcc tggaagcccc cgcgccgcag 300 tcgcagcgag acgaaccctg gctccatgga ggggacacag accctgaaga ggaacaatga 360 tgtcctgcaa cctcaggcaa agggcttcag atctgacaca gggaccccag aaccgcttga 420 cccacagggt tcactcagcc tcagccaacg cacctcgcag cttgaccctc cagccttgca 480 ggcccctcga cctctgccgc agctcctgga cgacccccat gaggtggaaa agtcgggtct 540 gctcaacatg accaagattg cccaaggggg gcgcaagctc aggaagaact ggggcccgtc 600 ttgggtggtg ttaacgggta acagcctggt gttctaccga gagccaccgc cgacagcgcc 660 ctcctcaggc tggggaccag cgggtagccg gcccgaaagt agcgtggacc tgcgcggggc 720 ggccctggcg cacggccgcc acctgtccag ccgccgcaac gtcctgcaca tccgcacgat 780 ccctggccac gagttcctgc tgcagtcgga ccacgagaca gagctgcgag cctggcaccg 840 cgcgctgcgg actgtcatcg agcggctgga tcgggagaac cccctggagc tgcgtctgtc 900 gggctctgga cccgcggagc tgagcgccgg ggaggacgaa gaagaggagt cggagctggt 960 gtccaagccg ctgctgcgcc tcagcagccg ccggagctcc attcgggggc ccgaaggcac 1020 cgagcagaac cgcgtgcgca acaaactaaa gcggctcatc gcgaagagac cgcccttaca 1080 aagcctgcag gagcggggtc tgctccgaga ccaggtgttc ggctgccagt tggaatcact 1140 ctgccagcgg gaaggagaca cggtgcccag ctttttgcgg ctctgcattg ctgctgtgga 1200 taaaagaggt ctagatgtgg atggcattta tcgggtgagc gggaacttgg cagtggtcca 1260 gaagcttcgc tttctggtgg acagagagcg tgcggtcacc tccgatggga ggtatgtgtt 1320 cccagaacag ccaggacaag aaggtcggtt agatttggac agtactgagt gggatgacat 1380 tcatgtggtc accggagccc tgaagctttt tctccgggag ctgccccagc ctctggtgcc 1440 accactgctg ctgccccatt tccgtgctgc ccttgcactc tccgaatcag agcagtgcct 1500 ctctcagata caagaattaa taggctcaat gccaaagccc aaccatgaca ctctacggta 1560 cctcctggag catttatgca gggtgatagc acactcagat aagaatcgca tgacacccca 1620 caacctggga attgtgtttg gaccaaccct gtttcggcca gagcaggaga catctgaccc 1680 agcagcccat gctctctacc cagggcagct ggtccagctg atgctcacca acttcaccag 1740 cctcttcccc tgatgcaggg aaggaagaag agaaaacata tttccggtca tctctggtgg 1800 tgagaggctg gtgttctgtt ttgaggatat ccctttaaat ctcccaaatg actgtctcta 1860 tcttcatgag tgtgacttga ggtgttggga tgggtgaggg agcttctcta aagaggaaag 1920 tgagtggatt aacccctgct tctcttcttg ttccctgtta tcattcctcc ccgaacataa 1980 taatacataa gt 1992 8 3144 DNA Homo sapiens 2726173CB1 8 ccttgatgcg ctggcggcct cggccgggaa ctccggggta gatgaccgtg gacagcagca 60 tgagcagtgg gtactgcagc ctggacgagg aactggaaga ctgcttcttc actgctaaga 120 ctaccttttt cagaaatgcg cagagcaaac atctttcaaa gaatgtctgt aaacctgtgg 180 aggagacaca gcgcccgccc acactgcagg agatcaagca gaagatcgac agctacaaca 240 cgcgagagaa gaactgcctg ggcatgaaac tgagtgaaga cggcacctac acgggtttca 300 tcaaagtgca tctgaaactc cggcggcctg tgacggtgcc tgctgggatc cggccccagt 360 ccatctatga tgccatcaag gaggtgaacc tggcggctac cacggacaag cggacatcct 420 tctacctgcc cctagatgcc atcaagcagc tgcacatcag cagcaccacc accgtcagtg 480 aggtcatcca ggggctgctc aagaagttca tggttgtgga caatccccag aagtttgcac 540 tttttaagcg gatacacaag gacggacaag tgctcttcca gaaactctcc attgctgacc 600 gccccctcta cctgcgcctg cttgctgggc ctgacacgga ggtcctcaac tttgtgctaa 660 aggagaatga aactggagag gtagagtggg atgccttctc catccctgaa cttcagaact 720 tcctaacaat cctggaaaaa gaggagcagg acaaaatcca acaagtgcaa aagaagtatg 780 acaagtttag gcagaaactg gaggaggcct taagagaatc ccagggcaaa cctgggtaac 840 cggtcctgct tcctctcctc ctggtgcatt cagatttatt tgtattatta attattattt 900 tgcaacagac actttttctc aggacatctc tggcaggtgc atttgtgcct gcccagcagt 960 tccagctgtg gcaaaagtct cttccatgga caagtgtttg cacgggggtt cagctgtgcc 1020 cgcccccagg ctgtgcccca ccacagattc tgccaaggat cagaactcat gtgaaacaaa 1080 cagctgacgt cctctctcga tctgcaagcc tttcaccaac caaatagttg cctctctcgt 1140 caccaaactg gaacctcaca ccagccggca aaggaaggaa gaaaggtttt agagctgtgt 1200 gttctttctc tggctttgat tcttctttga gttctcttac ttgccacgta caggaccatt 1260 atttatgagt gaaaagttgt agcacattcc ttttgcaggt ctgagctaag cccttgaaag 1320 cagggtaatg ctcataaaag gactgttccc gcggccccaa ggtgcctgtt gttcacactt 1380 aagggaagtt tataaagcta ctggccccag atgctcaggg taaggagcac caaagctgag 1440 gctggctcag agatctccag agaagctgca gcctgccctg gccctggctc tggccctggc 1500 ccacattgca catggaaacc caaaggcata tatctgcgta tgtgtggtac ttagtcacat 1560 ctttgtcaac aaactgttcg tttttaagtt acaaatttga atttaatgtt gtcatcatcg 1620 tcatgtgttt ccccaaaggg aagccagtca ttgaccattt aaaaagtctc ctgctaagta 1680 tggaaatcag acagtaagag aaagccaaaa agcaatgcag agaaaggtgt ccaagctgtc 1740 ttcagccttc cccagctaaa gagcagagga gggcctgggc tacttgggtt ccccatcggc 1800 ctccagcact gcctccctcc tcccactgcg actctgggat ctccaggtgc tgcccaagga 1860 gttgccttga ttacagagag gggagcctcc aattcggcca acttggagtc ctttctgttt 1920 tgaagcatgg gccagacccg gcactgcgct cggagagccg gtgggcctgg cctccccgtc 1980 gacctcagtg cctttttgtt ttcagagaga aataggagta gggcgagttt gcctgaagct 2040 ctgctgctgg cttctcctgc caggaagtga acaatggcgg cggtgtggga gacaaggcca 2100 ggagagcccg cgttcagtat gggttgaggg tcacagacct ccctcccatc tgggtgcctg 2160 agttttgact ccaatcagtg ataccagacc acattgacag ggaggatcaa attcctgact 2220 tacatttgca ctggcttctt gtttaggctg aatcctaaaa taaattagtc aaaaaattcc 2280 aacaagtagc caggactgca gagacactcc agtgcagagg gagaaggact tgtaattttc 2340 aaagcagggc tggttttcca acccagcctc tgagaaacca tttctttgct atcctctgcc 2400 ttcccaagtc cctcttgggt cggttcaagc ccaagcttgt tcgtgtagct tcagaagttc 2460 cctctccgac ccaggctgag tccatactgc ccctgatccc agaaggaatg ctgacccctc 2520 gtcgtatgaa ctgtgcatag tctccagagc ttcaaaggca acacaagctc gcaactctaa 2580 gattttttta aaccacaaaa accctggtta gccatctcat gctcagcctt atcacttccc 2640 tccctttaga aactctctcc ctgctgtata ttaaagggag caggtggaga gtcattttcc 2700 ttcgtcctgc atgtctctaa cattaataga aggcatggct cctgctgcaa ccgctgtgaa 2760 tgctgctgag aacctccctc tatggggatg gctattttat ttttgagaag gaaaaaaaaa 2820 gtcatgtata tatacacata aaggcatata gctatatata aagagataag ggtgtttatg 2880 aaatgagaaa attattggac aattcagact ttactaaagc acagttagac ccaaggccta 2940 tgctgaggtc taaacctctg aaaaaagtat agtatcgagt acccgttccc tcccagaggt 3000 gggagtaact gctggtagtg ccttctttgg ttgtgttgct cagtgtgtaa gtgtttgttt 3060 ccaggatatt ttctttttaa atgtctttct tatatgggtt ttaaaaaaaa gtaataaaag 3120 cctgttgcaa aaatgaaaaa aaaa 3144 9 2309 DNA Homo sapiens 3054032CB1 9 aaggggccca ggaagatcaa gttgctgagg agaaatgggg aggaagtttt cctgagtgcc 60 tatgatgacc taagtcccct tctgggacct aaacccccaa tctggaaggg ttcagggagt 120 ctggagggag aggcagcagg atgtggaagg caggctctgg gacagggtgg ggaagagcag 180 gcatgctggg aagttgggga ggacaagcag gctgagcctg gaggcaggct agacatcagg 240 gaagaggcag agggaagtcc agagaccaag gtggaggctg gaaaggccag tgaggataga 300 ggggaggctg ggggaagcca agagacaaaa gtcagattga gagaagggag tagggaagag 360 acagaggcca aggaagagaa gtccaaaggt cagaagaagg ctgacagtat ggaggctaaa 420 ggtgtggagg aaccaggagg agatgagtat acagatgaga aggaaaaaga aattgagaga 480 gaagaggatg aacaaagaga ggaagcccag gtagaagctg gaagggacct agagcaaggg 540 gcccaggaag atcaagttgc tgaggagaaa tgggaagttg tacagaaaca agaggctgag 600 ggagtcagag aggatgagga caaaggacag agggagaagg ggtaccatga agcaagaaaa 660 gaccaaggag atggtgaaga cagcagaagc ccagaagcag caactgaagg aggagcaggg 720 gaggtcagca aggaacggga gagtggggat ggagaggctg agggagacca gagggctgga 780 gggtactatt tagaagagga caccctctct gaaggttcag gtgtagcgtc cctggaggtt 840 gactgtgcca aagagggcaa tcctcactct tctgagatgg aagaggtagc cccacagcca 900 cctcagccag aggagatgga gcctgagggg cagcccagtc cagacggctg tctatgcccc 960 tgttctcttg gcctgggtgg cgtgggcatg cgtctagctt ccactctggt tcaggtccaa 1020 caggtccgct ctgtgcctgt ggtgcccccc aagccacagt ttgccaagat gcccagtgca 1080 atgtgtagca agattcatgt ggcacctgca aatccatgcc cgaggcctgg ccggcttgat 1140 gggactcctg gagaaagggc ttgggggtcc cgagcttctc gatcctcttg gaggaatggg 1200 ggtagtcttt cctttgatgc tgctgtggcc ctagcccggg accgccaaag gactgaggct 1260 caaggagttc ggcgaaccca gacctgtact gagggtgggg attactgcct catccccaga 1320 acctcccctt gtagcatgat ctctgcccat tctcctcggc cccttagctg cctggagctc 1380 ccatctgaag gtgcagaagg gtctggatcc cggagtcgtc ttagtctgcc ccccagagaa 1440 ccccaggttc ctgaccccct gttgtcctct cagcgcagat catatgcatt tgaaacacag 1500 gctaaccctg ggaaaggtga aggactgtga ttaggaccac agccctgggc aaaggggacc 1560 agcaagttgt cttgaatctc cagggttcct gactagctgt ctcctctgca gcatgagcag 1620 ctgtagtgcc caactctata ggctttggcc ctccagcttc tctctttgac tgtgggaggc 1680 actgccttgg ttggtttacc tgaacttgtc tccgacacaa agcacttatc tcttaggaga 1740 ttcccaagaa agtcaacaag atcttgttcc cagggagtgg gtcattggcc aaagggaaca 1800 taaggtaggc agaaaactta aaagagtttg ttaaagtgaa gactggagaa attcctccct 1860 tcctctgagc tgtgaatctc tcttcatgaa agccaaaggt agagacaggg aggacagggc 1920 caggttaggg ccttccacac acaaacactt ctagagttgc ccattcctgt tatgttcttg 1980 gaccctaaga tacctcctgt cccttttaaa tccagattaa gagaaacgtc caggaagagc 2040 tctttgaagc cctcaatatt tgttggaggg actggactcc tctccagctc cccaccctct 2100 gcctccagtc accatgtgca agagaggtcc tgtacagatc tctctgggct ctcctttctc 2160 ctttggaata acttgttcct atttcaggaa agggaaatgg tgtcactcag gccctgggac 2220 tgcttctcca gccaggctgg ggccacaggt cccactctag tgaaggtcaa tgtctcagaa 2280 taaaagctgt atttttacaa aaaaaaaaa 2309 10 1666 DNA Homo sapiens 3055142CB1 10 ctgctcgaga actgaatggc cctgtgcaga gccatagtcc cactgtgggt cctgcaatga 60 gcaggggctg ggagtagagg gtttctgggg cctcagggtt ctgggaaagc aacagctatc 120 agagagagaa gggccagacc ccatagcctc ttagattcct ggcagtagaa ggagaaggat 180 gggtaaattg acctctgaag tccctgacca ttagcatggt ctaggatcct ttctagaagg 240 aagatctgag gctctggtgc tcagggggat ggcttgggcc ttttctctca accttggctg 300 agcctacccc ttactttgcc aaagacttga ggaccctgta tgtctggagt tcagtcccct 360 cctctgtggg gctcaggtga ttgaaatgtg gatgaaacat ttctctactt caagaccacc 420 tctccctgca aacaccacac acacatggca tgcatgtacg cacatgcgca cacacacacg 480 cacacacctc aataatttct ctcaagtttc ctgagtctcc agaaaaacag cactaacgct 540 ggacctgtct actctcagaa cccggcacag attctctctt gatctccttt tggaatctga 600 gattcttaga agacaggata gggttaaatt tagtagcagc tcagttctag ctaaatcact 660 agaggaagtt aattaacttt aagccttcat ttctccagca ctaaaatgga gtggagagtt 720 ggggtggaaa taagacatcc ttaaaaggtt aaattgtctg caaagcacct agcccagtgc 780 cgagctccca gtaggtgttc agtaaagctt agtgcctgac tttctgaaca ctgattcctc 840 ctgtttggag tcactgggat actctcattg ccgttgggat gttcctcact ccttcccagt 900 tcgtggctga ggcagaaccc agactgaaga gggaagagac attccagagg aggattgcct 960 tcgtcagggt aaggggtggg ctgctcaggg gccctaccct tcaccccctt ctgtatcaga 1020 ttggccctcc cactcccatc tcactctgcg tgtacaatct tccatatccg caagttcact 1080 ggcactcttc tggcacctgg gcaagatccc agaacagagg atggagtgac tggcctcaca 1140 gagcttagtg cccgactcag gggaaatggg actggtgcat gggaaatggt cagcctagga 1200 taggacacga gagtctgaaa ttcaaagcaa ccagcttgaa gtggtttgag aagctggaag 1260 caaacatggg ctagagagat agggcagaag tcaagacgag gatctggact gatgtggaga 1320 aagtagccac ggaagcatga actgtatcct gcacaaagtc cctcttcccc gcctcctaat 1380 tcattatgcc caaaaggcct tacgtgaaat tccagcccag agtactcatg acttgagaga 1440 cgtggacaga gccagcttct accttgcctg gccgtctctc ccctgtctta atgtctgctc 1500 ttgctctaag ctccagaaga gtggcgggcc atgtatcttc aatatgtttt tgctgtatgg 1560 gcaggttgtc ttattatgtg atcaacagat gtccaggaac taatgagtgg aatttaatat 1620 tattgtcaaa taaaacttga tttgtcctat aaaaaaaaaa aaaaaa 1666 11 1751 DNA Homo sapiens 3507924CB1 11 tttagaggtt cctgtttgca tctctgcaac cacttcagaa ggcacgtgtt tggtttgctc 60 tgagcctaac ctagagtgct cgcagcagtc tttcagttga gcttggggac tgcagctgtg 120 gggagatttc agtgcattgc ctcccctggg tgctcttcat cttggatttg aaagttgaga 180 gcagcatgtt ttgcccactg aaactcatcc tgctgccagt gttactggat tattccttgg 240 gcctgaatga cttgaatgtt tccccgcctg agctaacagt ccatgtgggt gattcagctc 300 tgatgggatg tgttttccag agcacagaag acaaatgtat attcaagata gactggactc 360 tgtcaccagg agagcacgcc aaggacgaat atgtgctata ctattactcc aatctcagtg 420 tgcctattgg gcgcttccag aaccgcgtac acttgatggg ggacatctta tgcaatgatg 480 gctctctcct gctccaagat gtgcaagagg ctgaccaggg aacctatatc tgtgaaatcc 540 gcctcaaagg ggagagccag gtgttcaaga aggcggtggt actgcatgtg cttccagagg 600 agcccaaaga gctcatggtc catgtgggtg gattgattca gatgggatgt gttttccaga 660 gcacagaagt gaaacacgtg accaaggtag aatggatatt ttcaggacgg cgcgcaaagg 720 aggagattgt atttcgttac taccacaaac tcaggatgtc tgtggagtac tcccagagct 780 ggggccactt ccagaatcgt gtgaacctgg tgggggacat tttccgcaat gacggttcca 840 tcatgcttca aggagtgagg gagtcagatg gaggaaacta cacctgcagt atccacctag 900 ggaacctggt gttcaagaaa accattgtgc tgcatgtcag cccggaagag cctcgaacac 960 tggtgacccc ggcagccctg aggcctctgg tcttgggtgg taatcagttg gtgatcattg 1020 tgggaattgt ctgtgccaca atcctgctgc tccctgttct gatattgatc gtgaagaaga 1080 cctgtggaaa taagagttca gtgaattcta cagtcttggt gaagaacacg aagaagacta 1140 atccagagat aaaagaaaaa ccctgccatt ttgaaagatg tgaaggggag aaacacattt 1200 actccccaat aattgtacgg gaggtgatcg aggaagaaga accaagtgaa aaatcagagg 1260 ccacctacat gaccatgcac ccagtttggc cttctctgag gtcagatcgg aacaactcac 1320 ttgaaaaaaa gtcaggtggg ggaatgccaa aaacacagca agccttttga gaagaatgga 1380 gagtcccttc atctcagcag cggtggagac tctctcctgt gtgtgtcctg ggccactcta 1440 ccagtgattt cagactcccg ctctcccagc tgtcctcctg tctcattgtt tggtcaatac 1500 actgaagatg gagaatttgg agcctggcag agagactgga cagctctgga ggaacaggcc 1560 tgctgagggg aggggagcat ggacttggcc tctggagtgg gacactggcc ctgggaacca 1620 ggctgagctg agtggcctca aaccccccgt tggatcagac cctcctgtgg gcagggttct 1680 tagtggatga gttactggga agaatcagag ataaaaacca acccaaatca ttcctctggc 1740 aaaaaaaaaa a 1751 12 127 PRT Homo sapiens 402234CD1 12 Met Phe Gln Ala Ala Gly Ala Ala Gln Ala Thr Pro Ser His Asp 1 5 10 15 Ala Lys Gly Gly Gly Ser Ser Thr Val Gln Arg Ser Lys Ser Phe 20 25 30 Ser Leu Arg Ala Gln Val Lys Glu Thr Cys Ala Ala Cys Gln Lys 35 40 45 Thr Val Tyr Pro Met Glu Arg Leu Val Ala Asp Lys Leu Ile Phe 50 55 60 His Asn Ser Cys Phe Cys Cys Lys His Cys His Thr Lys Leu Ser 65 70 75 Leu Gly Ser Tyr Ala Ala Leu His Gly Glu Phe Tyr Cys Lys Pro 80 85 90 His Phe Gln Gln Leu Phe Lys Ser Lys Gly Asn Tyr Asp Glu Gly 95 100 105 Phe Gly Arg Lys Gln His Lys Glu Leu Trp Ala His Lys Glu Val 110 115 120 Asp Pro Gly Thr Lys Thr Ala 125 13 93 PRT Homo sapiens 569989CD1 13 Met Ile His Leu Gly His Ile Leu Phe Leu Leu Leu Leu Pro Val 1 5 10 15 Ala Ala Ala Gln Thr Thr Pro Gly Glu Arg Ser Ser Leu Pro Ala 20 25 30 Phe Tyr Pro Gly Thr Ser Gly Ser Cys Ser Gly Cys Gly Ser Leu 35 40 45 Ser Leu Pro Leu Leu Ala Gly Leu Val Ala Ala Asp Ala Val Ala 50 55 60 Ser Leu Leu Ile Val Gly Ala Val Phe Leu Cys Ala Arg Pro Arg 65 70 75 Arg Ser Pro Ala Gln Glu Asp Gly Lys Val Tyr Ile Asn Met Pro 80 85 90 Gly Arg Gly 14 225 PRT Homo sapiens 2349263CD1 14 Met Thr Ser Gln Pro Val Pro Asn Glu Thr Ile Ile Val Leu Pro 1 5 10 15 Ser Asn Val Ile Asn Phe Ser Gln Ala Glu Lys Pro Glu Pro Thr 20 25 30 Asn Gln Gly Gln Asp Ser Leu Lys Lys His Leu His Ala Glu Ile 35 40 45 Lys Val Ile Gly Thr Ile Gln Ile Leu Cys Gly Met Met Val Leu 50 55 60 Ser Leu Gly Ile Ile Leu Ala Ser Ala Ser Phe Ser Pro Asn Phe 65 70 75 Thr Gln Val Thr Ser Thr Leu Leu Asn Ser Ala Tyr Pro Phe Ile 80 85 90 Gly Pro Phe Phe Phe Ile Ile Ser Gly Ser Leu Ser Ile Ala Thr 95 100 105 Glu Lys Arg Leu Thr Lys Leu Leu Val His Ser Ser Leu Val Gly 110 115 120 Ser Ile Leu Ser Ala Leu Ser Ala Leu Val Gly Phe Ile Ile Leu 125 130 135 Ser Val Lys Gln Ala Thr Leu Asn Pro Ala Ser Leu Gln Cys Glu 140 145 150 Leu Asp Lys Asn Asn Ile Pro Thr Arg Ser Tyr Val Ser Tyr Phe 155 160 165 Tyr His Asp Ser Leu Tyr Thr Thr Asp Cys Tyr Thr Ala Lys Ala 170 175 180 Ser Leu Ala Gly Thr Leu Ser Leu Met Leu Ile Cys Thr Leu Leu 185 190 195 Glu Phe Cys Leu Ala Val Leu Thr Ala Val Leu Arg Trp Lys Gln 200 205 210 Ala Tyr Ser Asp Phe Pro Gly Val Ser Val Leu Ala Gly Phe Thr 215 220 225 15 547 PRT Homo sapiens 2471716CD1 15 Met Ser Glu Pro Pro Val Tyr Cys Asn Leu Val Asp Leu Arg Arg 1 5 10 15 Cys Pro Arg Ser Pro Pro Pro Gly Pro Ala Cys Pro Leu Leu Gln 20 25 30 Arg Leu Asp Ala Trp Glu Gln His Leu Asp Pro Asn Ser Gly Arg 35 40 45 Cys Phe Tyr Ile Asn Ser Leu Thr Gly Cys Lys Ser Trp Lys Pro 50 55 60 Pro Arg Arg Ser Arg Ser Glu Thr Asn Pro Gly Ser Met Glu Gly 65 70 75 Thr Gln Thr Leu Lys Arg Asn Asn Asp Val Leu Gln Pro Gln Ala 80 85 90 Lys Gly Phe Arg Ser Asp Thr Gly Thr Pro Glu Pro Leu Asp Pro 95 100 105 Gln Gly Ser Leu Ser Leu Ser Gln Arg Thr Ser Gln Leu Asp Pro 110 115 120 Pro Ala Leu Gln Ala Pro Arg Pro Leu Pro Gln Leu Leu Asp Asp 125 130 135 Pro His Glu Val Glu Lys Ser Gly Leu Leu Asn Met Thr Lys Ile 140 145 150 Ala Gln Gly Gly Arg Lys Leu Arg Lys Asn Trp Gly Pro Ser Trp 155 160 165 Val Val Leu Thr Gly Asn Ser Leu Val Phe Tyr Arg Glu Pro Pro 170 175 180 Pro Thr Ala Pro Ser Ser Gly Trp Gly Pro Ala Gly Ser Arg Pro 185 190 195 Glu Ser Ser Val Asp Leu Arg Gly Ala Ala Leu Ala His Gly Arg 200 205 210 His Leu Ser Ser Arg Arg Asn Val Leu His Ile Arg Thr Ile Pro 215 220 225 Gly His Glu Phe Leu Leu Gln Ser Asp His Glu Thr Glu Leu Arg 230 235 240 Ala Trp His Arg Ala Leu Arg Thr Val Ile Glu Arg Leu Asp Arg 245 250 255 Glu Asn Pro Leu Glu Leu Arg Leu Ser Gly Ser Gly Pro Ala Glu 260 265 270 Leu Ser Ala Gly Glu Asp Glu Glu Glu Glu Ser Glu Leu Val Ser 275 280 285 Lys Pro Leu Leu Arg Leu Ser Ser Arg Arg Ser Ser Ile Arg Gly 290 295 300 Pro Glu Gly Thr Glu Gln Asn Arg Val Arg Asn Lys Leu Lys Arg 305 310 315 Leu Ile Ala Lys Arg Pro Pro Leu Gln Ser Leu Gln Glu Arg Gly 320 325 330 Leu Leu Arg Asp Gln Val Phe Gly Cys Gln Leu Glu Ser Leu Cys 335 340 345 Gln Arg Glu Gly Asp Thr Val Pro Ser Phe Leu Arg Leu Cys Ile 350 355 360 Ala Ala Val Asp Lys Arg Gly Leu Asp Val Asp Gly Ile Tyr Arg 365 370 375 Val Ser Gly Asn Leu Ala Val Val Gln Lys Leu Arg Phe Leu Val 380 385 390 Asp Arg Glu Arg Ala Val Thr Ser Asp Gly Arg Tyr Val Phe Pro 395 400 405 Glu Gln Pro Gly Gln Glu Gly Arg Leu Asp Leu Asp Ser Thr Glu 410 415 420 Trp Asp Asp Ile His Val Val Thr Gly Ala Leu Lys Leu Phe Leu 425 430 435 Arg Glu Leu Pro Gln Pro Leu Val Pro Pro Leu Leu Leu Pro His 440 445 450 Phe Arg Ala Ala Leu Ala Leu Ser Glu Ser Glu Gln Cys Leu Ser 455 460 465 Gln Ile Gln Glu Leu Ile Gly Ser Met Pro Lys Pro Asn His Asp 470 475 480 Thr Leu Arg Tyr Leu Leu Glu His Leu Cys Arg Val Ile Ala His 485 490 495 Ser Asp Lys Asn Arg Met Thr Pro His Asn Leu Gly Ile Val Phe 500 505 510 Gly Pro Thr Leu Phe Arg Pro Glu Gln Glu Thr Ser Asp Pro Ala 515 520 525 Ala His Ala Leu Tyr Pro Gly Gln Leu Val Gln Leu Met Leu Thr 530 535 540 Asn Phe Thr Ser Leu Phe Pro 545 16 265 PRT Homo sapiens 2726173CD1 16 Met Thr Val Asp Ser Ser Met Ser Ser Gly Tyr Cys Ser Leu Asp 1 5 10 15 Glu Glu Leu Glu Asp Cys Phe Phe Thr Ala Lys Thr Thr Phe Phe 20 25 30 Arg Asn Ala Gln Ser Lys His Leu Ser Lys Asn Val Cys Lys Pro 35 40 45 Val Glu Glu Thr Gln Arg Pro Pro Thr Leu Gln Glu Ile Lys Gln 50 55 60 Lys Ile Asp Ser Tyr Asn Thr Arg Glu Lys Asn Cys Leu Gly Met 65 70 75 Lys Leu Ser Glu Asp Gly Thr Tyr Thr Gly Phe Ile Lys Val His 80 85 90 Leu Lys Leu Arg Arg Pro Val Thr Val Pro Ala Gly Ile Arg Pro 95 100 105 Gln Ser Ile Tyr Asp Ala Ile Lys Glu Val Asn Leu Ala Ala Thr 110 115 120 Thr Asp Lys Arg Thr Ser Phe Tyr Leu Pro Leu Asp Ala Ile Lys 125 130 135 Gln Leu His Ile Ser Ser Thr Thr Thr Val Ser Glu Val Ile Gln 140 145 150 Gly Leu Leu Lys Lys Phe Met Val Val Asp Asn Pro Gln Lys Phe 155 160 165 Ala Leu Phe Lys Arg Ile His Lys Asp Gly Gln Val Leu Phe Gln 170 175 180 Lys Leu Ser Ile Ala Asp Arg Pro Leu Tyr Leu Arg Leu Leu Ala 185 190 195 Gly Pro Asp Thr Glu Val Leu Asn Phe Val Leu Lys Glu Asn Glu 200 205 210 Thr Gly Glu Val Glu Trp Asp Ala Phe Ser Ile Pro Glu Leu Gln 215 220 225 Asn Phe Leu Thr Ile Leu Glu Lys Glu Glu Gln Asp Lys Ile Gln 230 235 240 Gln Val Gln Lys Lys Tyr Asp Lys Phe Arg Gln Lys Leu Glu Glu 245 250 255 Ala Leu Arg Glu Ser Gln Gly Lys Pro Gly 260 265 17 394 PRT Homo sapiens 3507924CD1 17 Met Phe Cys Pro Leu Lys Leu Ile Leu Leu Pro Val Leu Leu Asp 1 5 10 15 Tyr Ser Leu Gly Leu Asn Asp Leu Asn Val Ser Pro Pro Glu Leu 20 25 30 Thr Val His Val Gly Asp Ser Ala Leu Met Gly Cys Val Phe Gln 35 40 45 Ser Thr Glu Asp Lys Cys Ile Phe Lys Ile Asp Trp Thr Leu Ser 50 55 60 Pro Gly Glu His Ala Lys Asp Glu Tyr Val Leu Tyr Tyr Tyr Ser 65 70 75 Asn Leu Ser Val Pro Ile Gly Arg Phe Gln Asn Arg Val His Leu 80 85 90 Met Gly Asp Ile Leu Cys Asn Asp Gly Ser Leu Leu Leu Gln Asp 95 100 105 Val Gln Glu Ala Asp Gln Gly Thr Tyr Ile Cys Glu Ile Arg Leu 110 115 120 Lys Gly Glu Ser Gln Val Phe Lys Lys Ala Val Val Leu His Val 125 130 135 Leu Pro Glu Glu Pro Lys Glu Leu Met Val His Val Gly Gly Leu 140 145 150 Ile Gln Met Gly Cys Val Phe Gln Ser Thr Glu Val Lys His Val 155 160 165 Thr Lys Val Glu Trp Ile Phe Ser Gly Arg Arg Ala Lys Glu Glu 170 175 180 Ile Val Phe Arg Tyr Tyr His Lys Leu Arg Met Ser Val Glu Tyr 185 190 195 Ser Gln Ser Trp Gly His Phe Gln Asn Arg Val Asn Leu Val Gly 200 205 210 Asp Ile Phe Arg Asn Asp Gly Ser Ile Met Leu Gln Gly Val Arg 215 220 225 Glu Ser Asp Gly Gly Asn Tyr Thr Cys Ser Ile His Leu Gly Asn 230 235 240 Leu Val Phe Lys Lys Thr Ile Val Leu His Val Ser Pro Glu Glu 245 250 255 Pro Arg Thr Leu Val Thr Pro Ala Ala Leu Arg Pro Leu Val Leu 260 265 270 Gly Gly Asn Gln Leu Val Ile Ile Val Gly Ile Val Cys Ala Thr 275 280 285 Ile Leu Leu Leu Pro Val Leu Ile Leu Ile Val Lys Lys Thr Cys 290 295 300 Gly Asn Lys Ser Ser Val Asn Ser Thr Val Leu Val Lys Asn Thr 305 310 315 Lys Lys Thr Asn Pro Glu Ile Lys Glu Lys Pro Cys His Phe Glu 320 325 330 Arg Cys Glu Gly Glu Lys His Ile Tyr Ser Pro Ile Ile Val Arg 335 340 345 Glu Val Ile Glu Glu Glu Glu Pro Ser Glu Lys Ser Glu Ala Thr 350 355 360 Tyr Met Thr Met His Pro Val Trp Pro Ser Leu Arg Ser Asp Arg 365 370 375 Asn Asn Ser Leu Glu Lys Lys Ser Gly Gly Gly Met Pro Lys Thr 380 385 390 Gln Gln Ala Phe

Claims (55)

What is claimed is:
1. An isolated polypeptide selected from the group consisting of:
a) a polypeptide comprising an amino acid sequence of SEQ ID NO: 12-17,
b) a polypeptide comprising a naturally occurring an amino acid sequence at least 90% identical to an amino acid sequence of SEQ ID NO: 12-17.
c) a biologically active fragment of a polypeptide having an amino acid sequence of SEQ ID NO: 12-17, and
d) an antigenic epitope of a polypeptide having an amino acid sequence of SEQ ID NO: 12-17.
2. An isolated polypeptide of claim 1, comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17.
3. An isolated polynucleotide encoding a polypeptide of claim 1.
4. An isolated polynucleotide encoding a polypeptide of claim 2.
5. An isolated polynucleotide of claim 4, selected from the group consisting of SEQ ID NO: 1, 2, 6, 7, 8, and 11.
6. A recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide of claim 3.
7. A cell transformed with a recombinant polynucleotide of claim 6.
8. A transgenic organism comprising a recombinant polynucleotide of claim 6.
9. A method of producing a polypeptide of claim 1, the method comprising:
a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide comprises a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 1, and
b) recovering the polypeptide so expressed.
10. A method of claim 9, wherein the polypeptide is selected from the group consisting of SEQ ID NO:12-17.
11. An isolated antibody which specifically binds to a polypeptide of claim 1.
12. An isolated polynucleotide selected from the group consisting of:
a) a polynucleotide comprising a polynucleotide sequence of SEQ ID NO: 1-11,
b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence of SEQ ID NO: 1-11,
c) a polynucleotide complementary to a polynucleotide of a),
d) a polynucleotide complementary to a polynucleotide of b) and
e) an RNA equivalent of a)-d).
13. An isolated polynucleotide comprising at least 60 contiguous nucleotides of a polynucleotide of claim 12.
14. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising:
a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and
b) detecting the presence or absence of said hybridization complex, and, optionally, if present, the amount thereof.
15. A method of claim 14, wherein the probe comprises at least 60 contiguous nucleotides.
16. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising:
a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and
b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
17. A composition comprising a polypeptide of claim 1 and a pharmaceutically acceptable excipient.
18. A composition of claim 17, wherein the polypeptide comprises an amino acid selected from the group consisting of SEQ ID NO: 12-17.
19. A method for treating a disease or condition associated with decreased expression of functional HLHLP, comprising administering to a patient in need of such treatment the composition of claim 17.
20. A method of screening a compound for effectiveness as an agonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and
b) detecting agonist activity in the sample.
21. A composition comprising an agonist compound identified by a method of claim 20 and a pharmaceutically acceptable excipient.
22. A method for treating a disease or condition associated with decreased expression of functional HLHLP, comprising administering to a patient in need of such treatment a composition of claim 21.
23. A method of screening a compound for effectiveness as an antagonist of a polypeptide of claim 1, the method comprising:
a) exposing a sample comprising a polypeptide of claim 1 to a compound, and
b) detecting antagonist activity in the sample.
24. A composition comprising an antagonist compound identified by a method of claim 23 and a pharmaceutically acceptable excipient.
25. A method for treating a disease or condition associated with overexpression of functional HLHLP, comprising administering to a patient in need of such treatment a composition of claim 24.
26. A method of screening for a compound that specifically binds to the polypeptide of claim 1, the method comprising:
a) combining the polypeptide of claim 1 with at least one test compound under suitable conditions, and
b) detecting binding of the polypeptide of claim 1 to the test compound, thereby identifying a compound that specifically binds to the polypeptide of claim 1.
27. A method of screening for a compound that modulates the activity of the polypeptide of claim 1, said method comprising:
a) combining the polypeptide of claim 1 with at least one test compound under conditions permissive for the activity of the polypeptide of claim 1,
b) assessing the activity of the polypeptide of claim 1 in the presence of the test compound, and
c) comparing the activity of the polypeptide of claim 1 in the presence of the test compound with the activity of the polypeptide of claim 1 in the absence of the test compound, wherein a change in the activity of the polypeptide of claim 1 in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide of claim 1.
28. A method of screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence of claim 5, the method comprising:
a) exposing a sample comprising the target polynucleotide to a compound, under conditions suitable for the expression of the target polynucleotide,
b) detecting altered expression of the target polynucleotide, and
c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.
29. A method of assessing toxicity of a test compound, the method comprising:
a) treating a biological sample containing nucleic acids with the test compound,
b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 12 under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide comprising a polynucleotide sequence of a polynucleotide of claim 12 or fragment thereof,
c) quantifying the amount of hybridization complex, and
d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.
30. A diagnostic test for a condition or disease associated with the expression of SEQ ID NO: 12-17 in a biological sample, the method comprising:
a) combining the biological sample with an antibody of claim 11, under conditions suitable for the antibody to bind the polypeptide and form an antibody:polypeptide complex, and
b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
31. The antibody of claim 11, wherein the antibody is:
a) a chimeric antibody,
b) a single chain antibody,
c) a Fab fragment,
d) a F(ab′)2 fragment, or
e) a humanized antibody.
32. A composition comprising an antibody of claim 11 and an acceptable excipient.
33. A method of diagnosing a condition or disease associated with the expression of HLHLP in a subject, comprising administering to said subject an effective amount of the composition of claim 32.
34. A composition of claim 32, wherein the antibody is labeled.
35. A diagnostic test for a condition or disease associated with the expression of SEQ ID NO: 12-17 in a biological sample, the method comprising:
a) combining the biological sample with the composition of claim 34, under conditions suitable for the antibody to bind the polypeptide and form an antibody:polypeptide complex, and
b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
36. A method of preparing a polyclonal antibody with the specificity of the antibody of claim 11, the method comprising:
a) immunizing an animal with a polypeptide selected from the group consisting of SEQ ID NO: 12-17, or an immunogenic fragment thereof, under conditions to elicit an antibody response,
b) isolating antibodies from said animal, and
c) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17.
37. A polyclonal antibody produced by a method of claim 36.
38. A composition comprising the polyclonal antibody of claim 37 and a suitable carrier.
39. A method of making a monoclonal antibody with the specificity of the antibody of claim 11, the method comprising:
a) immunizing an animal with a polypeptide selected from the group consisting of SEQ ID NO: 12-17, or an immunogenic fragment thereof, under conditions to elicit an antibody response,
b) isolating antibody producing cells from the animal,
c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells,
d) culturing the hybridoma cells, and
e) isolating from the culture monoclonal antibody which binds specifically to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOS: 12-17.
40. A monoclonal antibody produced by a method of claim 39.
41. A composition comprising the monoclonal antibody of claim 40 and a suitable carrier.
42. The antibody of claim 11, wherein the monoclonal antibody is produced by screening a Fab expression library.
43. The antibody of claim 11, wherein the antibody is produced by screening a recombinant immunoglobulin library.
44. A method of detecting a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17 in a sample, the method comprising:
a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and
b) detecting specific binding, wherein specific binding indicates the presence of a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17 in the sample.
45. A method of purifying a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17 from a sample, the method comprising:
a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and
b) separating the antibody from the sample and obtaining the purified polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 12-17.
46. A microarray wherein at least one element of the microarray is a polynucleotide of claim 12.
47. A method of generating an expression profile of a sample which contains polynucleotides, the method comprising:
a) labeling the polynucleotides of the sample,
b) contacting the elements of the microarray of claim 46 with the labeled polynucleotides of the sample under conditions suitable for the formation of a hybridization complex, and
c) quantifying the expression of the polynucleotides in the sample.
48. An array comprising different nucleotide molecules affixed in distinct physical locations on a solid substrate, wherein at least one of said nucleotide molecules comprises a first oligonucleotide or polynucleotide sequence specifically hybridizable with at least 30 contiguous nucleotides of a target polynucleotide, and wherein said target polynucleotide is a polynucleotide of claim 12.
49. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 30 contiguous nucleotides of said target polynucleotide.
50. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 60 contiguous nucleotides of said target polynucleotide.
51. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to said target polynucleotide.
52. An array of claim 48, which is a microarray.
53. An array of claim 48, further comprising said target polynucleotide hybridized to a nucleotide molecule comprising said first oligonucleotide or polynucleotide sequence.
54. An array of claim 48, wherein a linker joins at least one of said nucleotide molecules to said solid substrate.
55. An array of claim 48, wherein each distinct physical location on the substrate contains multiple nucleotide molecules, and the multiple nucleotide molecules at any single distinct physical location have the same sequence, and each distinct physical location on the substrate contains nucleotide molecules having a sequence which differs from the sequence of nucleotide molecules at another distinct physical location on the substrate.
US10/440,366 1998-11-18 2003-05-16 Inflammation-associated polynucleotides Abandoned US20030181706A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/440,366 US20030181706A1 (en) 1998-11-18 2003-05-16 Inflammation-associated polynucleotides

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US19529298A 1998-11-18 1998-11-18
US09/855,323 US6602667B1 (en) 1998-11-18 2001-05-14 Inflammation-associated polynucleotides
US10/440,366 US20030181706A1 (en) 1998-11-18 2003-05-16 Inflammation-associated polynucleotides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/855,323 Division US6602667B1 (en) 1998-11-18 2001-05-14 Inflammation-associated polynucleotides

Publications (1)

Publication Number Publication Date
US20030181706A1 true US20030181706A1 (en) 2003-09-25

Family

ID=22720829

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/855,323 Expired - Fee Related US6602667B1 (en) 1998-11-18 2001-05-14 Inflammation-associated polynucleotides
US10/440,366 Abandoned US20030181706A1 (en) 1998-11-18 2003-05-16 Inflammation-associated polynucleotides

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/855,323 Expired - Fee Related US6602667B1 (en) 1998-11-18 2001-05-14 Inflammation-associated polynucleotides

Country Status (6)

Country Link
US (2) US6602667B1 (en)
EP (1) EP1131425A2 (en)
JP (1) JP2002530077A (en)
AU (1) AU1344400A (en)
CA (1) CA2350415A1 (en)
WO (1) WO2000029574A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022178367A3 (en) * 2021-02-19 2022-09-29 University Of Southern California Single-chain and multi-chain synthetic antigen receptors for diverse immune cells

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030069403A1 (en) * 1997-06-16 2003-04-10 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
IL178921A0 (en) * 1998-12-24 2007-03-08 Yeda Res & Dev Caspase-8 interacting proteins
JP4838961B2 (en) 1999-09-21 2011-12-14 中外製薬株式会社 Transporter genes OATP-B, C, D, and E
AU2001249698A1 (en) * 2000-03-31 2001-10-15 Millennium Pharmaceuticals, Inc. 32591, a novel human gtpase activating molecule and uses therefor
US6444443B1 (en) 2000-05-19 2002-09-03 Novartis Ag Gene
US6905827B2 (en) 2001-06-08 2005-06-14 Expression Diagnostics, Inc. Methods and compositions for diagnosing or monitoring auto immune and chronic inflammatory diseases
US8241622B2 (en) * 2001-07-13 2012-08-14 University Of Iowa Research Foundation Adeno-associated virus vectors with intravector heterologous terminal palindromic sequences
US20040009503A1 (en) * 2002-07-03 2004-01-15 Molecular Staging, Inc. Immune modulatory activity of human ribonucleases
DE10238922A1 (en) * 2002-08-22 2004-03-04 Universitätsklinikum Charité, Medizinische Fakultät der Humboldt-Universität zu Berlin Immune markers for diagnosis and therapy in connection with graft reactions
AU2003277537A1 (en) * 2002-10-30 2004-05-25 Ishihara Sangyo Kaisha, Ltd. REGULATION OF INTERACTION BETWEEN RAPL AND Rap1
EP2062981B1 (en) * 2005-06-16 2013-09-25 Life Technologies Corporation Gene expression profiling for identification and monitoring of multiple sclerosis
US8399666B2 (en) 2005-11-04 2013-03-19 Panmira Pharmaceuticals, Llc 5-lipoxygenase-activating protein (FLAP) inhibitors
GB2431927B (en) 2005-11-04 2010-03-17 Amira Pharmaceuticals Inc 5-Lipoxygenase-activating protein (FLAP) inhibitors
US7977359B2 (en) 2005-11-04 2011-07-12 Amira Pharmaceuticals, Inc. 5-lipdxygenase-activating protein (FLAP) inhibitors
JP2009521928A (en) * 2005-12-30 2009-06-11 ユニバーシテイ・オブ・アイオワ・リサーチ・フアウンデーシヨン Methods for identifying compounds useful in the treatment of neurodegenerative diseases
MX2010012814A (en) 2008-05-23 2010-12-20 Amira Pharmaceuticals Inc 5-lipoxygenase-activating protein inhibitor.
EP2646470B1 (en) 2010-11-30 2017-03-01 F. Hoffmann-La Roche AG Low affinity anti-transferrin receptor antibodies and their use to transfer therapeutic scfv across the blood brain barrier
JO3300B1 (en) 2012-06-06 2018-09-16 Novartis Ag Compounds and compositions for modulating egfr activity
EP4187486A1 (en) * 2014-12-30 2023-05-31 Ventana Medical Systems, Inc. Method for co-expression analysis
US11702672B2 (en) 2016-02-08 2023-07-18 University Of Iowa Research Foundation Methods to produce chimeric adeno-associated virus/bocavirus parvovirus
MX2018010842A (en) 2016-03-07 2019-07-04 Univ Iowa Res Found Aav-mediated expression using a synthetic promoter and enhancer.
US11142775B2 (en) 2017-01-13 2021-10-12 University Of Iowa Research Foundation Bocaparvovirus small noncoding RNA and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5876946A (en) * 1997-06-03 1999-03-02 Pharmacopeia, Inc. High-throughput assay
US6140488A (en) * 1997-10-01 2000-10-31 Zhang; Xian-Feng Ras-binding protein (PRE1)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5696076A (en) * 1994-06-22 1997-12-09 Human Genome Sciences, Inc. 5-lipoxygenase-activating protein II
DE69734890T2 (en) * 1996-10-11 2006-08-24 New York University RdgB PROTEINS
DE19813835A1 (en) * 1998-03-20 1999-09-23 Metagen Gesellschaft Fuer Genomforschung Mbh Human nucleic acid sequences and protein products from normal breast tissue, useful for breast cancer therapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5876946A (en) * 1997-06-03 1999-03-02 Pharmacopeia, Inc. High-throughput assay
US6140488A (en) * 1997-10-01 2000-10-31 Zhang; Xian-Feng Ras-binding protein (PRE1)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022178367A3 (en) * 2021-02-19 2022-09-29 University Of Southern California Single-chain and multi-chain synthetic antigen receptors for diverse immune cells

Also Published As

Publication number Publication date
EP1131425A2 (en) 2001-09-12
US6602667B1 (en) 2003-08-05
WO2000029574A3 (en) 2000-10-05
JP2002530077A (en) 2002-09-17
CA2350415A1 (en) 2000-05-25
WO2000029574A2 (en) 2000-05-25
AU1344400A (en) 2000-06-05

Similar Documents

Publication Publication Date Title
US6602667B1 (en) Inflammation-associated polynucleotides
US6524799B1 (en) DNA encoding sparc-related proteins
US20020102569A1 (en) Diagnostic marker for cancers
WO2002072596A1 (en) Steap-related protein
JP2002507124A (en) Human extracellular matrix protein
US20030082533A1 (en) Intelectin
US6262247B1 (en) Polycyclic aromatic hydrocarbon induced molecules
CA2415127A1 (en) Down syndrome critical region 1-like 1 proteins
US6566066B1 (en) Aquaporin-8 variant
US6500642B1 (en) Molecule associated with apoptosis
WO2002059311A2 (en) Colon cancer marker
US6444430B1 (en) Ndr2-related proteins
JP2004516813A (en) Related tumor markers
JP2004521611A (en) Genes expressed in the cell cycle
US6692923B2 (en) Tapasin-like protein
US20030104418A1 (en) Diagnostic markers for breast cancer
US20030044812A1 (en) Cell differentiation cDNAs induced by retinoic acid
US20030175787A1 (en) Vesicle membrane proteins
US20030170627A1 (en) cDNAs co-expressed with placental steroid synthesis genes
US20030087253A1 (en) Polynucleotide markers for ovarian cancer
US20020137166A1 (en) ASIP-related proteins
US20020137038A1 (en) Intestinal proteins
US20030124609A1 (en) Ankyrin repeat domain 2 protein variant
US20030099995A1 (en) Ras association domain containing protein
US20020055108A1 (en) Human Sec6 vesicle transport protein

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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