WO2002031152A2 - Intracellular signaling molecules - Google Patents

Intracellular signaling molecules Download PDF

Info

Publication number
WO2002031152A2
WO2002031152A2 PCT/US2001/032090 US0132090W WO0231152A2 WO 2002031152 A2 WO2002031152 A2 WO 2002031152A2 US 0132090 W US0132090 W US 0132090W WO 0231152 A2 WO0231152 A2 WO 0231152A2
Authority
WO
WIPO (PCT)
Prior art keywords
leu
ser
glu
ala
pro
Prior art date
Application number
PCT/US2001/032090
Other languages
French (fr)
Other versions
WO2002031152A3 (en
Inventor
Mariah R. Baughn
Li Ding
Vicki S. Elliott
Ameena R. Gandhi
Kimberly J. Gietzen
Jennifer A. Griffin
Rajagopal Gururajan
April J. A. Hafalia
Liam Kearney
Farrah A. Khan
Preeti Lal
Ernestine A. Lee
Dyung Aina M. Lu
Yan Lu
Danniel B. Nguyen
Chandra Arvizu
Jaya Ramkumar
Y. Tom Tang
Kavitha Thangavelu
Michael Thornton
Narinder K. Chawla
Bridget A. Warren
Yuming Xu
Monique G. Yao
Henry Yue
Original Assignee
Incyte Genomics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Genomics, Inc. filed Critical Incyte Genomics, Inc.
Priority to AU2002216625A priority Critical patent/AU2002216625A1/en
Priority to US10/399,456 priority patent/US20040043395A1/en
Publication of WO2002031152A2 publication Critical patent/WO2002031152A2/en
Publication of WO2002031152A3 publication Critical patent/WO2002031152A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material

Definitions

  • This invention relates to nucleic acid and amino acid sequences of intracellular signaling molecules and to the use of these sequences in the diagnosis, treatment, and prevention of cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of intracellular signaling molecules.
  • Cell-cell communication is essential for the growth, development, and survival of multicellular organisms.
  • Cells communicate by sending and receiving molecular signals.
  • An example of a molecular signal is a growth factor, which binds and activates a specific transmembrane receptor on the surface of a target cell. The activated receptor transduces the signal intracellularly, thus initiating a cascade of biochemical reactions that ultimately affect gene transcription and cell cycle progression in the target cell.
  • Intracellular signaling is the process by which cells respond to extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.) through a cascade of biochemical reactions that begins with the binding of a signaling molecule to a cell membrane receptor and ends with the activation of an intracellular target molecule.
  • Intermediate steps in the process involve the activation of various cytoplasmic proteins by phosphorylation via protein kinases, and their deactivation by protein phosphatases, and the eventual translocation of some of these activated proteins to the cell nucleus where the transcription of specific genes is triggered.
  • the intracellular signaling process regulates all types of cell functions including cell proliferation, cell differentiation, and gene transcription, and involves a diversity of molecules including protein kinases and phosphatases, and second messenger molecules such as cyclic nucleotides, calcium-calmodulin, inositol, and various mitogens that regulate protein phosphorylation.
  • Cells also respond to changing conditions by switching off signals. Many signal transduction proteins are short-lived and rapidly targeted for degradation by covalent ligation to ubiquitin, a highly conserved small protein. Cells also maintain mechanisms to monitor changes in the concentration of denatured or unfolded proteins in membrane-bound extracytoplasmic compartments, including a transmembrane receptor that monitors the concentration of available chaperone molecules in the endoplasmic reticulum and transmits a signal to the cytosol to activate the transcription of nuclear genes encoding chaperones in the endoplasmic reticulum. Certain proteins in intracellular signaling pathways serve to link or cluster other proteins involved in the signaling cascade. These proteins are referred to as scaffold, anchoring, or adaptor proteins.
  • Protein kinases and phosphatases play a key role in the intracellular signaling process by controlling the phosphorylation and activation of various signaling proteins.
  • the high energy phosphate for this reaction is generally transferred from the adenosine triphosphate molecule (ATP) to a particular protein by a protein kinase and removed from that protein by a protein phosphatase.
  • Protem kinases are roughly divided into two groups: those that phosphorylate serine or threonine residues (serine/threonine kinases, STK) and those that phosphorylate tyrosine residues (protem tyrosine kinases, PTK).
  • a few protein kinases have dual specificity for serine/threonine and tyrosine residues. Almost all kinases contain a conserved 250-300 amino acid catalytic domain containing specific residues and sequence motifs characteristic of the kinase family (Hardie, G. and S. Hanks (1995) The Protem Kinase Facts Books. Vol 1:7-20, Academic Press, San Diego, CA).
  • STKs include the second messenger dependent protein kinases such as the cyclic -AMP dependent protein kinases (PKA), involved in mediating hormone-induced cellular responses; calcium-calmodulin (CaM) dependent protein kinases, involved in regulation of smootii muscle contraction, glycogen breakdown, and neurotransmission; and the mitogen-activated protein kinases (MAP) which mediate signal transduction from the cell surface to the nucleus via phosphorylation cascades.
  • PKA cyclic -AMP dependent protein kinases
  • CaM calcium-calmodulin dependent protein kinases
  • MAP mitogen-activated protein kinases
  • Altered PKA expression is implicated in a variety of disorders and diseases including cancer, thyroid disorders, diabetes, atherosclerosis, and cardiovascular disease (Isselbacher, KJ. et al. (1994) Harrison's Principles of Internal Medicine. McGraw-Hill, New York, NY, pp. 416-431, 1887).
  • Transmembrane PTKs are receptors for most growth factors.
  • Non-receptor PTKs lack transmembrane regions and, instead, form complexes with the intracellular regions of cell surface receptors.
  • Receptors that function through non-receptor PTKs include those for cytokines and hormones (growth hormone and prolactin) and antigen-specific receptors on T and B lymphocytes. Many of these PTKs were first identified as the products of mutant oncogenes in cancer cells in which their activation was no longer subject to normal cellular controls.
  • HPK histidine protein kinase family
  • HPKs bear little homology with mammalian STKs or PTKs but have distinctive sequence motifs of their own (Davie, J.R. et al. (1995) J. Biol. Chem. 270:19861-19867).
  • a histidine residue in the N-terminal half of the molecule (region I) is an autophosphorylation site.
  • Three additional motifs located in the C-terminal half of the molecule include an invariant asparagine residue in region ⁇ and two glycine-rich loops characteristic of nucleotide binding domains in regions Ul and IV. Recently a branched chain alpha-ketoacid dehydrogenase kinase has been found with characteristics of HPK in rat (Davie et al., supra).
  • the two principal categories of protein phosphatases are the protein (serine/threonine) phosphatases (PPs) and the protein tyrosine phosphatases (PTPs).
  • PPs dephosphorylate phosphoserine/threonine residues and are important regulators of many cAMP-mediated hormone responses (Cohen, P. (1989) Annu. Rev. Biochem. 58:453-508).
  • PTPs reverse the effects of protem tyrosine kinases and play a significant role in cell cycle and cell signaling processes (Charbonneau and Tonks, supra).
  • PTPs may prevent or reverse cell transformation and the growth of various cancers by controlling the levels of tyrosine phosphorylation in cells. This hypothesis is supported by studies showing that overexpression of PTPs can suppress transformation in cells, and that specific inhibition of PTPs can enhance cell transformation (Charbonneau and Tonks, supra). Phospholipid and Inositol-phosphate Signaling
  • Inositol phospholipids are involved in an intracellular signaling pathway that begins with binding of a signaling molecule to a G-protein linked receptor in the plasma membrane. This leads to the phosphorylation of phosphatidylinositol (PI) residues on the inner side of the plasma membrane to the biphosphate state (PIP 2 ) by inositol kinases. Simultaneously, the G-protein linked receptor binding stimulates a trimeric G-protein which in turn activates a phosphoinositide-specific phospholipase C- ⁇ .
  • PI phosphatidylinositol
  • IP 3 inositol triphosphate
  • diacylglycerol acts as mediators for separate signaling events.
  • IP 3 diffuses through the plasma membrane to induce calcium release from the endoplasmic reticulum (ER), while diaacylglycerol remains in the membrane and helps activate protein kinase C, a serine-threonine kinase that phosphorylates selected proteins in the target cell.
  • the calcium response initiated by IP 3 is terminated by the dephosphorylation of IP 3 by specific inositol phosphatases.
  • Cellular responses that are mediated by this pathway are glycogen breakdown in the liver in response to vasopressin, smooth muscle contraction in response to acetylcholine, and thrombin-induced platelet aggregation.
  • Oxysterols are oxygenated derivatives of cholesterol and have a wide range of biological activities. Oxysterols mediate cholesterol homeostasis, steroid biosynthesis and sphingolipid metabolism within the cell, but can also diffuse through the plasma membrane and act as extracellular messengers, affecting such processes as platelet aggregation, cell growth and apoptosis. Oxysterols interact with a number of receptors, including the oxysterol binding protein (OSBP), the sterol regulatory element binding protein, the cellular nucleic acid binding protein, the LXR nuclear hormone receptors, and the LDL receptor (for a review, see Schroepfer, G.J. (2000) Physiol. Rev. 80:361-554).
  • OSBP oxysterol binding protein
  • sterol regulatory element binding protein the sterol regulatory element binding protein
  • the cellular nucleic acid binding protein the LXR nuclear hormone receptors
  • LDL receptor for a review, see Schroepfer, G.J. (2000) Physiol
  • OSBP is a high-affinity intracellular receptor for a variety of oxysterols that down-regulate cholesterol synthesis and stimulate cholesterol esterification.
  • OSBP Upon ligand binding, OSBP translocates from the cytoplasm to the Golgi. This movement seems to be dependent on the presence of a pleckstrin homology domain (Lagace, T.A. et al. (1997) Biochem. J. 326:205-213).
  • the oxysterol-induced apoptosis of leukemic T-cells seems to be mediated by OSBP occupancy (Bakos, J.T. et al. (1993) J. Steroid Biochem. Mol. Biol. 46:415-426).
  • Cyclic Nucleotide Signaling Cyclic nucleotides function as intracellular second messengers to transduce a variety of extracellular signals including hormones, light, and neurotransmitters.
  • cyclic-AMP dependent protein kinases PKA
  • PKA cyclic-AMP dependent protein kinases
  • Visual excitation and the phototransmission of light signals in the eye is controlled by cyclic-GMP regulated, Ca 2+ -specific channels. Because of the importance of cellular levels of cyclic nucleotides in mediating these various responses, regulating the synthesis and breakdown of cyclic nucleotides is an important matter.
  • adenylyl cyclase which synthesizes cAMP from AMP, is activated to increase cAMP levels in muscle by binding of adrenaline to ⁇ -adrenergic receptors, while activation of guanylate cyclase and increased cGMP levels in photoreceptors leads to reopening of the Ca 2+ -specific channels and recovery of the dark state in the eye.
  • hydrolysis of cyclic nucleotides by cAMP and cGMP-specific phosphodiesterases (PDEs) produces the opposite of these and other effects mediated by increased cyclic nucleotide levels.
  • PDEs appear to be particularly important in the regulation of cyclic nucleotides, considering the diversity found in this family of proteins. At least seven families of mammalian PDEs (PDE1-7) have been identified based on substrate specificity and affinity, sensitivity to cofactors, and sensitivity to inhibitory drugs (Beavo, J.A. (1995) Physiol. Rev. 75:725-748). PDE inhibitors have been found to be particularly useful in treating various clinical disorders. Rolipram, a specific inhibitor of PDE4, has been used in the treatment of depression, and similar inhibitors are undergoing evaluation as anti-inflammatory agents. Theophylline is a nonspecific PDE inhibitor used in the treatment of bronchial asthma and other respiratory diseases (Banner, K.H. and CP. Page (1995) Eur. Respir. J. 8:996-1000). G-Protein Signaling
  • G-proteins are critical mediators of signal transduction between a particular class of extracellular receptors, the G-protein coupled receptors (GPCRs), and intracellular second messengers such as cAMP and Ca 2+ .
  • G-proteins are linked to the cytosolic side of a GPCR such that activation of the GPCR by ligand binding stimulates binding of the G-protein to GTP, inducing an "active" state in the G-protein. In the active state, the G-protein acts as a signal to trigger other events in the cell such as the increase of cAMP levels or the release of Ca 2+ into the cytosol from the ER, which, in turn, regulate phosphorylation and activation of other intracellular proteins.
  • G-proteins consisting of three different subunits, and monomeric, low molecular weight (LMW), G-proteins consisting of a single polypeptide chain.
  • LMW low molecular weight
  • the three polypeptide subunits of heterotrimeric G-proteins are the ⁇ , ⁇ , and ⁇ subunits. The subunit binds and hydrolyzes GTP.
  • the ⁇ and ⁇ subunits form a tight complex that anchors the protein to the inner side of the plasma membrane.
  • the ⁇ subunits also known as G- ⁇ proteins or ⁇ transducins, contain seven tandem repeats of the WD-repeat sequence motif, a motif found in many proteins with regulatory functions. Mutations and variant expression of ⁇ transducfn proteins are linked with various disorders (Neer, E. J. et al. (1994) Nature 371 :297-300; Margottin, F. et al. (1998) Mol. Cell. 1:565-574).
  • LMW GTP-proteins are GTPases which regulate cell growth, cell cycle control, protein secretion, and intracellular vesicle interaction. They consist of single polypeptides which, like the ⁇ subunit of the heterotrimeric G-proteins, are able to bind and hydrolyze GTP, thus cycling between an inactive and an active state. At least sixty members of the LMW G-protein superfamily have been identified and are currently grouped into the six subfamilies of ras, rho, arf, sari, ran, and rab. Activated ras genes were initially found in human cancers, and subsequent studies confirmed that ras function is critical in determining whether cells continue to grow or become differentiated. Other members of the LMW G-protein superfamily have roles in signal transduction that vary with the function of the activated genes and the locations of the G-proteins.
  • Guanine nucleotide exchange factors regulate the activities of LMW G-proteins by determining whether GTP or GDP is bound.
  • GTPase-activating protein GAP
  • GTP-ras GTPase-activating protein
  • GDP-ras guanine nucleotide releasing protein
  • RGS G-protein signaling
  • cytokine interleukin
  • Ca 2+ is another second messenger molecule that is even more widely used as an intracellular mediator than cAMP.
  • Ca 2+ can enter the cytosol by two pathways, in response to extracellular signals.
  • One pathway acts primarily in nerve signal transduction where Ca 2+ enters a nerve terminal through a voltage-gated Ca 2+ channel.
  • the second is a more ubiquitous pathway in which Ca 2+ is released from the ER into the cytosol in response to binding of an extracellular signaling molecule to a receptor.
  • Ca 2+ directly activates regulatory enzymes, such as protein kinase C, which trigger signal transduction pathways.
  • Ca 2+ also binds to specific Ca + -binding proteins (CBPs) such as calmodulin (CaM) which then activate multiple target proteins in the cell including enzymes, membrane transport pumps, and ion channels.
  • CBPs Ca + -binding proteins
  • CaM interactions are involved in a multitude of cellular processes including, but not limited to, gene regulation, DNA synthesis, cell cycle progression, mitosis, cytokinesis, cytoskeletal organization, muscle contraction, signal transduction, ion homeostasis, exocytosis, and metabolic regulation (Celio, M.R. et al. (1996) Guidebook to Calcium-binding Proteins. Oxford University Press, Oxford, UK, pp. 15-20).
  • Ca 2+ binding proteins are characterized by the presence of one or more EF-hand Ca 2+ binding motifs, which are comprised of 12 amino acids flanked by ⁇ -helices (Celio, supra).
  • the regulation of CBPs has implications for the control of a variety of disorders.
  • Calcineurin a CaM-regulated protein phosphatase, is a target for inhibition by the immunosuppressive agents cyclosporin and FK506. This indicates the importance of calcineurin and CaM in the immune response and immune disorders (Schwaninger M. et al. (1993) J. Biol Chem. 268:23111-23115).
  • the level of CaM is increased several-fold in tumors and tumor-derived cell lines for various types of cancer (Rasmussen, CD. and A.R. Means (1989) Trends Neurosci. 12:433-438).
  • the annexins are a family of calcium-binding proteins that associate with the cell membrane (Towle, CA. and B.V. Treadwell (1992) J. Biol. Chem. 267:5416-5423). Annexins reversibly bind to negatively charged phospholipids (phosphatidylcholine and phosphatidylserine) in a calcium dependent manner.
  • Annexins participate in various processes pertaining to signal transduction at the plasma membrane, including membrane-cytoskeleton interactions, phospholipase inhibition, anticoagulation, and membrane fusion. Annexins contain four to eight repeated segments of about 60 residues. Each repeat folds into five alpha helices wound into a right-handed superhelix. Signaling Complex Protein Domains
  • PDZ domains were named for three proteins in which this domain was initially discovered. These proteins include PSD-95 (postsynaptic density 95), Dig (Drosophila lethal(l)discs large-1), and ZO-1 (zonula occludens-1). These proteins play important roles in neuronal synaptic transmission, tumor suppression, and cell junction formation, respectively. Since the discovery of these proteins, over sixty additional PDZ-containing proteins have been identified in diverse prokaryotic and eukaryotic organisms. This domain has been implicated in receptor and ion channel clustering and in the targeting of multiprotein signaling complexes to specialized functional regions of the cytosolic face of the plasma membrane. (For a review of PDZ domain-containing proteins, see Ponting, CP.
  • PDZ domains are found in the eukaryotic MAGUK (membrane-associated guanylate kinase) protein family, members of which bind to the intracellular domains of receptors and channels.
  • MAGUK membrane-associated guanylate kinase
  • PDZ domains are also found in diverse membrane-localized proteins such as protein tyrosine phosphatases, serine/threonine kinases, G-protein cofactors, and synapse-associated proteins such as syntrophins and neuronal nitric oxide synthase (nNOS).
  • the glutamate receptor interacting protein contains seven PDZ domains. GRIP is an adaptor that links certain glutamate receptors to other proteins and may be responsible for the clustering of these receptors at excitatory synapses in the brain (Dong, H. et al. (1997) Nature 386:279-284).
  • the Drosophila scribble (SCRIB) protein contains both multiple PDZ domains and leucine-rich repeats.
  • SCRIB is located at the epithelial septate junction, which is analogous to the vertebrate tight junction, at the boundary of the apical and basolateral cell surface. SCRIB is involved in the distribution of apical proteins and correct placement of adherens junctions to the basolateral cell surface (Bilder, D. and N. Perrimon (2000) Nature 403:676-680).
  • the PX domain is an example of a domain specialized for promoting protein-protein interactions. The PX domain is found in sorting nexins and in a variety of other proteins, including the PhoX components of NADPH oxidase and the Cpk class of phosphatidylinositol 3-kinase.
  • PX domains contain a polyproline motif which is characteristic of SH3 domain-binding proteins (Ponting, CP. (1996) Protein Sci. 5:2353-2357).
  • SH3 domain-mediated interactions involving the PhoX components of NADPH oxidase play a role in the formation of the NADPH oxidase multi-protein complex (Leto, T.L. et al. (1994) Proc. Natl. Acad. Sci. USA 91:10650-10654; Wilson, L. et al. (1997) Inflamm. Res. 46:265-271).
  • the SH3 domain is defined by homology to a region of the proto-oncogene c-Src, a cytoplasmic protein tyrosine kinase.
  • SH3 is a small domain of 50 to 60 amino acids that interacts with proline-rich ligands. SH3 domains are found in a variety of eukaryotic proteins involved in signal transduction, cell polarization, and membrane-cytoskeleton interactions. In some cases, SH3 domain- containing proteins interact directly with receptor tyrosine kinases.
  • the SLAP-130 protein is a substrate of the T-cell receptor (TCR) stimulated protein kinase.
  • SLAP-130 interacts via its SH3 domain with the protein SLP-76 to affect the TCR-induced expression of interleukin-2 (Musci, M. A. et al. (1997) J. Biol. Chem. 272: 11674-11677).
  • Another recently identified SH3 domain protein is macrophage actin-associated tyrosine-phosphorylated protein (MAYP) which is phosphorylated during the response of macrophages to colony stimulating factor- 1 (CSF-1) and is likely to play a role in regulating the CSF-1-induced reorganization of the actin cytoskeleton (Yeung, Y.-G. et al. (1998) J. Biol. Chem. 273:30638-30642).
  • the structure of the SH3 domain is characterized by two antiparallel beta sheets packed against each other at right angles. This packing forms a hydrophobic pocket lined with residues that are highly conserved between different SH3 domains. This pocket makes critical hydrophobic contacts with proline residues in the ligand (Feng, S. et al. (1994) Science 266:1241- 1247).
  • a novel domain, called the WW domain resembles the SH3 domain in its ability to bind proline-rich ligands. This domain was originally discovered in dystrophin, a cytoskeletal protein with direct involvement in Duchenne muscular dystrophy (Bork, P. and M. Sudol (1994) Trends Biochem. Sci. 19:531-533). WW domains have since been discovered in a variety of intracellular signaling molecules involved in development, cell differentiation, and cell proliferation. The structure of the WW domain is composed of beta strands grouped around four conserved aromatic residues, generally tryptophan.
  • SH2 domain is defined by homology to a region of c-Src.
  • SH2 domains interact directly with phospho-tyrosine residues, thus providing an immediate mechanism for the regulation and transduction of receptor tyrosine kinase-mediated signaling pathways.
  • SH2 domains are capable of binding to phosphorylated tyrosine residues in the activated PDGF receptor, thereby providing a highly coordinated and finely tuned response to ligand-mediated receptor activation.
  • the GSG domain (GRP33, Sam68, GLD-1) and the KH domain (an RNA binding domain), are found within Sam68, a 68-kDa Src substrate associated during mitosis protein, which is an RNA- binding protein with signaling properties. It is known to be a substrate for Src-family tyrosine kinases during mitosis and associates with various SH3 and SH2 domain-containing signaling molecules.
  • SLM-1 and SLM-2 (Sam68-like mammalian) proteins have sequence identity with Sam68, also contain the GSG domain, have proline-rich motifs, arginine-gylcine repeats, and a C-terminal tyrosine-rich region.
  • SLM-1 is a Src substrate during mitosis, suggesting a possible involvement in the steps of mitosis. It has been suggested by Di Fruscio et al. that Sam68/SLM defines a family in which the members have the potential to link tyrosine kinase signaling cascades with some aspects of RNA metabolism, possibly as multifunctional adapter proteins during mitosis (Di Fruscio, M. et al. (1999) Proc. Natl. Acad. Sci. U.S.A. 96:2710-2715.) The pleckstrin homology (PH) domain was originally identified in pleckstrin, the predominant substrate for protein kinase C in platelets.
  • PH pleckstrin homology
  • Proteins containing the pleckstrin homology domain include a variety of kinases, phospholipase-C isoforms, guanine nucleotide release factors, and GTPase activating proteins.
  • members of the FGD1 family contain both Rho-guanine nucleotide exchange factor (GEF) and PH domains, as well as a FYVE zinc finger domain.
  • GEF Rho-guanine nucleotide exchange factor
  • FGD1 is the gene responsible for faciogenital dysplasia, an inherited skeletal dysplasia (Pasteris, N.G. and J.L. Gorski (1999) Genomics 60:57-66).
  • PH domain proteins function in association with the plasma membrane, and this association appears to be mediated by the PH domain itself.
  • PH domains share a common structure composed of two antiparallel beta sheets flanked by an amphipathic alpha helix. Variable loops connecting the component beta strands generally occur within a positively charged environment and may function as ligand binding sites (Lemmon, M.A. et al. (1996) Cell 85:621-624).
  • Ankyrin (ANK) repeats mediate protein-protein interactions associated with diverse intracellular signaling functions.
  • ANK repeats are found in proteins involved in cell proliferation such as kinases, kinase inhibitors, tumor suppressors, and cell cycle control proteins. (See, for example, Kalus, W.
  • Myotrophin is an ANK repeat protein that plays a key role in the development of cardiac hypertrophy, a contributing factor to many heart diseases. Structural studies show that the myotrophin ANK repeats, like other ANK repeats, each form a helix-turn-helix core preceded by a protruding "tip.” These tips are of variable sequence and may play a role in protein- protein interactions. The helix-turn-helix region of the ANK repeats stack on top of one another and are stabilized by hydrophobic interactions (Yang, Y. et al. (1998) Structure 6:619-626).
  • TPR tetratricopeptide repeat
  • CDC16, CDC23, and CDC27 members of the anaphase promoting complex which targets proteins for degradation at the onset of anaphase.
  • Other processes involving TPR proteins include cell cycle control, transcription repression, stress response, and protein kinase inhibition (Lamb, J.R. et al. (1995) Trends Biochem. Sci. 20:257-259).
  • the armadillo/beta-catenin repeat is a 42 amino acid motif which forms a superhelix of alpha helices when tandemly repeated.
  • the structure of the armadillo repeat region from beta-catenin revealed a shallow groove of positive charge on one face of the superhelix, which is a potential binding surface.
  • the armadillo repeats of beta-catenin, plakoglobin, and pl20 cas bind the cytoplasmic domains of cadherins.
  • Beta-catenin/cadherin complexes are targets of regulatory signals that govern cell adhesion and mobility (Huber, A.H. et al. (1997) Cell 90:871-882).
  • G-beta beta-transducin
  • alpha, beta, and gamma beta-transducin
  • G proteins guanine nucleotide-binding proteins
  • the invention features purified polypeptides, intracellular signaling molecules, referred to collectively as “INTSIG” and individually as “INTSIG-1,” 'TNTSIG-2,” 'TNTSIG-3,” 'TNTSIG-4,” “ ⁇ NTSIG-5,” “ ⁇ NTSIG-6,” “ ⁇ NTSIG-7,” “ ⁇ NTSIG-8,” “ ⁇ NTSIG-9,” “ ⁇ NTSIG-IO,” “ ⁇ NTSIG- ⁇ ,” 'TNTSIG-12,” “INTSIG-13,” “INTSIG-14,” “INTSIG-15,” 'TNTSIG-16,” “INTSIG-17,” 'TNTSIG-
  • the invention provides an isolated polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 1-20.
  • the invention further provides an isolated polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:l- 20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ D3 NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-20.
  • the polynucleotide encodes a polypeptide selected from the group consisting of SEQ DD NO: 1-20.
  • the polynucleotide is selected from the group consisting of SEQ ID NO:21-40.
  • the invention provides a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ED NO: 1
  • the invention also provides a method for producing a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the method comprises a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recovering the polypeptide so expressed.
  • the invention provides an isolated antibody which specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the invention further provides an isolated polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d).
  • the polynucleotide comprises at least 60 contiguous nucleotides.
  • the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide .
  • a target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d).
  • the method comprises 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.
  • the probe comprises at least 60 contiguous nucleotides.
  • the invention further provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d).
  • the method comprises 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.
  • the invention further provides a composition comprising an effective amount of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and a pharmaceutically acceptable excipient.
  • the composition comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the invention additionally provides a method of treating a disease or condition associated with decreased expression of functional INTSIG, comprising administering to a patient in need of such treatment the composition.
  • the invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample.
  • the invention provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient.
  • the invention provides a method of treating a disease or condition associated with decreased expression of functional INTSIG, comprising administering to a patient in need of such treatment the composition.
  • the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample.
  • the invention provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient.
  • the invention provides a method of treating a disease or condition associated with overexpression of functional INTSIG, comprising administering to a patient in need of such treatment the composition.
  • the invention further provides a method of screening for a compound that specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
  • the method comprises a) combining the polypeptide with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide to the test compound, thereby identifying a compound that specifically binds to the polypeptide.
  • the invention further provides a method of screening for a compound that modulates the activity of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20.
  • the method comprises a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.
  • the invention further provides a method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, the method comprising a) exposing a sample comprising the target polynucleotide to a compound, and b) detecting altered expression of the target polynucleotide.
  • the invention further provides a method for assessing toxicity of a test compound, said 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 selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, iii) a polynucleotide having a sequence complementary to i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv
  • Hybridization occurs under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, iii) a polynucleotide complementary to the polynucleotide of i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv).
  • the target polynucleotide comprises a fragment of a polynucleotide sequence selected from the group consisting of i)-v) above; 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.
  • Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the present invention.
  • Table 2 shows the GenBank identification number and annotation of the nearest GenBank homolog for polypeptides of the invention. The probability score for the match between each polypeptide and its GenBank homolog is also shown.
  • Table 3 shows structural features of polypeptide sequences of the invention, including predicted motifs and domains, along with the methods, algorithms, and searchable databases used for analysis of the polypeptides.
  • Table 4 lists the cDNA and/or genomic DNA fragments which were used to assemble polynucleotide sequences of the invention, along with selected fragments of the polynucleotide sequences.
  • Table 5 shows the representative cDNA library for polynucleotides of the invention.
  • Table 6 provides an appendix which describes the tissues and vectors used for construction of the cDNA libraries shown in Table 5.
  • Table 7 shows the tools, programs, and algorithms used to analyze the polynucleotides and polypeptides of the invention, along with applicable descriptions, references, and threshold parameters.
  • a reference to “a host cell” includes a plurality of such host cells
  • 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.
  • all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs.
  • any machines, materials, and methods similar or equivalent to those described herein can be used to practice or test the present invention, the preferred machines, materials and methods are now described. All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. None herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention. DEFINITIONS
  • INTSIG refers to the amino acid sequences of substantially purified INTSIG obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, murine, equine, and human, and from any source, whether natural, synthetic, semi-synthetic, or recombinant.
  • agonist refers to a molecule which intensifies or mimics the biological activity of INTSIG.
  • Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of INTSIG either by directly interacting with INTSIG or by acting on components of the biological pathway in which INTSIG participates.
  • allelic variant is an alternative form of the gene encoding INTSIG. Allelic variants may result from at least one mutation in the nucleic acid sequence and may result in altered mRNAs or in polypeptides whose structure or function may or may not be altered. A gene may have none, one, or many allelic variants of its naturally occurring form. Common mutational changes which give rise to allelic variants are generally ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence.
  • altered nucleic acid sequences encoding INTSIG include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as INTSIG or a polypeptide with at least one functional characteristic of INTSIG. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding INTSIG, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding INTSIG.
  • the encoded protein may also be "altered,” and may contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent INTSIG.
  • Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of INTSIG is retained.
  • negatively charged amino acids may include aspartic acid and glutamic acid
  • positively charged amino acids may include lysine and arginine.
  • Amino acids with uncharged polar side chains having similar hydrophilicity values may ' include: asparagine and glutamine; and serine and threonine.
  • Amino acids with uncharged side chains having similar hydrophilicity values may include: leucine, isoleucine, and valine; glycine and alanine; and phenylalanine and tyrosine.
  • amino acid and amino acid sequence refer to an oligopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. Where "amino acid sequence” is recited to refer to a sequence of a naturally occurring protein molecule, “amino acid sequence” and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule. "Amplification” relates to the production of additional copies of a nucleic acid sequence.
  • Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art.
  • PCR polymerase chain reaction
  • Antagonist refers to a molecule which inhibits or attenuates the biological activity of INTSIG.
  • Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of rNTSIG either by directly interacting with INTSIG or by acting on components of the biological pathway in which INTSIG participates.
  • antibody refers to intact immunoglobulin molecules as well as to fragments thereof, such as Fab, F(ab') 2 , and Fv fragments, which are capable of binding an epitopic determinant.
  • Antibodies that bind INTSIG polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen.
  • the polypeptide or oligopeptide used to immunize an animal e.g., a mouse, a rat, or a rabbit
  • an animal e.g., a mouse, a rat, or a rabbit
  • Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal.
  • antigenic determinant refers to that region of a molecule (i.e., an epitope) that makes contact with a particular antibody.
  • an antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody.
  • aptamer refers to a nucleic acid or oligonucleotide molecule that binds to a specific molecular target.
  • Aptamers are derived from an in vitro evolutionary process (e.g., SELEX (Systematic Evolution of Ligands by Exponential Enrichment), described in U.S. Patent No. 5,270,163), which selects for target-specific aptamer sequences from large combinatorial libraries.
  • Aptamer compositions may be double-stranded or single-stranded, and may include deoxyribonucleotides, ribonucleotides, nucleotide derivatives, or other nucleotide-like molecules.
  • the nucleotide components of an aptamer may have modified sugar groups (e.g., the 2'-OH group of a ribonucleotide may be replaced by 2'-F or 2'-NH 2 ), which may improve a desired property, e.g., resistance to nucleases or longer lifetime in blood.
  • Aptamers may be conjugated to other molecules, e.g., a high molecular weight carrier to slow clearance of the aptamer from the circulatory system.
  • Aptamers may be specifically cross-linked to their cognate ligands, e.g., by photo-activation of a cross-linker. (See, e.g., Brody, E.N. and L. Gold (2000) J. Biotechnol. 74:5-13.)
  • RNA aptamer refers to an aptamer which is expressed in vivo.
  • a vaccinia virus-based RNA expression system has been used to express specific RNA aptamers at high levels in the cytoplasm of leukocytes (Blind, M. et al. (1999) Proc. Natl Acad. Sci. USA 96:3606-3610).
  • spiegelmer refers to an aptamer which includes L-DNA, L-RNA, or other left- handed nucleotide derivatives or nucleotide-like molecules. Aptamers containing left-handed nucleotides are resistant to degradation by naturally occurring enzymes, which normally act on substrates containing right-handed nucleotides.
  • antisense refers to any composition capable of base-pairing with the "sense" (coding) strand of a specific nucleic acid sequence.
  • Antisense compositions may include DNA; RNA; peptide nucleic acid (PNA); oligonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; oligonucleotides having modified sugar groups such as 2-methoxyethyl sugars or 2'-methoxyethoxy sugars; or oligonucleotides having modified bases such as 5-methyl cytosine, 2'-deoxyuracil, or 7-deaza-2'-deoxyguanosine.
  • Antisense molecules may be produced by any method including chemical synthesis or transcription. Once introduced into a cell, the complementary antisense molecule base-pairs with a naturally occurring nucleic acid sequence produced by the cell to form duplexes which block either transcription or translation.
  • the designation "negative” or “minus” can refer to the antisense strand, and the designation “positive” or “plus” can refer to the sense strand of a reference DNA molecule.
  • the term “biologically active” refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule.
  • immunologically active or “immunogenic” refers to the capability of the natural, recombinant, or synthetic INTSIG, or of any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.
  • “Complementary” describes the relationship between two single-stranded nucleic acid sequences that anneal by base-pairing. For example, 5'-AGT-3' pairs with its complement, 3'-TCA-5'.
  • composition comprising a given polynucleotide sequence and a “composition comprising a given amino acid sequence” refer broadly to any composition containing the given polynucleotide or amino acid sequence.
  • the composition may comprise a dry formulation or an aqueous solution.
  • compositions comprising polynucleotide sequences encoding INTSIG or fragments of INTSIG may be employed as hybridization probes.
  • the probes may be stored in freeze-dried form and may be associated with a stabilizing agent such as a carbohydrate.
  • the probe may be deployed in an aqueous solution containing salts (e.g., NaCl), detergents (e.g., sodium dodecyl sulfate; SDS), and other components (e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.).
  • salts e.g., NaCl
  • detergents e.g., sodium dodecyl sulfate
  • other components e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.
  • Consensus sequence refers to a nucleic acid sequence which has been subjected to repeated DNA sequence analysis to resolve uncalled bases, extended using the XL-PCR kit (Applied Biosystems, Foster City CA) in the 5' and/or the 3' direction, and resequenced, or which has been assembled from one or more overlapping cDNA, EST, or genomic DNA fragments using a computer program for fragment assembly, such as the GELVIEW fragment assembly system (GCG, Madison WI) or Phrap (University of Washington, Seattle WA). Some sequences have been both extended and assembled to produce the consensus sequence.
  • Constant amino acid substitutions are those substitutions that are predicted to least interfere with the properties of the original protein, i.e., the structure and especially the function of the protein is conserved and not significantly changed by such substitutions.
  • the table below shows amino acids which may be substituted for an original amino acid in a protein and which are regarded as conservative amino acid substitutions.
  • Tyr His, Phe, Trp VaT lie, Leu, Thr
  • Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.
  • a “deletion” refers to a change in the amino acid or nucleotide sequence that results in the absence of one or more amino acid residues or nucleotides.
  • derivative refers to a chemically modified polynucleotide or polypeptide. Chemical modifications of a polynucleotide can include, for example, replacement of hydrogen by an alkyl, acyl, hydroxyl, or amino group.
  • a derivative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule.
  • a derivative polypeptide is one modified by glycosylation, pegylation, or any similar process that retains at least one biological or immunological function of the polypeptide from which it was derived.
  • a “detectable label” refers to a reporter molecule or enzyme that is capable of generating a measurable signal and is covalently or noncovalently joined to a polynucleotide or polypeptide.
  • “Differential expression” refers to increased or upregulated; or decreased, downregulated, or absent gene or protein expression, determined by comparing at least two different samples. Such comparisons may be carried out between, for example, a treated and an untreated sample, or a diseased and a normal sample.
  • Exon shuffling refers to the recombination of different coding regions (exons). Since an exon may represent a structural or functional domain of the encoded protein, new proteins may be assembled through the novel reassortment of stable substructures,°thus allowing acceleration of the evolution of new protein functions.
  • a “fragment” is a unique portion of INTSIG or the polynucleotide encoding INTSIG which is identical in sequence to but shorter in length than the parent sequence.
  • a fragment may comprise up to the entire length of the defined sequence, minus one nucleotide/amino acid residue.
  • a fragment may comprise from 5 to 1000 contiguous nucleotides or amino acid residues.
  • a fragment used as a probe, primer, antigen, therapeutic molecule, or for other purposes may be at least 5, 10, 15, 16, 20, 25, 30, 40, 50, 60, 75, 100, 150, 250 or at least 500 contiguous nucleotides or amino acid residues in length. Fragments may be preferentially selected from certain regions of a molecule.
  • a polypeptide fragment may comprise a certain length of contiguous amino acids selected from the first 250 or 500 amino acids (or first 25% or 50%) of a polypeptide as shown in a certain defined sequence.
  • these lengths are exemplary, and any length that is supported by the specification, including the Sequence Listing, tables, and figures, may be encompassed by the present embodiments.
  • a fragment of SEQ DD NO :21-40 comprises a region of unique polynucleotide sequence that specifically identifies SEQ DD NO:21-40, for example, as distinct from any other sequence in the genome from which the fragment was obtained.
  • a fragment of SEQ DD NO:21-40 is useful, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ DD NO:21-40 from related polynucleotide sequences.
  • the precise length of a fragment of SEQ DD NO:21-40 and the region of SEQ DD NO:21-40 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.
  • a fragment of SEQ DD NO: 1-20 is encoded by a fragment of SEQ DD NO:21-40.
  • a fragment of SEQ DD NO: 1-20 comprises a region of unique amino acid sequence that specifically identifies SEQ DD NO: 1-20.
  • a fragment of SEQ DD NO: 1-20 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ DD NO: 1-20.
  • the precise length of a fragment of SEQ DD NO: 1-20 and the region of SEQ DD NO: 1-20 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.
  • a “full length” polynucleotide sequence is one containing at least a translation initiation codon (e.g., methionine) followed by an open reading frame and a translation termination codon.
  • a “full length” polynucleotide sequence encodes a “full length” polypeptide sequence.
  • “Homology” refers to sequence similarity or, interchangeably, sequence identity, between two or more polynucleotide sequences or two or more polypeptide sequences.
  • percent identity and % identity refer to the percentage of residue matches between at least two polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
  • NCBI National Center for Biotechnology Information
  • BLAST Basic Local Alignment Search Tool
  • NCBI National Center for Biotechnology Information
  • BLAST Basic Local Alignment Search Tool
  • the BLAST software suite includes various sequence analysis programs including "blastn,” that is used to align a known polynucleotide sequence with other polynucleotide sequences from a variety of databases.
  • BLAST 2 Sequences are commonly used with gap and other parameters set to default settings. For example, to compare two nucleotide sequences, one may use blastn with the "BLAST 2 Sequences” tool Version 2.0.12 (April-21-2000) set at default parameters. Such default parameters may be, for example: Matrix: BLOSUM62
  • Percent identity may be measured over the length of an entire defined sequence, for example, as defined by a particular SEQ DD number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined sequence, for instance, a fragment of at least 20, at least 30, at least 40, at least 50, at least 70, at least 100, or at least 200 contiguous nucleotides.
  • Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures, or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
  • Nucleic acid sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein.
  • the phrases "percent identity” and "% identity,” as applied to polypeptide sequences refer to the percentage of residue matches between at least two polypeptide sequences aligned using a standardized algorithm. Methods of polypeptide sequence alignment are well-known. Some alignment methods take into account conservative amino acid substitutions. Such conservative substitutions, explained in more detail above, generally preserve the charge and_hydrophobicity at the site of substitution, thus preserving the structure (and therefore function) of the polypeptide.
  • NCBI BLAST software suite may be used.
  • BLAST 2 Sequences Version 2.0.12 (April-21-2000) with blastp set at default parameters.
  • Such default parameters may be, for example:
  • Percent identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ DD number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues.
  • Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
  • HACs Human artificial chromosomes
  • HACs are linear rmcrocrrromosomes which may contain DNA sequences of about 6 kb to 10 Mb in size and which contain all of the elements required for chromosome replication, segregation and maintenance.
  • humanized antibody refers to an antibody molecule in which the amino acid sequence in the non-antigen binding regions has been altered so that the antibody more closely resembles a human antibody, and still retains its original binding ability.
  • Hybridization refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of complementarity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after the "washing" step(s). The washing step(s) is particularly important in determining the stringency of the hybridization process, with more stringent conditions allowing less non-specific binding, i.e., binding between pairs of nucleic acid strands that are not perfectly matched.
  • Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve the desired stringency, and therefore hybridization specificity. Permissive annealing conditions occur, for example, at 68°C in the presence of about 6 x SSC, about 1% (w/v) SDS, and about 100 ⁇ g/ml sheared, denatured salmon sperm DNA.
  • wash temperatures are typically selected to be about 5°C to 20°C lower than the thennal melting point (T m ) for the specific sequence at a defined ionic strength and pH.
  • T m is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • High stringency conditions for hybridization between polynucleotides of the present invention include wash conditions of 68°C in the presence of about 0.2 x SSC and about 0.1% SDS, for 1 hour. Alternatively, temperatures of about 65°C, 60°C, 55°C, or 42°C may be used. SSC concentration may be varied from about 0.1 to 2 x SSC, with SDS being present at about 0.1%.
  • blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 ⁇ g/ml.
  • Organic solvent such as formamide at a concentration of about 35-50% v/v
  • RNA:DNA hybridizations Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art.
  • Hybridization particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is strongly indicative of a similar role for the nucleotides and their encoded polypeptides.
  • hybridization complex refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases.
  • a hybridization complex may be formed in solution (e.g., C 0 t or R 0 t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed).
  • insertion and “addition” refer to changes in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively.
  • Immuno response can refer to conditions associated with inflammation, trauma, immune disorders, or infectious or genetic disease, etc. These conditions can be characterized by expression of various factors, e.g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems.
  • factors e.g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems.
  • an “immunogenic fragment” is a polypeptide or oligopeptide fragment of INTSIG which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal.
  • the term “immunogenic fragment” also includes any polypeptide or oligopeptide fragment of INTSIG which is useful in any of the antibody production methods disclosed herein or known in the art.
  • microarray refers to an arrangement of a plurality of polynucleotides, polypeptides, or other chemical compounds on a substrate.
  • element and “array element” refer to a polynucleotide, polypeptide, or other chemical compound having a unique and defined position on a microarray.
  • modulate refers to a change in the activity of INTSIG. For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of INTSIG.
  • nucleic acid and nucleic acid sequence refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof. These phrases also refer to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA), or to any DNA-like or RNA-like material.
  • PNA peptide nucleic acid
  • operably linked refers to the situation in which a first nucleic acid sequence is placed in a functional relationship with a second nucleic acid sequence.
  • a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence.
  • Operably linked DNA sequences may be in close proximity or contiguous and, where necessary to join two protein coding regions, in the same reading frame.
  • PNA protein nucleic acid
  • PNA refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition. PNAs preferentially bind complementary single stranded DNA or RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell.
  • Post-translational modification of an INTSIG may involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and other modifications known in the art. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cell type depending on the enzymatic milieu of INTSIG.
  • Probe refers to nucleic acid sequences encoding INTSIG, their complements, or fragments thereof, which are used to detect identical, allelic or related nucleic acid sequences.
  • Probes are isolated oligonucleotides or polynucleotides attached to a detectable label or reporter molecule. Typical labels include radioactive isotopes, ligands, chemiluminescent agents, and enzymes.
  • Primmers are short nucleic acids, usually DNA oligonucleotides, which may be annealed to a target polynucleotide by complementary base-pairing. The primer may then be extended along the target DNA strand by a DNA polymerase enzyme.
  • Probes and primers as used in the present invention typically comprise at least 15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and primers may also be employed, such as probes and primers that comprise at least 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or at least 150 consecutive nucleotides of the disclosed nucleic acid sequences. Probes and primers may be considerably longer than these examples, and it is understood that any length supported by the specification, including the tables, figures, and Sequence Listing, may be used.
  • PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambridge MA). Oligonucleotides for use as primers are selected using software known in the art for such purpose. For example, OLIGO 4.06 software is useful for the selection of PCR primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases. Similar primer selection programs have incorporated additional features for expanded capabilities.
  • the PrimOU primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas TX) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope.
  • the Primer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambridge MA) allows the user to input a "misprinting library," in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarray s.
  • the source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.
  • the PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences. Hence, this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments.
  • oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of oligonucleotide selection are not limited to those described above.
  • a "recombinant nucleic acid” is a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two or more otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques such as those described in Sambrook, supra.
  • the term recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion of a portion of the nucleic acid.
  • a recombinant nucleic acid may include a nucleic acid sequence operably linked to a promoter sequence. Such a recombinant nucleic acid may be part of a vector that is used, for example, to transform a cell.
  • such recombinant nucleic acids may be part of a viral vector, e.g., based on a vaccinia virus, that could be use to vaccinate a mammal wherein the recombinant nucleic acid is expressed, inducing a protective immunological response in the mammal.
  • a “regulatory element” refers to a nucleic acid sequence usually derived from untranslated regions of a gene and includes enhancers, promoters, introns, and 5' and 3' untranslated regions (UTRs). Regulatory elements interact with host or viral proteins which control transcription, translation, or RNA stability.
  • Reporter molecules are chemical or biochemical moieties used for labeling a nucleic acid, amino acid, or antibody. Reporter molecules include radionuclides; enzymes; fluorescent, chemiluminescent, or chromogenic agents; substrates; cofactors; inhibitors; magnetic particles; and other moieties known in the art.
  • RNA equivalent in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
  • sample is used in its broadest sense.
  • a sample suspected of containing INTSIG, nucleic acids encoding INTSIG, or fragments thereof may comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; a cell; genomic DNA, RNA, or cDNA, in solution or bound to a substrate; a tissue; a tissue print; etc.
  • specific binding and “specifically binding” refer to that interaction between a protein or peptide and an agonist, an antibody, an antagonist, a small molecule, or any natural or synthetic binding composition.
  • the interaction is dependent upon the presence of a particular structure of the protein, e.g., the antigenic determinant or epitope, recognized by the binding molecule.
  • a particular structure of the protein e.g., the antigenic determinant or epitope
  • the binding molecule e.g., the binding molecule for binding the binding molecule.
  • an antibody is specific for epitope "A”
  • the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody.
  • substantially purified refers to nucleic acid or amino acid sequences that are removed from their natural environment and are isolated or separated, and are at least 60% free, preferably at least 75% free, and most preferably at least 90% free from other components with which they are naturally associated.
  • substitution refers to the replacement of one or more amino acid residues or nucleotides by different amino acid residues or nucleotides, respectively.
  • Substrate refers to any suitable rigid or semi-rigid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillaries.
  • the substrate can have a variety of surface forms, such as wells, trenches, pins, channels and pores, to which polynucleotides or polypeptides are bound.
  • a “transcript image” refers to the collective pattern of gene expression by a particular cell type or tissue under given conditions at a given time.
  • Transformation describes a process by which exogenous DNA is introduced into a recipient cell. Transformation may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method for transformation is selected based on the type of host cell being transformed and may include, but is not limited to, bacteriophage or viral infection, electroporation, heat shock, lipofection, and particle bombardment.
  • transformed cells includes stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome, as well as transiently transformed cells which express the inserted DNA or RNA for limited periods of time.
  • a "transgenic organism,” as used herein, is any organism, including but not limited to animals and plants, in which one or more of the cells of the organism contains heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art.
  • the nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus.
  • the term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule.
  • the transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals.
  • the isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation. Techniques for transferring the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook et al. (1989), supra.
  • a "variant" of a particular nucleic acid sequence is defined as a nucleic acid sequence having at least 40% sequence identity to the particular nucleic acid sequence over a certain length of one of the nucleic acid sequences using blastn with the "BLAST 2 Sequences" tool Version 2.0.9 (May-07- 1999) set at default parameters.
  • Such a pair of nucleic acids may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at ' least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length.
  • a variant may be described as, for example, an "allelic” (as defined above), “splice,” “species,” or “polymorphic” variant.
  • a splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternate splicing of exons during mRNA processing.
  • the corresponding polypeptide may possess additional functional domains or lack domains that are present in the reference molecule.
  • Species variants are polynucleotide sequences that vary from one species to another. The resulting polypeptides will generally have significant amino acid identity relative to each other.
  • a polymorphic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species.
  • Polymorphic variants also may encompass "single nucleotide polymorphisms" (SNPs) in which the polynucleotide sequence varies by one nucleotide base.
  • SNPs single nucleotide polymorphisms
  • the presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state.
  • a "variant" of a particular polypeptide sequence is defined as a polypeptide sequence having at least 40% sequence identity to the particular polypeptide sequence over a certain length of one of the polypeptide sequences using blastp with the "BLAST 2 Sequences" tool Version 2.0.9 (May-07- 1999) set at default parameters.
  • Such a pair of polypeptides may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length of one of the polypeptides.
  • the invention is based on the discovery of new human intracellular signaling molecules (INTSIG), the polynucleotides encoding INTSIG, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders.
  • INTSIG new human intracellular signaling molecules
  • Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the invention. Each polynucleotide and its corresponding polypeptide are correlated to a single Incyte project identification number (Incyte Project DD). Each polypeptide sequence is denoted by both a polypeptide sequence identification number (Polypeptide SEQ DD NO:) and an Incyte polypeptide sequence number (Incyte Polypeptide DD) as shown.
  • Each polynucleotide sequence is denoted by both a polynucleotide sequence identification number (Polynucleotide SEQ DD NO:) and an Incyte polynucleotide consensus sequence number (Incyte Polynucleotide DD) as shown.
  • Table 2 shows sequences with homology to the polypeptides of the invention as identified by BLAST analysis against the GenBank protein (genpept) database. Columns 1 and 2 show the polypeptide sequence identification number (Polypeptide SEQ DD NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide DD) for polypeptides of the invention.
  • Column 3 shows the GenBank identification number (Genbank DD NO:) of the nearest GenBank homolog.
  • Column 4 shows the probability score for the match between each polypeptide and its GenBank homolog.
  • Column 5 shows the annotation of the GenBank homolog along with relevant citations where applicable, all of which are expressly incorporated by reference herein.
  • Table 3 shows various structural features of the polypeptides of the invention.
  • Columns 1 and 2 show the polypeptide sequence identification number (SEQ DD NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide DD) for each polypeptide of the invention.
  • Column 3 shows the number of amino acid residues in each polypeptide.
  • Column 4 shows potential phosphorylation sites, and column 5 shows potential glycosylation sites, as determined by the MOTIF'S program of the GCG sequence analysis software package (Genetics Computer Group, Madison WI).
  • Column 6 shows amino acid residues comprising signature sequences, domains, and motifs.
  • Column 7 shows analytical methods for protein structure/function analysis and in some cases, searchable databases to which the analytical methods were applied.
  • SEQ DD NO:2 is 37% identical to Schizosaccharomyces pombe beta transducin (GenBank DD g3451308) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 2.1e-146, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:2 also contains a G-beta repeat WD40 domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from MOTIFS analysis provides further corroborative evidence that SEQ DD NO:2 is a transducin.
  • HMM hidden Markov model
  • SEQ DD NO:6 is 85% identical to murine nedd-1 protein (GenBank DD g286103) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 0.0, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:6 also contains a WD domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS and MOTTFS analyses provide further corroborative evidence that SEQ DD NO: 6 is a protein involved in signal transduction.
  • HMM hidden Markov model
  • SEQ ID NO: 10 is 51% identical to the human rho GTPase activating protein pi 15 (GenBank DD g840786) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 5.2e-211, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance.
  • SEQ DD NO: 10 contains a rhoGAP domain, an SH3 domain, and a Fes/CIP4 actin cytoskeleton regulatory protein domain, as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains.
  • HMM hidden Markov model
  • SEQ DD NO: 10 is a GTPase activating protein.
  • SEQ DD NO: 16 is 49% identical to the human ras-related tumor suppressor NOEY2 (GenBank DD g4100355) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 3.6e-45, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance.
  • SEQ DD NO: 16 also contains a ras family domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS and MOTIFS analyses provide further corroborative evidence that SEQ DD NO: 16 is a signaling protein of the ras family.
  • HMM hidden Markov model
  • SEQ DD NO:20 is 95% identical to murine SLM-1 protein (GenBank DD g4426613) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 3.1e-183, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:20 also contains a KH domain (E- value is 0.11) as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains.
  • HMM hidden Markov model
  • SEQ DD NO:l SEQ DD NO:3-5, SEQ DD NO:7-9, SEQ DD NO: 11-13, SEQ DD NO: 14-15, and SEQ DD NO:17-19 were analyzed and annotated in a similar manner.
  • the algorithms and parameters for the analysis of SEQ DD NO:1-20 are described in Table 7.
  • the full length polynucleotide sequences of the present invention were assembled using cDNA sequences or coding (exon) sequences derived from genomic DNA, or any combination of these two types of sequences.
  • Columns 1 and 2 list the polynucleotide sequence identification number (Polynucleotide SEQ DD NO:) and the corresponding Incyte polynucleotide consensus sequence number (Incyte Polynucleotide DD) for each polynucleotide of the invention.
  • Column 3 shows the length of each polynucleotide sequence in basepairs.
  • Column 4 lists fragments of the polynucleotide sequences which are useful, for example, in hybridization or amplification technologies that identify SEQ DD NO:21-40 or that distinguish between SEQ DD NO:21-40 and related polynucleotide sequences.
  • Column 5 shows identification numbers corresponding to cDNA sequences, coding sequences (exons) predicted from genomic DNA, and/or sequence assemblages comprised of both cDNA and genomic DNA. These sequences were used to assemble the full length polynucleotide sequences of the invention.
  • Columns 6 and 7 of Table 4 show the nucleotide start (5') and stop (3') positions of the cDNA and/or genomic sequences in column 5 relative to their respective full length sequences.
  • the identification numbers in Column 5 of Table 4 may refer specifically, for example, to Incyte cDNAs along with their corresponding cDNA libraries.
  • 105283R6 is the identification number of an Incyte cDNA sequence
  • BMARNOT02 is the cDNA library from which it is derived.
  • Incyte cDNAs for which cDNA libraries are not indicated were derived from pooled cDNA libraries (e.g., 71206562V1).
  • the identification numbers in column 5 may refer to GenBank cDNAs or ESTs (e.g., g3034305) which contributed to the assembly of the full length polynucleotide sequences.
  • the identification numbers in column 5 may identify sequences derived from the ENSEMBL (The Sanger Centre, Cambridge, UK) database (i.e., those sequences including the designation "ENST”).
  • the identification numbers in column 5 may be derived from the NCBI RefSeq Nucleotide Sequence Records Database (i.e., those sequences including the designation "NM” or “NT”) or the NCBI RefSeq Protein Sequence Records (i.e., those sequences including the designation "NP”).
  • the identification numbers in column 5 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an "exon stitching" algorithm. For example, ⁇ L_XXXXXXJ!
  • XXXXX is the identification number of the cluster of sequences to which the algorithm was applied
  • YYYYY is the number of the prediction generated by the algorithm
  • N 1A3 _ Struktur if present, represent specific exons that may have been manually edited during analysis (See Example V).
  • the identification numbers in column 5 may refer to assemblages of exons brought together by an "exon-stretching" algorithm. For example,
  • FLXXXXXXX_gAAAAA_gBBBBB_l_N is the identification number of a "stretched" sequence, with XXXXX being the Incyte project identification number, gAAAAA being the GenBank identification number of the human genomic sequence to which the "exon-stretching" algorithm was applied, gBBBBB being the GenBank identification number or NCBI RefSeq identification number of the nearest GenBank protein homolog, and N referring to specific exons (See Example V).
  • a RefSeq identifier (denoted by " ⁇ M,” “ ⁇ P,” or “NT”) may be used in place of the GenBank identifier (i.e., gBBBBB).
  • a prefix identifies component sequences that were hand-edited, predicted from genomic DNA sequences, or derived from a combination of sequence analysis methods.
  • the following Table lists examples of component sequence prefixes and corresponding sequence analysis methods associated with the prefixes (see Example IV and Example V).
  • Incyte cDNA coverage redundant with the sequence coverage shown in column 5 was obtained to confirm the final consensus polynucleotide sequence, but the relevant Incyte cDNA identification numbers are not shown.
  • Table 5 shows the representative cDNA libraries for those full length polynucleotide sequences which were assembled using Incyte cDNA sequences.
  • the representative cDNA library is the Incyte cDNA library which is most frequently represented by the Incyte cDNA sequences which were used to assemble and confirm the above polynucleotide sequences.
  • the tissues and vectors which were used to construct the cDNA libraries shown in Table 5 are described in Table 6.
  • the invention also encompasses INTSIG variants.
  • a preferred INTSIG variant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% amino acid sequence identity to the INTSIG amino acid sequence, and which contains at least one functional or structural characteristic of INTSIG.
  • the invention also encompasses polynucleotides which encode INTSIG.
  • the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ DD NO:21-40, which encodes INTSIG.
  • the polynucleotide sequences of SEQ DD NO:21-40 as presented in the Sequence Listing, embrace the equivalent RNA sequences, wherein occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
  • the invention also encompasses a variant of a polynucleotide sequence encoding INTSIG.
  • a variant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding INTSIG.
  • a particular aspect of the invention encompasses a variant of a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ DD NO:21- 40 which has at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to a nucleic acid sequence selected from the group consisting of SEQ DD NO:21-40. Any one of the polynucleotide variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of INTSIG.
  • nucleotide sequences which encode INTSIG and its variants are generally capable of hybridizing to the nucleotide sequence of the naturally occurring INTSIG under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding INTSIG or its derivatives possessing a substantially different codon usage, e.g., inclusion of non- naturally occurring codons. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host.
  • RNA transcripts having more desirable properties such as a greater half-life, than transcripts produced from the naturally occurring sequence.
  • the invention also encompasses production of DNA sequences which encode INTSIG and INTSIG derivatives, or fragments thereof, entirely by synthetic chemistry.
  • the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents well known in the art.
  • synthetic chemistry may be used to introduce mutations into a sequence encoding INTSIG or any fragment thereof.
  • polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ DD NO:21-40 and fragments thereof under various conditions of stringency.
  • Hybridization conditions including annealing and wash conditions, are described in "Definitions.”
  • Methods for DNA sequencing are well known in the art and may be used to practice any of the embodiments of the invention.
  • the methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical, Cleveland OH), Taq polymerase (Applied Biosystems), thermostable T7 polymerase (Amersham Pharmacia Biotech, Piscataway NJ), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE amplification system (Life Technologies, Gaithersburg MD).
  • sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno NV), PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (Applied Biosystems). Sequencing is then carried out using either the ABI 373 or 377 DNA sequencing system (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (Molecular Dynamics, Sunnyvale CA), or other systems known in the art. The resulting sequences are analyzed using a variety of algorithms which are well known in the art. (See, e.g., Ausubel, F.M. (1997) Short Protocols in Molecular Biology. John Wiley & Sons, New York NY, unit 7.7; Meyers, R.A. (1995) Molecular Biology and Biotechnology. Wiley VCH, New York NY, pp. 856-853.)
  • the nucleic acid sequences encoding INTSIG may be extended utilizing a partial nucleotide sequence and employing various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements.
  • various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements.
  • restriction-site PCR uses universal and nested primers to amplify unknown sequence from genomic DNA within a cloning vector. (See, e.g., Sarkar, G. (1993) PCR Methods Applic. 2:318-322.)
  • inverse PCR uses primers that extend in divergent directions to amplify unknown sequence from a circularized template.
  • the template is derived from restriction fragments comprising a known genomic locus and surrounding sequences.
  • a third method, capture PCR involves PCR amplification of DNA fragments adjacent to known sequences in human and yeast artificial chromosome DNA. (See, e.g., Lagerstrom, M. et al. (1991) PCR Methods Applic.
  • primers may be designed using commercially available software, such as OLIGO 4.06 primer analysis software (National Biosciences, Plymouth MN) or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the template at temperatures of about 68°C to 72°C
  • Genomic libraries may be useful for extension of sequence into 5' non-transcribed regulatory regions.
  • Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products.
  • capillary sequencing may employ flowable polymers for electrophoretic separation, four different nucleotide- specific, laser-stimulated fluorescent dyes, and a charge coupled device camera for detection of the emitted wavelengths.
  • Output/light intensity may be converted to electrical signal using appropriate ' software (e.g., GENOTYPER and SEQUENCE NAVIGATOR, Applied Biosystems), and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled.
  • Capillary electrophoresis is especially preferable for sequencing small DNA fragments which may be present in limited amounts in a particular sample.
  • polynucleotide sequences or fragments thereof which encode INTSIG may be cloned in recombinant DNA molecules that direct expression of INTSIG, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced and used to express INTSIG.
  • nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter INTSIG-encoding 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 and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences.
  • oligonucleotide- mediated site-directed mutagenesis may be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.
  • the nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA; described in U.S. Patent No.
  • DNA shuffling is a process by which a library of gene variants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene variants with the desired properties.
  • sequences encoding INTSIG may be synthesized, in whole or in part, using chemical methods well known in the art.
  • INTSIG itself or a fragment thereof may be synthesized using chemical methods.
  • peptide synthesis can be performed using various solution-phase or solid-phase techniques.
  • the peptide may be substantially purified by preparative high performance liquid chromatography. (See, e.g., Chiez, R.M. and F.Z. Regnier (1990) Methods Enzymol. 182:392-421.)
  • the composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing. (See, e.g., Creighton, supra, pp. 28-53.)
  • an appropriate expression vector i.e., a vector which contains the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host.
  • These elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5' and 3' untranslated regions in the vector and in polynucleotide sequences encoding INTSIG. Such elements may vary in their strength and specificity. Specific initiation signals may also be used to achieve more efficient translation of sequences encoding INTSIG. Such signals include the ATG initiation codon and adjacent sequences, e.g. the Kozak sequence. In cases where sequences encoding INTSIG and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed.
  • exogenous translational control signals including an in-frame ATG initiation codon should be provided by the vector.
  • Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular host cell system used. (See, e.g., Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162.)
  • a variety of expression vector/host systems may be utilized to contain and express sequences encoding INTSIG. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e.g., baculovirus); plant cell systems transformed with viral expression vectors (e.g., cauliflower mosaic virus, CaMV, or tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors insect cell systems infected with viral expression vectors (e.g., baculovirus)
  • plant cell systems transformed with viral expression vectors e.g., cauliflower mosaic virus, CaMV, or tobacco
  • Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population.
  • the invention is not limited by the host cell employed.
  • cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding INTSIG.
  • routine cloning, subcloning, and propagation of polynucleotide sequences encoding INTSIG can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding INTSIG into the vector's multiple cloning site disrupts the lacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules.
  • vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence.
  • vectors which direct high level expression of INTSIG may be used.
  • vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used.
  • Yeast expression systems may be used for production of INTSIG.
  • a number of vectors containing constitutive or inducible promoters may be used in the yeast Saccharomyces cerevisiae or Pichia pastoris.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH promoters
  • such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation.
  • Plant systems may also be used for expression of INTSIG. Transcription of sequences encoding INTSIG may be driven by viral promoters, e.g., the 35S and 19S promoters of CaMV used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 3:17-311). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e.g., Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl.
  • a number of viral-based expression systems may be utilized.
  • sequences encoding INTSIG may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain infective virus which expresses INTSIG in host cells.
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
  • SV40 or EBV- based vectors may also be used for high-level protein expression.
  • HACs Human artificial chromosomes
  • HACs may also be employed to deliver larger fragments of DNA than can be contained in and expressed from a plasmid.
  • HACs of about 6 kb to 10 Mb are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes.
  • liposomes, polycationic amino polymers, or vesicles for therapeutic purposes.
  • INTSIG in cell lines is preferred.
  • sequences encoding INTSIG can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector.
  • expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector.
  • cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media.
  • the purpose of the selectable marker is to confer resistance to a ⁇ selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences.
  • Resistant clones of stably transformed cells may be propagated using tissue culture techniques appropriate to the cell type.
  • selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase and adenine phosphoribosyltransferase genes, for use in fk and apr cells, respectively. (See, e.g., Wigler, M. et al. (1977) Cell 11:223-232; Lowy, I. et al. (1980) Cell 22:817-823.) Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection.
  • dhfr confers resistance to methotrexate
  • neo confers resistance to the aminoglycosides neomycin and G-418
  • als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively.
  • Additional selectable genes have been described, e.g., trpB and hisD, which alter cellular requirements for metabolites.
  • Visible markers e.g., anthocyanins, green fluorescent proteins (GFP; Clontech), ⁇ glucuronidase and its substrate ⁇ -glucuronide, or luciferase and its substrate luciferin may be used. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system. (See, e.g., Rhodes, CA. (1995) Methods Mol. Biol. 55:121-131.)
  • marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed.
  • sequence encoding INTSIG is inserted within a marker gene sequence
  • transformed cells containing sequences encoding INTSIG can be identified by the absence of marker gene function.
  • a marker gene can be placed in tandem with a sequence encoding INTSIG under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
  • host cells that contain the nucleic acid sequence encoding INTSIG and that express INTSIG may 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 protein sequences. Immunological methods for detecting and measuring the expression of INTSIG 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
  • a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on INTSIG is preferred, but a competitive binding assay may be employed.
  • assays are well known in the art. (See, e.g., Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St. Paul MN, Sect. IV; Coligan, J.E. et al. (1997) Current Protocols in Immunology, Greene Pub. Associates and Wiley-Interscience, New York NY; and Pound, J.D. (1998) Immunochemical Protocols, Humana Press, Totowa NJ.)
  • Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding INTSIG include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.
  • sequences encoding INTSIG, or any fragments thereof may be cloned into a vector for the production of an mRNA probe.
  • a vector for the production of an mRNA probe Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Amersham Pharmacia Biotech, Promega (Madison WI), and US Biochemical. Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
  • Host cells transformed with nucleotide sequences encoding INTSIG may be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
  • the protein produced by a transformed cell may be secreted or retained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing polynucleotides which encode INTSIG may be designed to contain signal sequences which direct secretion of INTSIG through a prokaryotic or eukaryotic cell membrane.
  • a host cell strain may 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 polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
  • Post-translational processing which cleaves a "prepro” or “pro” form of the protein may 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 WI38) are available from the American Type Culture Collection (ATCC, Manassas VA) and may be chosen to ensure the correct modification and processing of the foreign protein.
  • ATCC American Type Culture Collection
  • natural, modified, or recombinant nucleic acid sequences encoding INTSIG may be ligated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems.
  • a chimeric INTSIG protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide libraries for inhibitors of INTSIG activity.
  • Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matrices.
  • Such moieties include, but are not limited to, glutathione S-transf erase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutinin (HA).
  • GST, MBP, Trx, CBP, and 6-His enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively.
  • FLAG, c-myc, and hemagglutinin (HA) enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags.
  • a fusion protein may also be engineered to contain a proteolytic cleavage site located between the INTSIG encoding sequence and the heterologous protein sequence, so that INTSIG may be cleaved away from the heterologous moiety following purification. Methods for fusion protein expression and purification are discussed in Ausubel (1995, supra, ch. 10). A variety of commercially available kits may also be used to facilitate expression and purification of fusion proteins.
  • synthesis of radiolabeled INTSIG may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcription and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled amino acid precursor, for example, 35 S-methionine.
  • INTSIG of the present invention or fragments thereof may be used to screen for compounds that specifically bind to INTSIG.
  • At least one and up to a plurality of test compounds may be screened for specific binding to INTSIG.
  • test compounds include antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules.
  • the compound thus identified is closely related to the natural ligand of INTSIG, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a ⁇ natural binding partner.
  • the compound can be closely related to the natural receptor to which INTSIG binds, or to at least a fragment of the receptor, e.g., the ligand binding site. In either case, the compound can be rationally designed using known techniques. In one embodiment, screening for these compounds involves producing appropriate cells which express INTSIG, either as a secreted protein or on the cell membrane.
  • Preferred cells include cells from mammals, yeast, Drosophila, or K coli. Cells expressing INTSIG or cell membrane fractions which contain INTSIG are then contacted with a test compound and binding, stimulation, or inhibition of activity of either INTSIG or the compound is analyzed.
  • An assay may simply test binding of a test compound to the polypeptide, wherein binding is detected by a fluorophore, radioisotope, enzyme conjugate, or other detectable label.
  • the assay may comprise the steps of combining at least one test compound with INTSIG, either in solution or affixed to a solid support, and detecting the binding of INTSIG to the compound.
  • the assay may detect or measure binding of a test compound in the presence of a labeled competitor.
  • the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support.
  • INTSIG of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of INTSIG.
  • Such compounds may include agonists, antagonists, or partial or inverse agonists.
  • an assay is performed under conditions permissive for INTSIG activity, wherein INTSIG is combined with at least one test compound, and the activity of INTSIG in the presence of a test compound is compared with the activity of INTSIG in the absence of the test compound. A change in the activity of INTSIG in the presence of the test compound is indicative of a compound that modulates the activity of INTSIG.
  • a test compound is combined with an in vitro or cell-free system comprising INTSIG under conditions suitable for INTSIG activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of INTSIG may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened.
  • polynucleotides encoding INTSIG or their mammalian homologs may be "knocked out" in an animal model system using homologous recombination in embryonic stem (ES) cells.
  • ES embryonic stem
  • Such techniques are well known in the art and are useful for the generation of animal models of human disease.
  • mouse ES cells such as the mouse 129/SvJ cell line, are derived from the early mouse embryo and grown in culture.
  • the ES cells are transformed with a vector containing the gene of interest disrupted by a marker gene, e.g., the neomycin phosphotransferase gene (neo; Capecchi, M.R. (1989) Science 244:1288-1292).
  • a marker gene e.g., the neomycin phosphotransferase gene (neo; Capecchi, M.R. (1989) Science 244:1288-1292).
  • the vector integrates into the corresponding region of the host genome by homologous recombination.
  • homologous recombination takes place using the Cre-loxP system to knockout a gene of interest in a tissue- or developmental stage-specific manner (Marth, J.D. (1996) Clin. Invest. 97:1999-2002; Wagner, K.U. et al. (1997) Nucleic Acids Res. 25:4323-4330).
  • Transformed ES cells are identified and microinjected into mouse cell blastocysts such as those from the C57BL/6 mouse strain.
  • the blastocysts are surgically transferred to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains.
  • Transgenic animals thus generated may be tested with potential therapeutic or toxic agents.
  • Polynucleotides encoding INTSIG may also be manipulated in vitro in ES cells derived from human blastocysts.
  • Human ES cells have the potential to differentiate into at least eight separate cell lineages including endoderm, mesoderm, and ectodermal cell types. These cell lineages differentiate into, for example, neural cells, hematopoietic lineages, and cardiomyocytes (Thomson, J.A. et al. (1998) Science 282:1145-1147).
  • Polynucleotides encoding INTSIG can also be used to create "knockin” humanized animals (pigs) or transgenic animals (mice or rats) to model human disease.
  • knockin technology a region of a polynucleotide encoding INTSIG is injected into animal ES cells, and the injected sequence integrates into the animal cell genome.
  • Transformed cells are injected into blastulae, and the blastulae are implanted as described above.
  • Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease.
  • a mammal inbred to overexpress INTSIG e.g., by secreting INTSIG in its milk, may also serve as a convenient source of that protein (Janne, J. et al. (1998) Biotechnol. Annu. Rev. 4:55-74).
  • INTSIG Intracellular signaling molecules
  • the expression of INTSIG is closely associated with brain and neurological tissues including thoracic dorsal root ganglion tissue, dermal tissue, reproductive tissue, digestive and hemic/immune tissue, diseased prostate tissue, and tumorous tissues including bladder, tongue, and testicular. Therefore, INTSIG appears to play a role in cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders. In the treatment of disorders associated with increased INTSIG expression or activity, it is desirable to decrease the expression or activity of INTSIG. In the treatment of disorders associated with decreased INTSIG expression or activity, it is desirable to increase the expression or activity of INTSIG.
  • INTSIG or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG.
  • disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung
  • Parkinson's disease and other extrapyramidal disorders amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and
  • Gerstmann-Straussler-Scheinker syndrome fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear pal
  • compositions comprising a substantially purified INTSIG in conjunction with a suitable pharmaceutical carrier may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG including, but not limited to, those provided above.
  • an agonist which modulates the activity of INTSIG may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG including, but not limited to, those listed above.
  • an antagonist of INTSIG may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of INTSIG. Examples of such disorders include, but are not limited to, those cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders described above.
  • an antibody which specifically binds INTSIG may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express INTSIG.
  • a vector expressing the complement of the polynucleotide encoding INTSIG may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of INTSIG including, but not limited to, those described above.
  • any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • An antagonist of INTSIG may be produced using methods which are generally known in the art.
  • purified INTSIG may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind INTSIG.
  • Antibodies to INTSIG may also be generated using methods that are well known in the art. Such antibodies may 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 (i.e., those which inhibit dimer formation) are generally preferred for therapeutic use.
  • various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with INTSIG or with any fragment or oligopeptide thereof which has immunogenic properties.
  • various adjuvants may be used to increase immunological response.
  • adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are especially preferable.
  • the oligopeptides, peptides, or fragments used to induce antibodies to INTSIG have an amino acid sequence consisting of at least about 5 amino acids, and generally will consist of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein. Short stretches of INTSIG amino acids may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced.
  • Monoclonal antibodies to INTSIG may 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 technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D. et al. (1985) J. Immunol. Methods 81:31-42; Cote, R.J. et al. (1983) Proc. Natl. Acad. Sci. USA 80:2026-2030; and Cole, S.P. et al. (1984) Mol. Cell Biol. 62:109-120.)
  • chimeric antibodies such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity.
  • techniques developed for the production of "chimeric antibodies” such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used.
  • techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce INTSIG-specific single chain antibodies.
  • Antibodies with related specificity, but of distinct idiotypic composition may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (See, e.g., Burton, D.R. (1991) Proc. Natl. Acad. Sci. USA 88:10134-10137.)
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (See, e.g., Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Winter, G. et al. (1991) Nature 349:293-299.)
  • Antibody fragments which contain specific binding sites for INTSIG may also be generated.
  • fragments include, but are not limited to, F(ab') 2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e.g., Huse, W.D. et al. (1989) Science 246:1275-1281.)
  • immunoassays may be used for screening 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.
  • Such immunoassays typically involve the measurement of complex formation between INTSIG and its specific antibody.
  • a two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering INTSIG epitopes is generally used, but a competitive binding assay may also be employed (Pound, supra).
  • K a is defined as the molar concentration of INTSIG-antibody complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions.
  • the K determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple INTSIG epitopes, represents the average affinity, or avidity, of the antibodies for INTSIG.
  • the K a determined for a preparation of monoclonal antibodies, which are monospecific for a particular INTSIG epitope represents a true measure of affinity.
  • High-affinity antibody preparations with K ranging from about 10 9 to 10 12 L/mole are preferred for use in immunoassays in which the INTSIG- antibody complex must withstand rigorous manipulations.
  • Low-affinity antibody preparations with K a ranging from about 10 6 to 10 7 L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of INTSIG, preferably in active form, from the antibody (Catty, D. (1988) Antibodies, Volume I: A Practical Approach. IRL Press, Washington DC; Liddell, J.E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies, John Wiley & Sons, New York NY).
  • polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications.
  • a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml, preferably 5-10 mg specific antibody/ml is generally employed in procedures requiring precipitation of INTSIG-antibody complexes.
  • Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available. (See, e.g., Catty, supra, and Coligan et al. supra.)
  • the polynucleotides encoding INTSIG may be used for therapeutic purposes.
  • modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified oligonucleotides) to the coding or regulatory regions of the gene encoding INTSIG.
  • complementary sequences or antisense molecules DNA, RNA, PNA, or modified oligonucleotides
  • antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding INTSIG. (See, e.g., Agrawal, S., ed. (1996) Antisense Therapeutics, Humana Press Inc., Totawa NJ.)
  • Antisense sequences can be delivered intracellularly in the form of an expression plasmid which, upon transcription, produces a sequence complementary to at least a portion of the cellular sequence encoding the target protein.
  • Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors.
  • polynucleotides encoding INTSIG may be used for somatic or germline gene therapy.
  • Gene therapy may be performed to (i) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCDD)-Xl disease characterized by X- linked inheritance (Cavazzana-Calvo, M. et al. (2000) Science 288:669-672), severe combined immunodeficiency syndrome associated with an inherited adenosine deaminase (ADA) deficiency (Blaese, R.M. et al. (1995) Science 270:475-480; Bordignon, C. et al. (1995) Science 270:470-475), cystic fibrosis (Zabner, J. et al. (1993) Cell 75:207-216; Crystal, R.G. et al. (1995) Hum. Gene
  • hepatitis B or C virus HBV, HCV
  • fungal parasites such as Candida albicans and Paracoccidioides brasiliensis
  • protozoan parasites such as Plasmodium falciparum and Trvpanosoma cruzi.
  • the expression of INTSIG from an appropriate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency.
  • diseases or disorders caused by deficiencies in INTSIG are treated by constructing mammalian expression vectors encoding INTSIG and introducing these vectors by mechanical means into INTSIG-deficient cells.
  • Mechanical transfer technologies for use with cells in vivo or ex vitro include (i) direct DNA microinjection into individual cells, (ii) ballistic gold particle delivery, (iii) liposome-mediated transfection, (iv) receptor-mediated gene transfer, and (v) the use of DNA transposons (Morgan, R.A. and W.F. Anderson (1993) Annu. Rev. Biochem. 62:191-217; Ivies, Z. (1997) Cell 91:501-510; Boulay, J-L. and H. Recipon (1998) Curr. Opin. Biotechnol. 9:445-450).
  • Expression vectors that may be effective for the expression of INTSIG include, but are not limited to, the PCDNA 3.1, EPITAG, PRCCMV2, PREP, PVAX, PCR2-TOPOTA vectors (Invitrogen, Carlsbad CA), PCMV-SCRIPT, PCMV-TAG, PEGSH/PERV (Stratagene, La Jolla CA), and PTET-OFF, PTET-ON, PTRE2, PTRE2-LUC, PTK-HYG (Clontech, Palo Alto CA).
  • INTSIG may be expressed using (i) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or ⁇ -actin genes), (ii) an inducible promoter (e.g., the tetracycline-regulated promoter (Gossen, M. and H. Bujard (1992) Proc. Natl. Acad. Sci. USA 89:5547-5551; Gossen, M. et al. (1995) Science 268:1766-1769; Rossi, F.M.V. and H.M. Blau (1998) Curr. Opin. Biotechnol.
  • a constitutively active promoter e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or ⁇ -actin genes
  • TRANSFECTION KIT available from Invitrogen
  • transformation is performed using the calcium phosphate method (Graham, F.L. and A.J. Eb (1973) Virology 52:456-467), or by electroporation (Neumann, E. et al. (1982) EMBO J. 1:841-845).
  • the introduction of DNA to primary cells requires modification of these standardized mammalian transfection protocols.
  • diseases or disorders caused by genetic defects with respect to INTSIG expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding INTSIG under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (ii) appropriate RNA packaging signals, and (iii) a Rev-responsive element (RRE) along with additional retrovirus cz ' s-acting RNA sequences and coding sequences required for efficient vector propagation.
  • Retrovirus vectors e.g., PFB and PFBNEO
  • the vector is propagated in an appropriate vector producing cell line (VPCL) that expresses an envelope gene with a tropism for receptors on the target cells or a promiscuous envelope protein such as VSVg (Armentano, D. et al. (1987) J. Virol. 61:1647-1650; Bender, M.A. et al. (1987) J. Virol. 61:1639-1646; Adam, M.A. and A.D. Miller (1988) J. Virol. 62:3802-3806; Dull, T. et al. (1998) J. Virol. 72:8463-8471; Zufferey, R. et al.
  • VSVg vector producing cell line
  • U.S. Patent No. 5,910,434 to Rigg discloses a method for obtaining retrovirus packaging cell lines, and is hereby incorporated by reference. Propagation of retrovirus vectors, transduction of a population of cells (e.g., CD4 + T-cells), and the return of transduced cells to a patient are procedures well known to persons skilled in the art of gene therapy and have been well documented (Ranga, U. et al. (1997) J. Virol. 71:7020-7029; Bauer, G. et al.
  • an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding INTSIG to cells which have one or more genetic abnormalities with respect to the expression of INTSIG.
  • the construction and packaging of adenovirus-based vectors are well known to those with ordinary skill in the art. Replication defective adenovirus vectors have proven to be versatile for importing genes encoding immunoregulatory proteins into intact islets in the pancreas (Csete, M.E. et al. (1995) Transplantation 27:263-268). Potentially useful adenoviral vectors are described in U.S. Patent No. 5,707,618 to Armentano ("Adenovirus vectors for gene therapy”), hereby incorporated by reference.
  • a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding INTSIG to target cells which have one or more genetic abnormalities with respect to the expression of INTSIG.
  • the use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing INTSIG to cells of the central nervous system, for which HSV has a tropism.
  • the construction and packaging of herpes-based vectors are well known to those with ordinary skill in the art.
  • a replication-competent herpes simplex virus (HSV) type 1 -based vector has been used to deliver a reporter gene to the eyes of primates (Liu, X. et al. (1999) Exp. Eye Res. 169:385-395).
  • HSV-1 virus vector has also been disclosed in detail in U.S. Patent No. 5,804,413 to DeLuca ("Herpes simplex virus strains for gene transfer"), which is hereby incorporated by reference.
  • U.S. Patent No. 5,804,413 teaches the use of recombinant HSV d92 which consists of a genome containing at least one exogenous gene to be transferred to a cell under the control of the appropriate promoter for purposes including human gene therapy. Also taught by this patent are the construction and use of recombinant HSV strains deleted for ICP4, ICP27 and ICP22.
  • HSV vectors see also Goins, W.F. et al. (1999) J. Virol.
  • herpesvirus sequences The manipulation of cloned herpesvirus sequences, the generation of recombinant virus following the transfection of multiple plasmids containing different segments of the large herpesvirus genomes, the growth and propagation of herpesvirus, and the infection of cells with herpesvirus are techniques well known to those of ordinary skill in the art.
  • an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding INTSIG to target cells.
  • SFV Semliki Forest Virus
  • This subgenomic RNA replicates to higher levels than the full length genomic RNA, resulting in the overproduction of capsid proteins relative to the viral proteins with enzymatic activity (e.g., protease and polymerase).
  • enzymatic activity e.g., protease and polymerase.
  • inserting the coding sequence for INTSIG into the alphavirus genome in place of the capsid-coding region results in the production of a large number of INTSIG-coding RNAs and the synthesis of high levels of INTSIG in vector transduced cells.
  • alphavirus infection is typically associated with cell lysis within a few days
  • the ability to establish a persistent infection in hamster normal kidney cells (BHK-21) with a variant of Sindbis virus (SIN) indicates that the lytic replication of alphaviruses can be altered to suit the needs of the gene therapy application (Dryga, S.A. et al. (1997) Virology 228:74-83).
  • the wide host range of alphaviruses will allow the introduction of INTSIG into a variety of cell types.
  • the specific transduction of a subset of cells in a population may require the sorting of cells prior to transduction.
  • the methods of manipulating infectious cDNA clones of alphaviruses, performing alphavirus cDNA and RNA transfections, and performing alphavirus infections, are well known to those with ordinary skill in the art.
  • Oligonucleotides derived from the transcription initiation site may also be employed to inhibit gene expression. 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, J.E. et al. (1994) in Huber, B.E. and B.I. Carr, Molecular and Immunologic Approaches, Furura Publishing, Mt. Kisco NY, pp. 163-177.) A complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
  • Ribozymes enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • engineered hammerhead motif ribozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding INTSIG.
  • RNA sequences of between 15 and 20 ribonucleotides, corresponding to the region of the target gene containing the cleavage site, may be evaluated for secondary structural features which may render the oligonucleotide inoperable.
  • the suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
  • RNA molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding INTSIG. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6.
  • these cDNA constructs that synthesize complementary RNA, constitutively or inducibly, can be introduced into cell lines, cells, or tissues.
  • RNA molecules may 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 phosphodiesterase linkages within the backbone of the molecule.
  • An additional embodiment of the invention encompasses a method for screening for a compound which is effective in altering expression of a polynucleotide encoding INTSIG.
  • Compounds which may be effective in altering expression of a specific polynucleotide may include, but are not limited to, oligonucleotides, antisense oligonucleotides, triple helix-forming oligonucleotides, transcription factors and other polypeptide transcriptional regulators, and non-macromolecular chemical entities which are capable of interacting with specific polynucleotide sequences. Effective compounds may alter polynucleotide expression by acting as either inhibitors or promoters of polynucleotide expression.
  • a compound which specifically inhibits expression of the polynucleotide encoding INTSIG may be therapeutically useful, and in the treatment of disorders associated with decreased INTSIG expression or activity, a compound which specifically promotes expression of the polynucleotide encoding INTSIG may be therapeutically useful.
  • test compounds may be screened for effectiveness in altering expression of a specific polynucleotide.
  • a test compound may be obtained by any method commonly known in the art, including chemical modification of a compound known to be effective in altering polynucleotide expression; selection from an existing, commercially-available or proprietary library of naturally-occurring or non-natural chemical compounds; rational design of a compound based on chemical and/or structural properties of the target polynucleotide; and selection from a library of chemical compounds created combinatorially or randomly.
  • a sample comprising a polynucleotide encoding INTSIG is exposed to at least one test compound thus obtained.
  • the sample may comprise, for example, an intact or permeabilized cell, or an in vitro cell-free or reconstituted biochemical system.
  • Alterations in the expression of a polynucleotide encoding INTSIG are assayed by any method commonly known in the art.
  • the expression of a specific nucleotide is detected by hybridization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding INTSIG.
  • the amount of hybridization may be quantified, thus forming the basis for a comparison of the expression of the polynucleotide both with and without exposure to one or more test compounds.
  • a screen for a compound effective in altering expression of a specific polynucleotide can be carried out, for example, using a Schizosaccharomvces pombe gene expression system (Atkins, D. et al. (1999) U.S. Patent No. 5,932,435; Arndt, G.M. et al. (2000) Nucleic Acids Res. 28:E15) or a human cell line such as HeLa cell (Clarke, ML. et al. (2000) Biochem. Biophys. Res. Commun.
  • a particular embodiment of the present invention involves screening a combinatorial library of oligonucleotides (such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides) for antisense activity against a specific polynucleotide sequence (Bruice, T.W. et al. (1997) U.S. Patent No. 5,686,242; Bruice, T.W. et al. (2000) U.S. Patent No. 6,022,691).
  • oligonucleotides such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides
  • vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient. Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See, e.g., Goldman, C.K. et al. (1997) Nat. Biotechnol. 15:462-466.)
  • any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as humans, dogs, cats, cows, horses, rabbits, and monkeys.
  • An additional embodiment of the invention relates to the administration of a composition which generally comprises an active ingredient formulated with a pharmaceutically acceptable excipient.
  • Excipients may include, for example, sugars, starches, celluloses, gums, and proteins.
  • Various formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton PA).
  • Such compositions may consist of INTSIG, antibodies to INTSIG, and mimetics, agonists, antagonists, or inhibitors of INTSIG.
  • compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, pulmonary, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
  • compositions for pulmonary administration may be prepared in liquid or dry powder form. These compositions are generally aerosolized immediately prior to inhalation by the patient.
  • small molecules e.g. traditional low molecular weight organic drugs
  • aerosol delivery of fast- acting formulations is well-known in the art.
  • macromolecules e.g. larger peptides and proteins
  • Pulmonary delivery has the advantage of administration without needle injection, and obviates the need for potentially toxic penetration enhancers.
  • compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose.
  • the determination of an effective dose is well within the capability of those skilled in the art.
  • compositions may be prepared for direct intracellular delivery of macromolecules comprising INTSIG or fragments thereof.
  • liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule.
  • INTSIG or a fragment thereof may be joined to a short cationic N- terminal portion from the HIV Tat-1 protein. Fusion proteins thus generated have been found to transduce into the cells of all tissues, including the brain, in a mouse model system (Schwarze, S.R. et al. (1999) Science 285:1569-1572).
  • the therapeutically effective dose can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models such as mice, rats, rabbits, dogs, monkeys, or pigs. An animal model may also be used to determine the appropriate 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, for example
  • compositions which exhibit large therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies are used to formulate a range of dosage for human use.
  • the dosage contained in such compositions is preferably within a range of circulating concentrations that includes the ED 50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.
  • Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. Long-acting compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation.
  • Normal dosage amounts may vary from about 0.1 ⁇ g to 100,000 ⁇ g, up to a total dose of about 1 gram, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc. DIAGNOSTICS
  • antibodies which specifically bind INTSIG may be used for the diagnosis of disorders characterized by expression of INTSIG, or in assays to monitor patients being treated with INTSIG or agonists, antagonists, or inhibitors of INTSIG.
  • Antibodies useful for diagnostic purposes may be prepared in the same manner as described above for therapeutics. Diagnostic assays for INTSIG include methods which utilize the antibody and a label to detect INTSIG in human body fluids or in extracts of cells or tissues.
  • the antibodies may be used with or without modification, and may be labeled by covalent or non-covalent attachment of a reporter molecule.
  • a wide variety of reporter molecules, several of which are described above, are known in the art and may be used.
  • a variety of protocols for measuring INTSIG including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of INTSIG expression.
  • Normal or standard values for INTSIG expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to INTSIG under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of INTSIG expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
  • the polynucleotides encoding INTSIG may be used for diagnostic purposes.
  • the polynucleotides which may be used include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs.
  • the polynucleotides may be used to detect and quantify gene expression in biopsied tissues in which expression of INTSIG may be correlated with disease.
  • the diagnostic assay may be used to determine absence, presence, and excess expression of INTSIG, and to monitor regulation of INTSIG levels during therapeutic intervention.
  • hybridization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding INTSIG or closely related molecules may be used to identify nucleic acid sequences which encode INTSIG.
  • the specificity of the probe whether it is made from a highly specific region, e.g., the 5' regulatory region, or from a less specific region, e.g., a conserved motif, and the stringency of the hybridization or amplification will determine whether the probe identifies only naturally occurring sequences encoding INTSIG, allelic variants, or related sequences.
  • Probes may also be used for the detection of related sequences, and may have at least 50% sequence identity to any of the INTSIG encoding sequences.
  • the hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ DD NO:21-40 or from genomic sequences including promoters, enhancers, and introns of the INTSIG gene.
  • Means for producing specific hybridization probes for DNAs encoding ESfTSIG include the cloning of polynucleotide sequences encoding INTSIG or INTSIG derivatives into vectors for the production of mRNA probes.
  • vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides.
  • Hybridization probes may be labeled by a variety of reporter groups, for example, by radionuclides such as 32 P or 35 S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
  • Polynucleotide sequences encoding INTSIG may be used for the diagnosis of disorders associated with expression of INTSIG.
  • disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancrea
  • the polynucleotide sequences encoding INTSIG may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and multiforrnat ELISA-like assays; and in microarrays utilizing fluids or tissues from patients to detect altered INTSIG expression. Such qualitative or quantitative methods are well known in the art.
  • the nucleotide sequences encoding INTSIG may be useful in assays that detect the presence of associated disorders, particularly those mentioned above.
  • the nucleotide sequences encoding INTSIG may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes. After a suitable incubation period, the sample is washed and the signal is quantified and compared with a standard value. If the amount of signal in the patient sample is significantly altered in comparison to a control sample then the presence of altered levels of nucleotide sequences encoding INTSIG in the sample indicates the presence of the associated disorder.
  • Such assays may 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 patient.
  • a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, encoding INTSIG, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with values from an experiment in which a known amount of a substantially purified polynucleotide is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder.
  • hybridization assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in the normal subject.
  • the results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.
  • the presence of an abnormal amount of transcript (either under- or overexpressed) in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms.
  • a more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
  • Additional diagnostic uses for oligonucleotides designed from the sequences encoding INTSIG may involve the use of PCR. These oligomers may be chemically synthesized, generated enzymatically, or produced in vitro.
  • Oligomers will preferably contain a fragment of a polynucleotide encoding INTSIG, or a fragment of a polynucleotide complementary to the polynucleotide encoding INTSIG, and will be employed under optimized conditions for identification of a specific gene or condition. Oligomers may also be employed under less stringent conditions for detection or quantification of closely related DNA or RNA sequences.
  • oligonucleotide primers derived from the polynucleotide sequences encoding INTSIG may be used to detect single nucleotide polymorphisms (SNPs).
  • SNPs are substitutions, insertions and deletions that are a frequent cause of inherited or acquired genetic disease in humans.
  • Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods.
  • SSCP single-stranded conformation polymorphism
  • fSSCP fluorescent SSCP
  • oligonucleotide primers derived from the polynucleotide sequences encoding INTSIG are used to amplify DNA using the polymerase chain reaction (PCR).
  • the DNA may be derived, for example, from diseased or normal tissue, biopsy samples, bodily fluids, and the like.
  • SNPs in the DNA cause differences in the secondary and tertiary structures of PCR products in single-stranded form, and these differences are detectable using gel electrophoresis in non-denaturing gels.
  • the oligonucleotide primers are fluorescently labeled, which allows detection of the amplimers in high-throughput equipment such as DNA sequencing machines.
  • sequence database analysis methods termed in silico SNP (isSNP) are capable of identifying polymorphisms by comparing the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence.
  • SNPs may be detected and characterized by mass spectrometry using, for example, the high throughput MASSARRAY system (Sequenom, Inc., San Diego CA).
  • Methods which may also be used to quantify the expression of INTSIG include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves.
  • radiolabeling or biotinylating nucleotides include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves.
  • the speed of quantitation of multiple samples may be accelerated by running the assay in a high-throughput format where the oligomer or polynucleotide of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.
  • oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as elements on a microarray.
  • the microarray can be used in transcript imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as described below.
  • the microarray may also be used to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic basis of a disorder, to diagnose a disorder, to monitor progression/regression of disease as a function of gene expression, and to develop and monitor the activities of therapeutic agents in the treatment of disease.
  • this information may be used to develop a pharmacogenomic profile of a patient in order to select the most appropriate and effective treatment regimen for that patient.
  • therapeutic agents which are highly effective and display the fewest side effects may be selected for a patient based on his/her pharmacogenomic profile.
  • INTSIG, fragments of INTSIG, or antibodies specific for INTSIG may be used as elements on a microarray.
  • the microarray may be used to monitor or measure protein-protein interactions, drug-target interactions, and gene expression profiles, as described above.
  • a particular embodiment relates to the use of the polynucleotides of the present invention to generate a transcript image of a tissue or cell type.
  • a transcript image represents the global pattern of gene expression by a particular tissue or cell type. Global gene expression patterns are analyzed by quantifying the number of expressed genes and their relative abundance under given conditions and at a given time. (See Seilhamer et al., "Comparative Gene Transcript Analysis," U.S. Patent No. 5,840,484, expressly incorporated by reference herein.)
  • a transcript image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcripts or reverse transcripts of a particular tissue or cell type.
  • the hybridization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements comprise a subset of a plurality of elements on a microarray.
  • the resultant transcript image would provide a profile of gene activity.
  • Transcript images may be generated using transcripts isolated from tissues, cell lines, biopsies, or other biological samples. The transcript image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or in vitro, as in the case of a cell line.
  • Transcript images which profile the expression of the polynucleotides of the present invention may also be used in conjunction with in vitro model systems and preclinical evaluation of pharmaceuticals, as well as toxicological testing of industrial and naturally-occurring environmental compounds. All compounds induce characteristic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E.F. et al. (1999) Mol. Carcinog. 24:153-159; Steiner, S. and N.L. Anderson (2000) Toxicol. Lett. 112-113:467-471, expressly incorporated by reference herein). If a test compound has a signature similar to that of a compound with known toxicity, it is likely to share those toxic properties.
  • the toxicity of a test compound is assessed by treating a biological sample containing nucleic acids with the test compound.
  • Nucleic acids that are expressed in the treated biological sample are hybridized with one or more probes specific to the polynucleotides of the present invention, so that transcript levels corresponding to the polynucleotides of the present invention may be quantified.
  • the transcript levels in the treated biological sample are compared with levels in an untreated biological sample. Differences in the transcript levels between the two samples are indicative of a toxic response caused by the test compound in the treated sample.
  • Another particular embodiment relates to the use of the polypeptide sequences of the present invention to analyze the proteome of a tissue or cell type.
  • proteome refers to the global pattern of protein expression in a particular tissue or cell type.
  • proteome expression patterns, or profiles are analyzed by quantifying the number of expressed proteins and their relative abundance under given conditions and at a given time.
  • a profile of a cell's proteome may thus be generated by separating and analyzing the polypeptides of a particular tissue or cell type.
  • the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectric focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Steiner and Anderson, supra).
  • the proteins are visualized in the gel as discrete and uniquely positioned spots, typically by staining the gel with an agent such as Coomassie Blue or silver or fluorescent stains.
  • the optical density of each protein spot is generally proportional to the level of the protein in the sample.
  • the optical densities of equivalently positioned protein spots from different samples, for example, from biological samples either treated or untreated with a test compound or therapeutic agent, are compared to identify any changes in protein spot density related to the treatment.
  • the proteins in the spots are partially sequenced using, for , example, standard methods employing chemical or enzymatic cleavage followed by mass spectrometry.
  • the identity of the protein in a spot may be determined by comparing its partial sequence, preferably of at least 5 contiguous amino acid residues, to the polypeptide sequences of the present invention. In some cases, further sequence data may be obtained for definitive protein identification.
  • a proteomic profile may also be generated using antibodies specific for INTSIG to quantify the levels of INTSIG expression.
  • the antibodies are used as elements on a microarray, and protein expression levels are quantified by exposing the microarray to the sample and detecting the levels of protein bound to each array element (Lueking, A. et al. (1999) Anal. Biochem. 270:103-111; Mendoze, L.G. et al. (1999) Biotechniques 27:778-788). Detection may be performed by a variety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or amino-reactive fluorescent compound and detecting the amount of fluorescence bound at each array element.
  • Toxicant signatures at the proteome level are also useful for toxicological screening, and should be analyzed in parallel with toxicant signatures at the transcript level.
  • There is a poor correlation between transcript and protein abundances for some proteins in some tissues (Anderson, N.L. and J. Seilhamer (1997) Electrophoresis 18:533-537), so proteome toxicant signatures may be useful in the analysis of compounds which do not significantly affect the transcript image, but which alter the proteomic profile.
  • the analysis of transcripts in body fluids is difficult, due to rapid degradation of mRNA, so proteomic profiling may be more reliable and informative in such cases.
  • the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound.
  • Proteins that are expressed in the treated biological sample are separated so that the amount of each protein can be quantified.
  • the amount of each protein is compared to the amount of the corresponding protein in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample.
  • Individual proteins are identified by sequencing the amino acid residues of the individual proteins and comparing these partial sequences to the polypeptides of the present invention.
  • the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins from the biological sample are incubated with antibodies specific to the polypeptides of the present invention. The amount of protein recognized by the antibodies is quantified. The amount of protein in the treated biological sample is compared with the amount in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample.
  • Microarrays may be prepared, used, and analyzed using methods known in the art.
  • methods known in the art See, e.g., Brennan, T.M. et al. (1995) U.S. Patent No. 5,474,796; Schena, M. et al. (1996) Proc. Natl. Acad. Sci. USA 93:10614-10619; Baldeschweiler et al. (1995) PCT application W095/251116; Shalon, D. et al. (1995) PCT application WO95/35505; Heller, R.A. et al. (1997) Proc. Natl. Acad. Sci. USA 94:2150-2155; and Heller, MJ. et al. (1997) U.S.
  • nucleic acid sequences encoding INTSIG may be used to generate hybridization probes useful in mapping the naturally occurring genomic sequence. Either coding or noncoding sequences may be used, and in some instances, noncoding sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a multi-gene family may potentially cause undesired cross hybridization during chromosomal mapping.
  • sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, e.g., human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial PI constructions, or single chromosome cDNA libraries.
  • HACs human artificial chromosomes
  • YACs yeast artificial chromosomes
  • BACs bacterial artificial chromosomes
  • PI constructions or single chromosome cDNA libraries.
  • the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the inheritance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP).
  • RFLP restriction fragment length polymorphism
  • FISH Fluorescent in situ hybridization
  • Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMDV1) World Wide Web site. Correlation between the location of the gene encoding INTSIG on a physical map and a specific disorder, or a predisposition to a specific disorder, may help define the region of DNA associated with that disorder and thus may further positional cloning efforts. In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps. Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the exact chromosomal locus is not known.
  • any sequences mapping to that area may represent associated or regulatory genes for further investigation.
  • the nucleotide sequence of the instant invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals.
  • INTSIG its catalytic or immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds in any of a variety of drug screening techniques.
  • the fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between INTSIG and the agent being tested may be measured.
  • Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest.
  • This method large numbers of different small test compounds are synthesized on a solid substrate. The test compounds are reacted with INTSIG, or fragments thereof, and washed. Bound INTSIG is then detected by methods well known in the art. Purified INTSIG can also be coated directly onto plates for use in the aforementioned drug screening techniques. Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
  • nucleotide sequences which encode INTSIG may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions.
  • Incyte cDNAs were derived from cDNA libraries described in the LJ-FESEQ GOLD database (Incyte Genomics, Palo Alto CA) and shown in Table 4, column 5. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL (Life Technologies), a monophasic solution of phenol and guanidine isothiocyanate. The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated from the lysates with either isopropanol or sodium acetate and ethanol, or by other routine methods.
  • poly(A)+ RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA purification kit (QIAGEN).
  • RNA was provided with RNA and constructed the corresponding cDNA libraries.
  • cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Life Technologies), using the recommended procedures or similar methods known in the art. (See, e.g., Ausubel, 1997, supra, units 5.1-6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes.
  • the cDNA was size-selected (300- 1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis.
  • cDNAs were ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Life Technologies), PCDNA2.1 plasmid (Invitrogen, Carlsbad CA), PBK-CMV plasmid (Stratagene), PCR2-TOPOTA plasmid (Invitrogen), PCMV-ICIS plasmid (Stratagene), pIGEN (Incyte Genomics, Palo Alto CA), or pTNCY (Incyte Genomics), or derivatives thereof.
  • Recombinant plasmids were transformed into competent E. coli cells including XLl-Blue, XLl-BlueMRF, or SOLR from Stratagene or DH5 ⁇ , DH10B, or ElectroMAX DH10B from Life Technologies.
  • Plasmids obtained as described in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Miniprep purification kit (Edge Biosystems, Gaithersburg MD); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid purification systems or the R.E.A.L. PREP 96 plasmid purification kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4°C
  • plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao, V.B. (1994) Anal. Biochem. 216:1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).
  • PICOGREEN dye Molecular Probes, Eugene OR
  • FLUOROSKAN II fluorescence scanner Labsystems Oy, Helsinki, Finland.
  • Incyte cDNA recovered in plasmids as described in Example II were sequenced as follows. Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (Applied Biosystems) thermal cycler or the PTC-200 thermal cycler (MJ Research) in conjunction with the HYDRA microdispenser (Robbins Scientific) or the MICROLAB 2200 (Hamilton) liquid transfer system. cDNA sequencing reactions were prepared using reagents provided by Amersham Pharmacia Biotech or supplied in ABI sequencing kits such as the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems).
  • Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were carried out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (Applied Biosystems) in conjunction with standard ABI protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, 1997, supra, unit 7.7). Some of the cDNA sequences were selected for extension using the techniques disclosed in Example VDI.
  • the polynucleotide sequences derived from Incyte cDNAs were validated by removing vector, linker, and poly(A) sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programming, and dinucleotide nearest neighbor analysis.
  • the Incyte cDNA sequences or translations thereof were then queried against a selection of public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM, and hidden Markov model (HMM)-based protein family databases such as PFAM.
  • HMM hidden Markov model
  • Incyte cDNA sequences were assembled to produce full length polynucleotide sequences.
  • GenBank cDNAs, GenBank ESTs, stitched sequences, stretched sequences, or Genscan-predicted coding sequences were used to extend Incyte cDNA assemblages to full length.
  • MACDNASIS PRO Hitachi Software Engineering, South San Francisco CA
  • LASERGENE software DNASTAR
  • Polynucleotide and polypeptide sequence alignments are generated using default parameters specified by the CLUSTAL algorithm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences.
  • Table 7 summarizes the tools, programs, and algorithms used for the analysis and assembly of Incyte cDNA and full length sequences and provides applicable descriptions, references, and threshold parameters.
  • the first column of Table 7 shows the tools, programs, and algorithms used, the second column provides brief descriptions thereof, the third column presents appropriate references, all of which are incorporated by reference herein in their entirety, and the fourth column presents, where applicable, the scores, probability values, and other parameters used to evaluate the strength of a match between two sequences (the higher the score or the lower the probability value, the greater the identity between two sequences).
  • Genscan is a general-purpose gene identification program which analyzes genomic DNA sequences from a variety of organisms (See Burge, C. and S. Karlin (1997) J. Mol. Biol. 268:78-94, and Burge, C. and S. Karlin (1998) Curr. Opin. Struct. Biol. 8:346-354).
  • the program concatenates predicted exons to form an assembled cDNA sequence extending from a methionine to a stop codon.
  • the output of Genscan is a FASTA database of polynucleotide and polypeptide sequences.
  • Genscan The maximum range of sequence for Genscan to analyze at once was set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode intracellular signaling molecules, the encoded polypeptides were analyzed by querying against PFAM models for intracellular signaling molecules. Potential intracellular signaling molecules were also identified by homology to Incyte cDNA sequences that had been annotated as intracellular signaling molecules. These selected Genscan-predicted sequences were then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences were then edited by comparison to the top BLAST hit from genpept to correct errors in the sequence predicted by Genscan, such as extra or omitted exons.
  • BLAST analysis was also used to find any Incyte cDNA or public cDNA coverage of the Genscan- predicted sequences, thus providing evidence for transcription. When Incyte cDNA coverage was available, this information was used to correct or confirm the Genscan predicted sequence.
  • Full length polynucleotide sequences were obtained by assembling Genscan-predicted coding sequences with Incyte cDNA sequences and/or public cDNA sequences using the assembly process described in Example III. Alternatively, full length polynucleotide sequences were derived entirely from edited or unedited Genscan-predicted coding sequences.
  • Partial cDNA sequences were extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example III were mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster was analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that were subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval was present on more than one sequence in the cluster were identified, and intervals thus identified were considered to be equivalent by transitivity.
  • Partial DNA sequences were extended to full length with an algorithm based on BLAST analysis.
  • GenBank primate such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases
  • the nearest GenBank protein homolog was then compared by BLAST analysis to either Incyte cDNA sequences or GenScan exon predicted sequences described in Example IV.
  • a chimeric protein was generated by using the resultant high-scoring segment pairs (HSPs) to map the translated sequences onto the GenBank protein homolog. Insertions or deletions may occur in the chimeric protein with respect to the original GenBank protein homolog.
  • HSPs high-scoring segment pairs
  • GenBank protein homolog The GenBank protein homolog, the chimeric protein, or both were used as probes to search for homologous genomic sequences from the public human genome databases. Partial DNA sequences were therefore "stretched” or extended by the addition of homologous genomic sequences. The resultant stretched sequences were examined to determine whether it contained a complete gene. VI. Chromosomal Mapping of INTSIG Encoding Polynucleotides The sequences which were used to assemble SEQ DD NO:21-40 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith-Waterman algorithm.
  • centiMorgans The map position of an interval, in centiMorgans, is measured relative to the terminus of the chromosome's p- arm.
  • centiMorgan is a unit of measurement based on recombination frequencies between chromosomal markers. On average, 1 cM is roughly equivalent to 1 megabase (Mb) of DNA in humans, although this can vary widely due to hot and cold spots of recombination.
  • Mb megabase
  • the cM distances are based on genetic markers mapped by Genethon which provide boundaries for radiation hybrid markers whose sequences were included in each of the clusters.
  • Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (See, e.g., Sambrook, supra, ch. 7; Ausubel (1995) supra, ch. 4 and 16.)
  • the product score takes into account both the degree of similarity between two sequences and the length of the sequence match.
  • the product score is a normalized value between 0 and 100, and is calculated as follows: the BLAST score is multiplied by the percent nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences).
  • the BLAST score is calculated by assigning a score of +5 for every base that matches in a high-scoring segment pair (HSP), and -4 for every mismatch. Two sequences may share more than one HSP (separated by gaps). If there is more than one HSP, then the pair with the highest BLAST score is used to calculate the product score.
  • the product score represents a balance between fractional overlap and quality in a BLAST alignment.
  • a product score of 100 is produced only for 100% identity over the entire length of the shorter of the two sequences being compared.
  • a product score of 70 is produced either by 100% identity and 70% overlap at one end, or by 88% identity and 100% overlap at the other.
  • a product score of 50 is produced either by 100% identity and 50% overlap at one end, or 79% identity and 100% overlap.
  • polynucleotide sequences encoding INTSIG are analyzed with respect to the tissue sources from which they were derived. For example, some full length sequences are assembled, at least in part, with overlapping Incyte cDNA sequences (see Example ID). Each cDNA sequence is derived from a cDNA library constructed from a human tissue.
  • Each human tissue is classified into one of the following organ/tissue categories: cardiovascular system; connective tissue; digestive system; embryonic structures; endocrine system; exocrine glands; genitalia, female; genitalia, male; germ cells; hemic and immune system; liver; musculoskeletal system; nervous system; pancreas; respiratory system; sense organs; skin; stomatognathic system; unclassified/mixed; or urinary tract.
  • the number of libraries in each category is counted and divided by the total number of libraries across all categories.
  • each human tissue is classified into one of the following disease/condition categories: cancer, cell line, developmental, inflammation, neurological, trauma, cardiovascular, pooled, and other, and the number of libraries in each category is counted and divided by the total number of libraries across all categories. The resulting percentages reflect the tissue- and disease-specific expression of cDNA encoding INTSIG.
  • cDNA sequences and cDNA library/tissue information are found in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA).
  • SEQ DD NO:30 shows a strong association with neurological tissues. 1292 libraries present in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA) isolated from 20 tissue types were examined.
  • SEQ ID NO:30 was found in 73 libraries, 43 (59%) of which were isolated from neurological tissues. Of 113 incidences of SEQ DD NO:30 in all libraries, 75 were in nervous system libraries. SEQ IN NO:30 is useful for distinguishing between nervous tissues and, for example, cardiovascular or endocrine tissues. VIII. Extension of INTSIG Encoding Polynucleotides Full length polynucleotide sequences were also produced by extension of an appropriate fragment of the full length molecule using oligonucleotide primers designed from this fragment. One primer was synthesized to initiate 5' extension of the known fragment, and the other primer was synthesized to initiate 3' extension of the known fragment.
  • the initial primers were designed using OLIGO 4.06 software (National Biosciences), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68 °C to about 72 °C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
  • Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed. High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the PTC-200 thermal cycler (MJ Research, Inc.).
  • the reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg 2+ , (NH 4 ) 2 S0 4 , and 2-mercaptoethanol, Taq DNA polymerase (Amersham Pharmacia Biotech), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B: Step 1 : 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 68 °C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68 °C, 5 min; Step 7: storage at 4°C
  • the parameters for primer pair T7 and SK+ were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68
  • the plate was scanned in a Fluoroskan II (Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA.
  • a 5 ⁇ l to 10 ⁇ l aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose gel to determine which reactions were successful in extending the sequence.
  • the extended nucleotides were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC 18 vector (Amersham Pharmacia Biotech).
  • CviJI cholera virus endonuclease Molecular Biology Research, Madison WI
  • sonicated or sheared prior to religation into pUC 18 vector
  • the digested nucleotides were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with Agar ACE (Promega).
  • Extended clones were religated using T4 ligase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Pharmacia Biotech), treated with Pfu DNA polymerase (Stratagene) to fill-in restriction site overhangs, and transfected into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37 °C in 384- well plates in LB/2x carb liquid media.
  • the cells were lysed, and DNA was amplified by PCR using Taq DNA polymerase (Amersham Pharmacia Biotech) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 72°C, 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72°C, 5 min; Step 7: storage at 4°C DNA was quantified by PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions as described above.
  • Hybridization probes derived from SEQ DD NO:21-40 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments. Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 software (National Biosciences) and labeled by combining 50 p ol of each oligomer, 250 Ci of [ ⁇ - 32 P] adenosine rriphosphate (Amersham Pharmacia Biotech), and T4 polynucleotide kinase (DuPont NEN, Boston MA).
  • the labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech). An aliquot containing 10 7 counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl D, Eco RI, Pst I, Xba I, or Pvu II (DuPont NEN).
  • the DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham NH). Hybridization is carried out for 16 hours at 40 °C. To remove nonspecific signals, blots are sequentially washed at room temperature under conditions of up to, for example, 0.1 x saline sodium citrate and 0.5% sodium d ⁇ decyl sulfate. Hybridization patterns are visualized using autoradiography or an alternative imaging means and compared. X. Microarrays
  • the linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectric printing (ink-jet printing, See, e.g., Baldeschweiler, supra.), mechanical microspotting technologies, and derivatives thereof.
  • the substrate in each of the aforementioned technologies should be uniform and solid with a non-porous surface (Schena (1999), supra). Suggested substrates include silicon, silica, glass slides, glass chips, and silicon wafers. Alternatively, a procedure analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures.
  • a typical array may be produced using available methods and machines well known to those of ordinary skill in the art and may contain any appropriate number of elements. (See, e.g., Schena, M. et al. (1995) Science 270:467-470; Shalon, D. et al. (1996) Genome Res. 6:639-645; Marshall, A. and J. Hodgson (1998) Nat. Biotechnol. 16:27-31.)
  • Full length cDNAs, Expressed Sequence Tags (ESTs), or fragments or oligomers thereof may comprise the elements of the microarray. Fragments or oligomers suitable for hybridization can be selected using software well known in the art such as LASERGENE software (DNASTAR).
  • the array elements are hybridized with polynucleotides in a biological sample.
  • the polynucleotides in the biological sample are conjugated to a fluorescent label or other molecular tag for ease of detection.
  • a fluorescence scanner is used to detect hybridization at each array element.
  • laser desorbtion and mass spectrometry may be used for detection of hybridization.
  • the degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed.
  • microarray preparation and usage is described in detail below.
  • Total RNA is isolated from tissue samples using the guanidinium thiocyanate method and poly(A) + RNA is purified using the oligo-(dT) cellulose method.
  • Each poly(A) + RNA sample is reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/ ⁇ l oligo-(dT) primer (21mer), IX first strand buffer, 0.03 units/ ⁇ l RNase inhibitor, 500 ⁇ M dATP, 500 ⁇ M dGTP, 500 ⁇ M dTTP, 40 ⁇ M dCTP, 40 ⁇ M dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech).
  • the reverse transcription reaction is performed in a 25 ml volume containing 200 ng poly(A) + RNA with GEMBRIGHT kits (Incyte).
  • Specific control poly(A) + RNAs are synthesized by in vitro transcription from non-coding yeast genomic DNA. After incubation at 37° C for 2 hr, each reaction sample (one with Cy3 and another with Cy5 labeling) is treated with 2.5 ml of 0.5M sodium hydroxide and incubated for 20 minutes at 85° C to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (CLONTECH Laboratories, Inc.
  • Sequences of the present invention are used to generate array elements.
  • Each array element is amplified from bacterial cells containing vectors with cloned cDNA inserts.
  • PCR amplification uses primers complementary to the vector sequences flanking the cDNA insert.
  • Array elements are amplified in thirty cycles of PCR from an initial quantity of 1-2 ng to a final quantity greater than 5 ⁇ g.
  • Amplified array elements are then purified using SEPHACRYL-400 (Amersham Pharmacia Biotech). Purified array elements are immobilized on polymer-coated glass slides. Glass microscope slides (Corning) are cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments.
  • Array elements are applied to the coated glass substrate using a procedure described in U.S. Patent No. 5,807,522, incorporated herein by reference.
  • 1 ⁇ l of the array element DNA, at an average concentration of 100 ng/ ⁇ l, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 nl of array element sample per slide.
  • Microarrays are UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene). Microarrays are washed at room temperature once in 0.2% SDS and three times in distilled water. Non-specific binding sites are blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (PBS) (Tropix, Inc., Bedford MA) for 30 minutes at 60° C followed by washes in 0.2% SDS and distilled water as before. Hybridization
  • Hybridization reactions contain 9 ⁇ l of sample mixture consisting of 0.2 ⁇ g each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer.
  • the sample mixture is heated to 65° C for 5 minutes and is aliquoted onto the microarray surface and covered with an 1.8 cm 2 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 5X SSC in a corner of the chamber.
  • the chamber containing the arrays is incubated for about 6.5 hours at 60° C.
  • the arrays are washed for 10 min at 45° C in a first wash buffer (IX SSC, 0.1% SDS), three times for 10 minutes each at 45° C in a second wash buffer (0.1X SSC), and dried. Detection
  • Reporter-labeled hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Inc., Santa Clara CA) 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 20X microscope objective (Nikon, Inc., Melville NY).
  • the slide containing the array is placed on a computer-controlled X-Y stage on the microscope and raster- scanned past the objective.
  • the 1.8 cm x 1.8 cm array used in the present example is scanned with a resolution of 20 micrometers.
  • a mixed gas multiline laser excites the two fluorophores sequentially. Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477,
  • 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 calibration is done by labeling samples of the calibrating cDNA with the two fluorophores and adding identical amounts of each to the hybridization mixture.
  • the output of the photomultiplier tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Inc., Norwood MA) 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 each fluorophore's emission spectrum.
  • 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 gene expression analysis program (Incyte).
  • Sequences complementary to the INTSIG-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring INTSIG. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of INTSIG. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the INTSIG-encoding transcript. XII. Expression of INTSIG
  • INTSIG expression and purification of INTSIG is achieved using bacterial or virus-based expression systems.
  • cDNA is subcloned into an appropriate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcription.
  • promoters include, but are not limited to, the trp-lac (tac) hybrid promoter and the T5 or T7 bacteriophage promoter in conjunction with the lac operator regulatory element.
  • Recombinant vectors are transformed into suitable bacterial hosts, e.g., BL21(DE3).
  • Antibiotic resistant bacteria express INTSIG upon induction with isopropyl beta-D- thiogalactopyranoside (IPTG).
  • INTSIG in eukaryotic cells
  • baculovirus recombinant Autographica calif ornica nuclear polyhedrosis virus
  • AcMNPV Autographica calif ornica nuclear polyhedrosis virus
  • the nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding INTSIG by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates. Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of cDNA transcription.
  • Recombinant baculovirus is used to infect Spodoptera frugiperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases.
  • INTSIG is synthesized as a fusion protein with, e.g., glutathione S-transferase (GST) or a peptide epitope tag, such as FLAG or 6-His, permitting rapid, single-step, affinity-based purification of recombinant fusion protein from crude cell lysates.
  • GST glutathione S-transferase
  • a peptide epitope tag such as FLAG or 6-His
  • FLAG an 8-amino acid peptide
  • 6- His a stretch of six consecutive histidine residues, enables purification on metal-chelate resins (QIAGEN). Methods for protein expression and purification are discussed in Ausubel (1995, supra, ch. 10 and 16). Purified INTSIG obtained by these methods can be used directly in the assays shown in Examples XVI, XV ⁇ , and XVDI, where applicable. XIII. Functional Assays
  • INTSIG function is assessed by expressing the sequences encoding INTSIG at physiologically elevated levels in mammalian cell culture systems.
  • cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA expression.
  • Vectors of choice include PCMV SPORT (Life Technologies) and PCR3.1 (Invitrogen, Carlsbad CA), both of which contain the cytomegalovirus promoter. 5-10 ⁇ g of recombinant vector are transiently transfected into a human cell line, for example, an endothelial or hematopoietic cell line, using either liposome formulations or elecrroporation.
  • 1-2 ⁇ g of an additional plasmid containing sequences encoding a marker protein are co-transfected.
  • Expression of a marker protein provides a means to distinguish transfected cells from nontransfected cells and is a reliable predictor of cDNA expression from the recombinant vector.
  • Marker proteins of choice include, e.g., Green Fluorescent Protein (GFP; Clontech), CD64, or a CD64-GFP fusion protein.
  • FCM Flow cytometry
  • FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death. These events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide; changes in cell size and granularity as measured by forward light scatter and 90 degree side light scatter; down-regulation of DNA synthesis as measured by decrease in bromodeoxyuridine uptake; alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell surface. Methods in flow cytometry are discussed in Ormerod, M.G. (1994) Flow Cytometry, Oxford, New York NY.
  • the influence of INTSIG on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding INTSIG and either CD64 or CD64-GFP.
  • CD64 and CD64-GFP are expressed on the surface of transfected cells and bind to conserved regions of human immunoglobulin G (IgG).
  • Transfected cells are efficiently separated from nontransfected cells using magnetic beads coated with either human IgG or antibody against CD64 (DYNAL, Lake Success NY).
  • mRNA can be purified from the cells using methods well known by those of skill in the art. Expression of mRNA encoding INTSIG and other genes of interest can be analyzed by northern analysis or microarray techniques.
  • INTSIG substantially purified using polyacrylamide gel electrophoresis (PAGE; see, e.g., Harrington, M.G. (1990) Methods Enzymol. 182:488-495), or other purification techniques, is used to immunize rabbits and to produce antibodies using standard protocols.
  • PAGE polyacrylamide gel electrophoresis
  • the INTSIG amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art.
  • LASERGENE software DNASTAR
  • Methods for selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions are well described in the art. (See, e.g., Ausubel, 1995, supra, ch. 11.)
  • oligopeptides of about 15 residues in length are synthesized using an ABI 431 A peptide synthesizer (Applied Biosystems) using FMOC chemistry and coupled to KLH (Sigma- Aldrich, St. Louis MO) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) to increase immunogenicity.
  • ABI 431 A peptide synthesizer Applied Biosystems
  • KLH Sigma- Aldrich, St. Louis MO
  • MBS N-maleimidobenzoyl-N-hydroxysuccinimide ester
  • Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant.
  • Resulting antisera are tested for antipeptide and anti-INTSIG activity by, for example, binding the peptide or INTSIG to a substrate, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG.
  • Naturally occurring or recombinant INTSIG is substantially purified by immunoaffinity chromatography using antibodies specific for INTSIG.
  • An immunoaffinity column is constructed by covalently coupling anti-INTSIG antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions. Media containing INTSIG are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of INTSIG (e.g., high ionic strength buffers in the presence of detergent).
  • the column is eluted under conditions that disrupt antibody/TNTSIG binding (e.g., a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and INTSIG is collected.
  • a chaotrope such as urea or thiocyanate ion
  • INTSIG or biologically active fragments thereof, are labeled with 125 I Bolton-Hunter reagent.
  • Bolton-Hunter reagent See, e.g., Bolton, A.E. and W.M. Hunter (1973) Biochem. J. 133:529-539.
  • Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled INTSIG, washed, and any wells with labeled INTSIG complex are assayed. Data obtained using different concentrations of INTSIG are used to calculate values for the number, affinity, and association of INTSIG with the candidate molecules.
  • molecules interacting with INTSIG are analyzed using the yeast two-hybrid system as described in Fields, S. and O. Song (1989) Nature 340:245-246, or using commercially available kits based on the two-hybrid system, such as the MATCHMAKER system (Clontech).
  • INTSIG may also be used in the PATHCALLING process (CuraGen Corp., New Haven CT) which employs the yeast two-hybrid system in a high-throughput manner to determine all interactions between the proteins encoded by two large libraries of genes (Nandabalan, K. et al. (2000) U.S. Patent No. 6,057,101). XVII. Demonstration of INTSIG Activity
  • INTSIG activity is associated with its ability to form protein-protein complexes and is measured by its ability to regulate growth characteristics of NDT3T3 mouse fibroblast cells.
  • a cDNA encoding INTSIG is subcloned into an appropriate eukaryotic expression vector. This vector is transfected into NIH3T3 cells using methods known in the art. Transfected cells are compared with non-transfected cells for the following quantifiable properties: growth in culture to high density, reduced attachment of cells to the substrate, altered cell morphology, and ability to induce tumors when injected into immunodeficient mice.
  • the activity of INTSIG is proportional to the extent of increased growth or frequency of altered cell morphology in NIH3T3 cells transfected with INTSIG.
  • INTSIG activity is measured by binding of INTSIG to radiolabeled for in polypeptides containing the proline-rich region that specifically binds to SH3 containing proteins (Chan, D.C et al. (1996) EMBO J. 15:1045-1054).
  • Samples of INTSIG are run on SDS-PAGE gels, and transferred onto nitrocellulose by electroblotting. The blots are blocked for 1 hr at room temperature in TBST (137 mM NaCl, 2.7 mM KC1, 25 mM Tris (pH 8.0) and 0.1% Tween-20) containing non-fat dry milk. Blots are then incubated with TBST containing the radioactive formin polypeptide for 4 hrs to overnight.
  • the blots After washing the blots four times with TBST, the blots are exposed to autoradiographic film. Radioactivity is quantitated by cutting out the radioactive spots and counting them in a radioisotope counter. The amount of radioactivity recovered is proportional to the activity of INTSIG in the assay.
  • INTSIG protein kinase activity is measured by quantifying the phosphorylation of an appropriate substrate in the presence of gamma-labeled 32 P-ATP.
  • INTSIG is incubated with the substrate, 32 P-ATP, and an appropriate kinase buffer.
  • the 32 P incorporated into the product is separated from free 32 P-ATP by electrophoresis, and the incorporated 32 P is quantified using a beta radioisotope counter.
  • the amount of incorporated 3 P is proportional to the protein kinase activity of INTSIG in the assay.
  • a determination of the specific amino acid residue phosphorylated by protein kinase activity is made by phosphoamino acid analysis of the hydrolyzed protein.
  • an assay for INTSIG protein phosphatase activity measures the hydrolysis of para-nitrophenyl phosphate (PNPP).
  • INTSIG is incubated together with PNPP in HEPES buffer pH 7.5, in the presence of 0.1% ⁇ -mercaptoethanol at 37°C for 60 min.
  • the reaction is stopped by the addition of 6 ml of 10 N NaOH, and the increase in light absorbance of the reaction mixture at 410 nm resulting from the hydrolysis of PNPP is measured using a spectrophotometer.
  • the increase in light absorbance is proportional to the activity of INTSIG in the assay (Diamond, R.H. et al. (1994) Mol. Cell Biol. 14:3752-3762).
  • An alternative assay measures INTSIG-mediated G-protein signaling activity by monitoring the mobilization of Ca 2+ as an indicator of the signal transduction pathway stimulation.
  • the assay requires preloading neutrophils or T cells with a fluorescent dye such as FURA-2 or BCECF (Universal Imaging Corp, Westchester PA) whose emission characteristics are altered by Ca ++ binding.
  • Ca ++ flux takes place. This flux can be observed and quantified by assaying the cells in a fluorometer or fluorescent activated cell sorter. Measurements of Ca** flux are compared between cells in their normal state and those transfected with INTSIG. Increased Ca ++ mobilization attributable to increased INTSIG concentration is proportional to INTSIG activity.
  • activating stimuli e.g., anti-CD3 antibody ligation of the T cell receptor
  • physiologically e.g., by allogeneic stimulation
  • INTSIG activity is measured by binding of INTSIG to a substrate which recognizes WD-40 repeats, such as ElonginB, by coimmunoprecipitation (Kamura, T. et al. (1998) Genes Dev. 12:3872-3881). Briefly, epitope tagged substrate and INTSIG are mixed and immunoprecipitated with commercial antibody against the substrate tag. The reaction solution is run on SDS-PAGE and the presence of INTSIG visualized using an antibody to the INTSIG tag. Substrate binding is proportional to INTSIG activity.
  • INTSIG activity is measured by measuring oxysterol binding.
  • Epitope-tagged INTSIG is incubated with a radio-labeled oxysterol ligand, such as 3 H-25-hydroxycholesterol.
  • INTSIG is collected by immunoprecipitation with a commercial antibody against the epitope, and bound hydroxycholesterol quantitated by scintillation counting.
  • INTSIG activity is proportional to the amount of ligand bound.
  • the binding of INTSIG to RNA can be assayed using a solid phase RNA binding assay.
  • Hemagglutinin- (HA) tagged wild type and mutant INTSIG in pcDNA3 are transiently transfected into COS cells using LipofectAMINE reagent (Life Technologies, Inc.) for expression and analysis of RNA binding to multiple, simutaneously purified INTSIG proteins.
  • Anti-HA immunoprecipitated INTSIG bound to protein G-Sepharose is incubated with 30 ng of 32 P-labeled G8-5 RNA in 30 ⁇ l of RNA binding buffer containing 1 ⁇ g/ ⁇ l poly(C) at room temperature for 20 min. with occasional shaking.
  • RNA binding buffer washed twice with 700 ⁇ l of RNA binding buffer and resuspended in 20 ⁇ l of SDS-polyacrylamide gel electrophoresis sample buffer.
  • the protein and RNA were separated by 10% SDS-polyacrylamdie gel electrophoresis.
  • the RNA bands ran with a mobility equivalent to 25-35 kDa, and this part of the gel is cut out and dried for autoradiography.
  • the upper part of the gel is transferred to a polyvinylidene difluoride membrane and blotted with anti-HA antibody to detect HA-INTSIG (Lin, et al. (1997) J. Biol. Chem. 272:27274-27280).
  • ABI FACTURA A program that removes vector sequences and Applied Biosystems, Foster City, CA. masks ambiguous bases in nucleic acid sequences.
  • ABI/PARACEL FDF A Fast Data Finder useful in comparing and Applied Biosystems, Foster City, CA; Mismatch ⁇ 50% annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA.
  • ABI AutoAssembler A program that assembles nucleic acid sequences. Applied Biosystems, Foster City, CA.
  • BLAST includes five Nucleic Acids Res. 25:3389-3402. Full Length sequences: Probabilit functions: blastp, blastn, blasts, tblastn, and tblastx. valuer l.OE-10 or less
  • fastx score 100 or greater
  • HMM hidden Markov model
  • Phred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res. sequencer traces with high sensitivity and probability. 8:175-185; Ewing, B. and P. Green (1998) Genome Res. 8:186-194.
  • TMHMMER A program that uses a hidden Markov model (HMM) to Sonnhammer, E.L. et al. (1998) Proc. Sixth Intl. delineate transmembrane segments on protein sequences Conf. on Intelligent Systems for Mol. Biol., and determine orientation. Glasgow et al., eds., The Am. Assoc. for Artificial Intelligence Press, Menlo Park, CA, pp. 175-182.
  • HMM hidden Markov model

Landscapes

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

Abstract

The invention provides human intracellular signaling molecules (INTSIG) and polynucleotides which identify and encode INTSIG. The invention also provides expression vectors, host cells, antibodies, agonists, and antagonists. The invention also provides methods for diagnosing, treating, or preventing disorders associated with aberrant expression of INTSIG.

Description

INTRACELLULAR SIGNALING MOLECULES
TECHNICAL FIELD
This invention relates to nucleic acid and amino acid sequences of intracellular signaling molecules and to the use of these sequences in the diagnosis, treatment, and prevention of cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of intracellular signaling molecules.
BACKGROUND OF THE INVENTION
Cell-cell communication is essential for the growth, development, and survival of multicellular organisms. Cells communicate by sending and receiving molecular signals. An example of a molecular signal is a growth factor, which binds and activates a specific transmembrane receptor on the surface of a target cell. The activated receptor transduces the signal intracellularly, thus initiating a cascade of biochemical reactions that ultimately affect gene transcription and cell cycle progression in the target cell.
Intracellular signaling is the process by which cells respond to extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.) through a cascade of biochemical reactions that begins with the binding of a signaling molecule to a cell membrane receptor and ends with the activation of an intracellular target molecule. Intermediate steps in the process involve the activation of various cytoplasmic proteins by phosphorylation via protein kinases, and their deactivation by protein phosphatases, and the eventual translocation of some of these activated proteins to the cell nucleus where the transcription of specific genes is triggered. The intracellular signaling process regulates all types of cell functions including cell proliferation, cell differentiation, and gene transcription, and involves a diversity of molecules including protein kinases and phosphatases, and second messenger molecules such as cyclic nucleotides, calcium-calmodulin, inositol, and various mitogens that regulate protein phosphorylation.
Cells also respond to changing conditions by switching off signals. Many signal transduction proteins are short-lived and rapidly targeted for degradation by covalent ligation to ubiquitin, a highly conserved small protein. Cells also maintain mechanisms to monitor changes in the concentration of denatured or unfolded proteins in membrane-bound extracytoplasmic compartments, including a transmembrane receptor that monitors the concentration of available chaperone molecules in the endoplasmic reticulum and transmits a signal to the cytosol to activate the transcription of nuclear genes encoding chaperones in the endoplasmic reticulum. Certain proteins in intracellular signaling pathways serve to link or cluster other proteins involved in the signaling cascade. These proteins are referred to as scaffold, anchoring, or adaptor proteins. (For review, see Pawson, T. and J.D. Scott (1997) Science 278:2075-2080.) As many intracellular signaling proteins such as protem kinases and phosphatases have relatively broad substrate specificities, the adaptors help to organize the component signaling proteins into specific biochemical pathways. Many of the above signaling molecules are characterized by the presence of particular domains that promote protein-protein interactions. A sampling of these domains is discussed below, along with other important intracellular messengers.
Intracellular Signaling Second Messenger Molecules
Protem Phosphorylation
Protein kinases and phosphatases play a key role in the intracellular signaling process by controlling the phosphorylation and activation of various signaling proteins. The high energy phosphate for this reaction is generally transferred from the adenosine triphosphate molecule (ATP) to a particular protein by a protein kinase and removed from that protein by a protein phosphatase. Protem kinases are roughly divided into two groups: those that phosphorylate serine or threonine residues (serine/threonine kinases, STK) and those that phosphorylate tyrosine residues (protem tyrosine kinases, PTK). A few protein kinases have dual specificity for serine/threonine and tyrosine residues. Almost all kinases contain a conserved 250-300 amino acid catalytic domain containing specific residues and sequence motifs characteristic of the kinase family (Hardie, G. and S. Hanks (1995) The Protem Kinase Facts Books. Vol 1:7-20, Academic Press, San Diego, CA).
STKs include the second messenger dependent protein kinases such as the cyclic -AMP dependent protein kinases (PKA), involved in mediating hormone-induced cellular responses; calcium-calmodulin (CaM) dependent protein kinases, involved in regulation of smootii muscle contraction, glycogen breakdown, and neurotransmission; and the mitogen-activated protein kinases (MAP) which mediate signal transduction from the cell surface to the nucleus via phosphorylation cascades. Altered PKA expression is implicated in a variety of disorders and diseases including cancer, thyroid disorders, diabetes, atherosclerosis, and cardiovascular disease (Isselbacher, KJ. et al. (1994) Harrison's Principles of Internal Medicine. McGraw-Hill, New York, NY, pp. 416-431, 1887). PTKs are divided into transmembrane, receptor PTKs and nontransmembrane, non-receptor
PTKs. Transmembrane PTKs are receptors for most growth factors. Non-receptor PTKs lack transmembrane regions and, instead, form complexes with the intracellular regions of cell surface receptors. Receptors that function through non-receptor PTKs include those for cytokines and hormones (growth hormone and prolactin) and antigen-specific receptors on T and B lymphocytes. Many of these PTKs were first identified as the products of mutant oncogenes in cancer cells in which their activation was no longer subject to normal cellular controls. In fact, about one third of the known oncogenes encode PTKs, and it is well known that cellular transformation (oncogenesis) is often accompanied by increased tyrosine phosphorylation activity (Charbonneau H. and N.K. Tonks (1992) Annu. Rev. Cell Biol. 8:463-493).
An additional family of protein kinases previously thought to exist only in prokaryotes is the histidine protein kinase family (HPK). HPKs bear little homology with mammalian STKs or PTKs but have distinctive sequence motifs of their own (Davie, J.R. et al. (1995) J. Biol. Chem. 270:19861-19867). A histidine residue in the N-terminal half of the molecule (region I) is an autophosphorylation site. Three additional motifs located in the C-terminal half of the molecule include an invariant asparagine residue in region π and two glycine-rich loops characteristic of nucleotide binding domains in regions Ul and IV. Recently a branched chain alpha-ketoacid dehydrogenase kinase has been found with characteristics of HPK in rat (Davie et al., supra).
Protein phosphatases regulate the effects of protein kinases by removing phosphate groups from molecules previously activated by kinases. The two principal categories of protein phosphatases are the protein (serine/threonine) phosphatases (PPs) and the protein tyrosine phosphatases (PTPs). PPs dephosphorylate phosphoserine/threonine residues and are important regulators of many cAMP-mediated hormone responses (Cohen, P. (1989) Annu. Rev. Biochem. 58:453-508). PTPs reverse the effects of protem tyrosine kinases and play a significant role in cell cycle and cell signaling processes (Charbonneau and Tonks, supra). As previously noted, many PTKs are encoded by oncogenes, and oncogenesis is often accompanied by increased tyrosine phosphorylation activity. It is therefore possible that PTPs may prevent or reverse cell transformation and the growth of various cancers by controlling the levels of tyrosine phosphorylation in cells. This hypothesis is supported by studies showing that overexpression of PTPs can suppress transformation in cells, and that specific inhibition of PTPs can enhance cell transformation (Charbonneau and Tonks, supra). Phospholipid and Inositol-phosphate Signaling
Inositol phospholipids (phosphoinositides) are involved in an intracellular signaling pathway that begins with binding of a signaling molecule to a G-protein linked receptor in the plasma membrane. This leads to the phosphorylation of phosphatidylinositol (PI) residues on the inner side of the plasma membrane to the biphosphate state (PIP2) by inositol kinases. Simultaneously, the G-protein linked receptor binding stimulates a trimeric G-protein which in turn activates a phosphoinositide-specific phospholipase C-β. Phospholipase C-β then cleaves PIP2 into two products, inositol triphosphate (IP3) and diacylglycerol. These two products act as mediators for separate signaling events. IP3 diffuses through the plasma membrane to induce calcium release from the endoplasmic reticulum (ER), while diaacylglycerol remains in the membrane and helps activate protein kinase C, a serine-threonine kinase that phosphorylates selected proteins in the target cell. The calcium response initiated by IP3 is terminated by the dephosphorylation of IP3 by specific inositol phosphatases. Cellular responses that are mediated by this pathway are glycogen breakdown in the liver in response to vasopressin, smooth muscle contraction in response to acetylcholine, and thrombin-induced platelet aggregation.
Oxysterols are oxygenated derivatives of cholesterol and have a wide range of biological activities. Oxysterols mediate cholesterol homeostasis, steroid biosynthesis and sphingolipid metabolism within the cell, but can also diffuse through the plasma membrane and act as extracellular messengers, affecting such processes as platelet aggregation, cell growth and apoptosis. Oxysterols interact with a number of receptors, including the oxysterol binding protein (OSBP), the sterol regulatory element binding protein, the cellular nucleic acid binding protein, the LXR nuclear hormone receptors, and the LDL receptor (for a review, see Schroepfer, G.J. (2000) Physiol. Rev. 80:361-554). OSBP is a high-affinity intracellular receptor for a variety of oxysterols that down-regulate cholesterol synthesis and stimulate cholesterol esterification. Upon ligand binding, OSBP translocates from the cytoplasm to the Golgi. This movement seems to be dependent on the presence of a pleckstrin homology domain (Lagace, T.A. et al. (1997) Biochem. J. 326:205-213). The oxysterol-induced apoptosis of leukemic T-cells seems to be mediated by OSBP occupancy (Bakos, J.T. et al. (1993) J. Steroid Biochem. Mol. Biol. 46:415-426). Cyclic Nucleotide Signaling Cyclic nucleotides (cAMP and cGMP) function as intracellular second messengers to transduce a variety of extracellular signals including hormones, light, and neurotransmitters. In particular, cyclic-AMP dependent protein kinases (PKA) are thought to account for all of the effects of cAMP in most mammalian cells, including various hormone-induced cellular responses. Visual excitation and the phototransmission of light signals in the eye is controlled by cyclic-GMP regulated, Ca2+-specific channels. Because of the importance of cellular levels of cyclic nucleotides in mediating these various responses, regulating the synthesis and breakdown of cyclic nucleotides is an important matter. Thus adenylyl cyclase, which synthesizes cAMP from AMP, is activated to increase cAMP levels in muscle by binding of adrenaline to β-adrenergic receptors, while activation of guanylate cyclase and increased cGMP levels in photoreceptors leads to reopening of the Ca2+-specific channels and recovery of the dark state in the eye. In contrast, hydrolysis of cyclic nucleotides by cAMP and cGMP-specific phosphodiesterases (PDEs) produces the opposite of these and other effects mediated by increased cyclic nucleotide levels. PDEs appear to be particularly important in the regulation of cyclic nucleotides, considering the diversity found in this family of proteins. At least seven families of mammalian PDEs (PDE1-7) have been identified based on substrate specificity and affinity, sensitivity to cofactors, and sensitivity to inhibitory drugs (Beavo, J.A. (1995) Physiol. Rev. 75:725-748). PDE inhibitors have been found to be particularly useful in treating various clinical disorders. Rolipram, a specific inhibitor of PDE4, has been used in the treatment of depression, and similar inhibitors are undergoing evaluation as anti-inflammatory agents. Theophylline is a nonspecific PDE inhibitor used in the treatment of bronchial asthma and other respiratory diseases (Banner, K.H. and CP. Page (1995) Eur. Respir. J. 8:996-1000). G-Protein Signaling
Guanine nucleotide binding proteins (G-proteins) are critical mediators of signal transduction between a particular class of extracellular receptors, the G-protein coupled receptors (GPCRs), and intracellular second messengers such as cAMP and Ca2+. G-proteins are linked to the cytosolic side of a GPCR such that activation of the GPCR by ligand binding stimulates binding of the G-protein to GTP, inducing an "active" state in the G-protein. In the active state, the G-protein acts as a signal to trigger other events in the cell such as the increase of cAMP levels or the release of Ca2+ into the cytosol from the ER, which, in turn, regulate phosphorylation and activation of other intracellular proteins. Recycling of the G-protein to the inactive state involves hydrolysis of the bound GTP to GDP by a GTPase activity in the G-protein. (See Alberts, B. et al. (1994) Molecular Biology of the Cell Garland Publishing, Inc. New York, NY, pp.734-759.) Two structurally distinct classes of G- proteins are recognized: heterotrimeric G-proteins, consisting of three different subunits, and monomeric, low molecular weight (LMW), G-proteins consisting of a single polypeptide chain. The three polypeptide subunits of heterotrimeric G-proteins are the α, β, and γ subunits. The subunit binds and hydrolyzes GTP. The β and γ subunits form a tight complex that anchors the protein to the inner side of the plasma membrane. The β subunits, also known as G-β proteins or β transducins, contain seven tandem repeats of the WD-repeat sequence motif, a motif found in many proteins with regulatory functions. Mutations and variant expression of β transducfn proteins are linked with various disorders (Neer, E. J. et al. (1994) Nature 371 :297-300; Margottin, F. et al. (1998) Mol. Cell. 1:565-574).
LMW GTP-proteins are GTPases which regulate cell growth, cell cycle control, protein secretion, and intracellular vesicle interaction. They consist of single polypeptides which, like the α subunit of the heterotrimeric G-proteins, are able to bind and hydrolyze GTP, thus cycling between an inactive and an active state. At least sixty members of the LMW G-protein superfamily have been identified and are currently grouped into the six subfamilies of ras, rho, arf, sari, ran, and rab. Activated ras genes were initially found in human cancers, and subsequent studies confirmed that ras function is critical in determining whether cells continue to grow or become differentiated. Other members of the LMW G-protein superfamily have roles in signal transduction that vary with the function of the activated genes and the locations of the G-proteins.
Guanine nucleotide exchange factors regulate the activities of LMW G-proteins by determining whether GTP or GDP is bound. GTPase-activating protein (GAP) binds to GTP-ras and induces it to hydrolyze GTP to GDP. In contrast, guanine nucleotide releasing protein (GNRP) binds to GDP-ras and induces the release of GDP and the binding of GTP.
Other regulators of G-protein signaling (RGS) also exist that act primarily by negatively regulating the G-protein pathway by an unknown mechanism (Druey, K.M. et al. (1996) Nature 379:742-746). Some 15 members of the RGS family have been identified. RGS family members are related structurally through similarities in an approximately 120 amino acid region termed the RGS domain and functionally by their ability to inhibit the interleukin (cytokine) induction of MAP kinase in cultured mammalian 293T cells (Druey et al., supra). Calcium Signaling Molecules
Ca2+ is another second messenger molecule that is even more widely used as an intracellular mediator than cAMP. Ca2+ can enter the cytosol by two pathways, in response to extracellular signals. One pathway acts primarily in nerve signal transduction where Ca2+ enters a nerve terminal through a voltage-gated Ca2+ channel. The second is a more ubiquitous pathway in which Ca2+ is released from the ER into the cytosol in response to binding of an extracellular signaling molecule to a receptor. Ca2+ directly activates regulatory enzymes, such as protein kinase C, which trigger signal transduction pathways. Ca2+ also binds to specific Ca +-binding proteins (CBPs) such as calmodulin (CaM) which then activate multiple target proteins in the cell including enzymes, membrane transport pumps, and ion channels. CaM interactions are involved in a multitude of cellular processes including, but not limited to, gene regulation, DNA synthesis, cell cycle progression, mitosis, cytokinesis, cytoskeletal organization, muscle contraction, signal transduction, ion homeostasis, exocytosis, and metabolic regulation (Celio, M.R. et al. (1996) Guidebook to Calcium-binding Proteins. Oxford University Press, Oxford, UK, pp. 15-20). Some Ca2+ binding proteins are characterized by the presence of one or more EF-hand Ca2+ binding motifs, which are comprised of 12 amino acids flanked by α-helices (Celio, supra). The regulation of CBPs has implications for the control of a variety of disorders. Calcineurin, a CaM-regulated protein phosphatase, is a target for inhibition by the immunosuppressive agents cyclosporin and FK506. This indicates the importance of calcineurin and CaM in the immune response and immune disorders (Schwaninger M. et al. (1993) J. Biol Chem. 268:23111-23115). The level of CaM is increased several-fold in tumors and tumor-derived cell lines for various types of cancer (Rasmussen, CD. and A.R. Means (1989) Trends Neurosci. 12:433-438). The annexins are a family of calcium-binding proteins that associate with the cell membrane (Towle, CA. and B.V. Treadwell (1992) J. Biol. Chem. 267:5416-5423). Annexins reversibly bind to negatively charged phospholipids (phosphatidylcholine and phosphatidylserine) in a calcium dependent manner. Annexins participate in various processes pertaining to signal transduction at the plasma membrane, including membrane-cytoskeleton interactions, phospholipase inhibition, anticoagulation, and membrane fusion. Annexins contain four to eight repeated segments of about 60 residues. Each repeat folds into five alpha helices wound into a right-handed superhelix. Signaling Complex Protein Domains
PDZ domains were named for three proteins in which this domain was initially discovered. These proteins include PSD-95 (postsynaptic density 95), Dig (Drosophila lethal(l)discs large-1), and ZO-1 (zonula occludens-1). These proteins play important roles in neuronal synaptic transmission, tumor suppression, and cell junction formation, respectively. Since the discovery of these proteins, over sixty additional PDZ-containing proteins have been identified in diverse prokaryotic and eukaryotic organisms. This domain has been implicated in receptor and ion channel clustering and in the targeting of multiprotein signaling complexes to specialized functional regions of the cytosolic face of the plasma membrane. (For a review of PDZ domain-containing proteins, see Ponting, CP. et al. (1997) Bioessays 19:469-479.) A large proportion of PDZ domains are found in the eukaryotic MAGUK (membrane-associated guanylate kinase) protein family, members of which bind to the intracellular domains of receptors and channels. However, PDZ domains are also found in diverse membrane-localized proteins such as protein tyrosine phosphatases, serine/threonine kinases, G-protein cofactors, and synapse-associated proteins such as syntrophins and neuronal nitric oxide synthase (nNOS). Generally, about one to three PDZ domains are found in a given protein, although up to nine PDZ domains have been identified in a single protein. The glutamate receptor interacting protein (GRIP) contains seven PDZ domains. GRIP is an adaptor that links certain glutamate receptors to other proteins and may be responsible for the clustering of these receptors at excitatory synapses in the brain (Dong, H. et al. (1997) Nature 386:279-284). The Drosophila scribble (SCRIB) protein contains both multiple PDZ domains and leucine-rich repeats. SCRIB is located at the epithelial septate junction, which is analogous to the vertebrate tight junction, at the boundary of the apical and basolateral cell surface. SCRIB is involved in the distribution of apical proteins and correct placement of adherens junctions to the basolateral cell surface (Bilder, D. and N. Perrimon (2000) Nature 403:676-680). The PX domain is an example of a domain specialized for promoting protein-protein interactions. The PX domain is found in sorting nexins and in a variety of other proteins, including the PhoX components of NADPH oxidase and the Cpk class of phosphatidylinositol 3-kinase. Most PX domains contain a polyproline motif which is characteristic of SH3 domain-binding proteins (Ponting, CP. (1996) Protein Sci. 5:2353-2357). SH3 domain-mediated interactions involving the PhoX components of NADPH oxidase play a role in the formation of the NADPH oxidase multi-protein complex (Leto, T.L. et al. (1994) Proc. Natl. Acad. Sci. USA 91:10650-10654; Wilson, L. et al. (1997) Inflamm. Res. 46:265-271).
The SH3 domain is defined by homology to a region of the proto-oncogene c-Src, a cytoplasmic protein tyrosine kinase. SH3 is a small domain of 50 to 60 amino acids that interacts with proline-rich ligands. SH3 domains are found in a variety of eukaryotic proteins involved in signal transduction, cell polarization, and membrane-cytoskeleton interactions. In some cases, SH3 domain- containing proteins interact directly with receptor tyrosine kinases. For example, the SLAP-130 protein is a substrate of the T-cell receptor (TCR) stimulated protein kinase. SLAP-130 interacts via its SH3 domain with the protein SLP-76 to affect the TCR-induced expression of interleukin-2 (Musci, M. A. et al. (1997) J. Biol. Chem. 272: 11674-11677). Another recently identified SH3 domain protein is macrophage actin-associated tyrosine-phosphorylated protein (MAYP) which is phosphorylated during the response of macrophages to colony stimulating factor- 1 (CSF-1) and is likely to play a role in regulating the CSF-1-induced reorganization of the actin cytoskeleton (Yeung, Y.-G. et al. (1998) J. Biol. Chem. 273:30638-30642). The structure of the SH3 domain is characterized by two antiparallel beta sheets packed against each other at right angles. This packing forms a hydrophobic pocket lined with residues that are highly conserved between different SH3 domains. This pocket makes critical hydrophobic contacts with proline residues in the ligand (Feng, S. et al. (1994) Science 266:1241- 1247). A novel domain, called the WW domain, resembles the SH3 domain in its ability to bind proline-rich ligands. This domain was originally discovered in dystrophin, a cytoskeletal protein with direct involvement in Duchenne muscular dystrophy (Bork, P. and M. Sudol (1994) Trends Biochem. Sci. 19:531-533). WW domains have since been discovered in a variety of intracellular signaling molecules involved in development, cell differentiation, and cell proliferation. The structure of the WW domain is composed of beta strands grouped around four conserved aromatic residues, generally tryptophan.
Like SH3, the SH2 domain is defined by homology to a region of c-Src. SH2 domains interact directly with phospho-tyrosine residues, thus providing an immediate mechanism for the regulation and transduction of receptor tyrosine kinase-mediated signaling pathways. For example, as many as ten distinct SH2 domains are capable of binding to phosphorylated tyrosine residues in the activated PDGF receptor, thereby providing a highly coordinated and finely tuned response to ligand-mediated receptor activation. (Reviewed in Schaffhausen, B. (1995) Biochim. Biophys. Acta. 1242:61-75.)
The GSG domain (GRP33, Sam68, GLD-1) and the KH domain (an RNA binding domain), are found within Sam68, a 68-kDa Src substrate associated during mitosis protein, which is an RNA- binding protein with signaling properties. It is known to be a substrate for Src-family tyrosine kinases during mitosis and associates with various SH3 and SH2 domain-containing signaling molecules. SLM-1 and SLM-2 (Sam68-like mammalian) proteins have sequence identity with Sam68, also contain the GSG domain, have proline-rich motifs, arginine-gylcine repeats, and a C-terminal tyrosine-rich region. SLM-1 is a Src substrate during mitosis, suggesting a possible involvement in the steps of mitosis. It has been suggested by Di Fruscio et al. that Sam68/SLM defines a family in which the members have the potential to link tyrosine kinase signaling cascades with some aspects of RNA metabolism, possibly as multifunctional adapter proteins during mitosis (Di Fruscio, M. et al. (1999) Proc. Natl. Acad. Sci. U.S.A. 96:2710-2715.) The pleckstrin homology (PH) domain was originally identified in pleckstrin, the predominant substrate for protein kinase C in platelets. Since its discovery, this domain has been identified in over 90 proteins involved in intracellular signaling or cytoskeletal organization. Proteins containing the pleckstrin homology domain include a variety of kinases, phospholipase-C isoforms, guanine nucleotide release factors, and GTPase activating proteins. For example, members of the FGD1 family contain both Rho-guanine nucleotide exchange factor (GEF) and PH domains, as well as a FYVE zinc finger domain. FGD1 is the gene responsible for faciogenital dysplasia, an inherited skeletal dysplasia (Pasteris, N.G. and J.L. Gorski (1999) Genomics 60:57-66). Many PH domain proteins function in association with the plasma membrane, and this association appears to be mediated by the PH domain itself. PH domains share a common structure composed of two antiparallel beta sheets flanked by an amphipathic alpha helix. Variable loops connecting the component beta strands generally occur within a positively charged environment and may function as ligand binding sites (Lemmon, M.A. et al. (1996) Cell 85:621-624). Ankyrin (ANK) repeats mediate protein-protein interactions associated with diverse intracellular signaling functions. For example, ANK repeats are found in proteins involved in cell proliferation such as kinases, kinase inhibitors, tumor suppressors, and cell cycle control proteins. (See, for example, Kalus, W. et al. (1997) FEBS Lett. 401 : 127-132; Ferrante, A.W. et al. (1995) Proc. Natl. Acad. Sci. USA 92:1911-1915.) These proteins generally contain multiple ANK repeats, each composed of about 33 amino acids. Myotrophin is an ANK repeat protein that plays a key role in the development of cardiac hypertrophy, a contributing factor to many heart diseases. Structural studies show that the myotrophin ANK repeats, like other ANK repeats, each form a helix-turn-helix core preceded by a protruding "tip." These tips are of variable sequence and may play a role in protein- protein interactions. The helix-turn-helix region of the ANK repeats stack on top of one another and are stabilized by hydrophobic interactions (Yang, Y. et al. (1998) Structure 6:619-626).
The tetratricopeptide repeat (TPR) is a 34 amino acid repeated motif found in organisms from bacteria to humans. TPRs are predicted to form ampipathic helices, and appear to mediate protein- protein interactions. TPR domains are found in CDC16, CDC23, and CDC27, members of the the anaphase promoting complex which targets proteins for degradation at the onset of anaphase. Other processes involving TPR proteins include cell cycle control, transcription repression, stress response, and protein kinase inhibition (Lamb, J.R. et al. (1995) Trends Biochem. Sci. 20:257-259). The armadillo/beta-catenin repeat is a 42 amino acid motif which forms a superhelix of alpha helices when tandemly repeated. The structure of the armadillo repeat region from beta-catenin revealed a shallow groove of positive charge on one face of the superhelix, which is a potential binding surface. The armadillo repeats of beta-catenin, plakoglobin, and pl20cas bind the cytoplasmic domains of cadherins. Beta-catenin/cadherin complexes are targets of regulatory signals that govern cell adhesion and mobility (Huber, A.H. et al. (1997) Cell 90:871-882).
Eight tandem repeats of about 40 residues (WD-40 repeats), each containing a central Trp-Asp motif, make up beta-transducin (G-beta), which is one of the three subunits (alpha, beta, and gamma) of the guanine nucleotide-binding proteins (G proteins). In higher eukaryotes G-beta exists as a small multigene family of highly conserved proteins of about 340 amino acid residues. The discovery of new intracellular signaling molecules, and the polynucleotides encoding them, satisfies a need in the art by providing new compositions which are useful in the diagnosis, prevention, and treatment of cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of intracellular signaling molecules.
SUMMARY OF THE INVENTION
The invention features purified polypeptides, intracellular signaling molecules, referred to collectively as "INTSIG" and individually as "INTSIG-1," 'TNTSIG-2," 'TNTSIG-3," 'TNTSIG-4," "ΓNTSIG-5," "ΓNTSIG-6," "ΓNTSIG-7," "ΓNTSIG-8," "ΓNTSIG-9," "ΓNTSIG-IO," "ΓNTSIG-Π," 'TNTSIG-12," "INTSIG-13," "INTSIG-14," "INTSIG-15," 'TNTSIG-16," "INTSIG-17," 'TNTSIG-
18," "INTSIG-19," and 'TNTSIG-20." In one aspect, the invention provides an isolated polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. In one alternative, the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 1-20.
The invention further provides an isolated polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO:l- 20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ D3 NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO:1-20. In one alternative, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ DD NO: 1-20. In another alternative, the polynucleotide is selected from the group consisting of SEQ ID NO:21-40. Additionally, the invention provides a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ED NO: 1-20. In one alternative, the invention provides a cell transformed with the recombinant polynucleotide. In another alternative, the invention provides a transgenic organism comprising the recombinant polynucleotide.
The invention also provides a method for producing a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The method comprises a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recovering the polypeptide so expressed.
Additionally, the invention provides an isolated antibody which specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The invention further provides an isolated polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). In one alternative, the polynucleotide comprises at least 60 contiguous nucleotides.
Additionally, the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide. having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises 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. In one alternative, the probe comprises at least 60 contiguous nucleotides. The invention further provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO:21-40, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises 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. The invention further provides a composition comprising an effective amount of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and a pharmaceutically acceptable excipient. In one embodiment, the composition comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The invention additionally provides a method of treating a disease or condition associated with decreased expression of functional INTSIG, comprising administering to a patient in need of such treatment the composition.
The invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample. In one alternative, the invention provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with decreased expression of functional INTSIG, comprising administering to a patient in need of such treatment the composition. Additionally, the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample. In one alternative, the invention provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with overexpression of functional INTSIG, comprising administering to a patient in need of such treatment the composition.
The invention further provides a method of screening for a compound that specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20. The method comprises a) combining the polypeptide with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide to the test compound, thereby identifying a compound that specifically binds to the polypeptide.
The invention further provides a method of screening for a compound that modulates the activity of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ DD NO: 1-20. The method comprises a) combining the polypeptide with at least one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.
The invention further provides a method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, the method comprising a) exposing a sample comprising the target polynucleotide to a compound, and b) detecting altered expression of the target polynucleotide.
The invention further provides a method for assessing toxicity of a test compound, said 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 selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, iii) a polynucleotide having a sequence complementary to i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Hybridization occurs under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ DD NO:21-40, iii) a polynucleotide complementary to the polynucleotide of i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Alternatively, the target polynucleotide comprises a fragment of a polynucleotide sequence selected from the group consisting of i)-v) above; 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.
BRIEF DESCRIPTION OF THE TABLES
Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the present invention.
Table 2 shows the GenBank identification number and annotation of the nearest GenBank homolog for polypeptides of the invention. The probability score for the match between each polypeptide and its GenBank homolog is also shown.
Table 3 shows structural features of polypeptide sequences of the invention, including predicted motifs and domains, along with the methods, algorithms, and searchable databases used for analysis of the polypeptides.
Table 4 lists the cDNA and/or genomic DNA fragments which were used to assemble polynucleotide sequences of the invention, along with selected fragments of the polynucleotide sequences.
Table 5 shows the representative cDNA library for polynucleotides of the invention.
Table 6 provides an appendix which describes the tissues and vectors used for construction of the cDNA libraries shown in Table 5. Table 7 shows the tools, programs, and algorithms used to analyze the polynucleotides and polypeptides of the invention, along with applicable descriptions, references, and threshold parameters.
DESCRIPTION OF THE INVENTION Before the present proteins, nucleotide sequences, and methods are described, it is understood that this invention is not limited to the particular machines, materials and methods described, as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. It must be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include 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. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any machines, materials, and methods similar or equivalent to those described herein can be used to practice or test the present invention, the preferred machines, materials and methods are now described. All publications mentioned herein are cited for the purpose of describing and disclosing the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention. DEFINITIONS
"INTSIG" refers to the amino acid sequences of substantially purified INTSIG obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, murine, equine, and human, and from any source, whether natural, synthetic, semi-synthetic, or recombinant.
The term "agonist" refers to a molecule which intensifies or mimics the biological activity of INTSIG. Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of INTSIG either by directly interacting with INTSIG or by acting on components of the biological pathway in which INTSIG participates.
An "allelic variant" is an alternative form of the gene encoding INTSIG. Allelic variants may result from at least one mutation in the nucleic acid sequence and may result in altered mRNAs or in polypeptides whose structure or function may or may not be altered. A gene may have none, one, or many allelic variants of its naturally occurring form. Common mutational changes which give rise to allelic variants are generally ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence.
"Altered" nucleic acid sequences encoding INTSIG include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as INTSIG or a polypeptide with at least one functional characteristic of INTSIG. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding INTSIG, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding INTSIG. The encoded protein may also be "altered," and may contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent INTSIG. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of INTSIG is retained. For example, negatively charged amino acids may include aspartic acid and glutamic acid, and positively charged amino acids may include lysine and arginine. Amino acids with uncharged polar side chains having similar hydrophilicity values may ' include: asparagine and glutamine; and serine and threonine. Amino acids with uncharged side chains having similar hydrophilicity values may include: leucine, isoleucine, and valine; glycine and alanine; and phenylalanine and tyrosine. The terms "amino acid" and "amino acid sequence" refer to an oligopeptide, peptide, polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. Where "amino acid sequence" is recited to refer to a sequence of a naturally occurring protein molecule, "amino acid sequence" and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule. "Amplification" relates to the production of additional copies of a nucleic acid sequence.
Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art.
The term "antagonist" refers to a molecule which inhibits or attenuates the biological activity of INTSIG. Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of rNTSIG either by directly interacting with INTSIG or by acting on components of the biological pathway in which INTSIG participates.
The term "antibody" refers to intact immunoglobulin molecules as well as to fragments thereof, such as Fab, F(ab')2, and Fv fragments, which are capable of binding an epitopic determinant. Antibodies that bind INTSIG polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or oligopeptide used to immunize an animal (e.g., a mouse, a rat, or a rabbit) can be derived from the translation of RNA, or synthesized chemically, and can be conjugated to a carrier protein if desired. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal.
The term "antigenic determinant" refers to that region of a molecule (i.e., an epitope) that makes contact with a particular antibody. When a protein or a fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to antigenic determinants (particular regions or three-dimensional structures on the protein). An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody.
The term "aptamer" refers to a nucleic acid or oligonucleotide molecule that binds to a specific molecular target. Aptamers are derived from an in vitro evolutionary process (e.g., SELEX (Systematic Evolution of Ligands by Exponential Enrichment), described in U.S. Patent No. 5,270,163), which selects for target-specific aptamer sequences from large combinatorial libraries. Aptamer compositions may be double-stranded or single-stranded, and may include deoxyribonucleotides, ribonucleotides, nucleotide derivatives, or other nucleotide-like molecules. The nucleotide components of an aptamer may have modified sugar groups (e.g., the 2'-OH group of a ribonucleotide may be replaced by 2'-F or 2'-NH2), which may improve a desired property, e.g., resistance to nucleases or longer lifetime in blood. Aptamers may be conjugated to other molecules, e.g., a high molecular weight carrier to slow clearance of the aptamer from the circulatory system. Aptamers may be specifically cross-linked to their cognate ligands, e.g., by photo-activation of a cross-linker. (See, e.g., Brody, E.N. and L. Gold (2000) J. Biotechnol. 74:5-13.)
The term "intramer" refers to an aptamer which is expressed in vivo. For example, a vaccinia virus-based RNA expression system has been used to express specific RNA aptamers at high levels in the cytoplasm of leukocytes (Blind, M. et al. (1999) Proc. Natl Acad. Sci. USA 96:3606-3610). The term "spiegelmer" refers to an aptamer which includes L-DNA, L-RNA, or other left- handed nucleotide derivatives or nucleotide-like molecules. Aptamers containing left-handed nucleotides are resistant to degradation by naturally occurring enzymes, which normally act on substrates containing right-handed nucleotides.
The term "antisense" refers to any composition capable of base-pairing with the "sense" (coding) strand of a specific nucleic acid sequence. Antisense compositions may include DNA; RNA; peptide nucleic acid (PNA); oligonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; oligonucleotides having modified sugar groups such as 2-methoxyethyl sugars or 2'-methoxyethoxy sugars; or oligonucleotides having modified bases such as 5-methyl cytosine, 2'-deoxyuracil, or 7-deaza-2'-deoxyguanosine. Antisense molecules may be produced by any method including chemical synthesis or transcription. Once introduced into a cell, the complementary antisense molecule base-pairs with a naturally occurring nucleic acid sequence produced by the cell to form duplexes which block either transcription or translation. The designation "negative" or "minus" can refer to the antisense strand, and the designation "positive" or "plus" can refer to the sense strand of a reference DNA molecule. The term "biologically active" refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule. Likewise, "immunologically active" or "immunogenic" refers to the capability of the natural, recombinant, or synthetic INTSIG, or of any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies. "Complementary" describes the relationship between two single-stranded nucleic acid sequences that anneal by base-pairing. For example, 5'-AGT-3' pairs with its complement, 3'-TCA-5'.
A "composition comprising a given polynucleotide sequence" and a "composition comprising a given amino acid sequence" refer broadly to any composition containing the given polynucleotide or amino acid sequence. The composition may comprise a dry formulation or an aqueous solution.
Compositions comprising polynucleotide sequences encoding INTSIG or fragments of INTSIG may be employed as hybridization probes. The probes may be stored in freeze-dried form and may be associated with a stabilizing agent such as a carbohydrate. In hybridizations, the probe may be deployed in an aqueous solution containing salts (e.g., NaCl), detergents (e.g., sodium dodecyl sulfate; SDS), and other components (e.g., Denhardt's solution, dry milk, salmon sperm DNA, etc.). "Consensus sequence" refers to a nucleic acid sequence which has been subjected to repeated DNA sequence analysis to resolve uncalled bases, extended using the XL-PCR kit (Applied Biosystems, Foster City CA) in the 5' and/or the 3' direction, and resequenced, or which has been assembled from one or more overlapping cDNA, EST, or genomic DNA fragments using a computer program for fragment assembly, such as the GELVIEW fragment assembly system (GCG, Madison WI) or Phrap (University of Washington, Seattle WA). Some sequences have been both extended and assembled to produce the consensus sequence.
"Conservative amino acid substitutions" are those substitutions that are predicted to least interfere with the properties of the original protein, i.e., the structure and especially the function of the protein is conserved and not significantly changed by such substitutions. The table below shows amino acids which may be substituted for an original amino acid in a protein and which are regarded as conservative amino acid substitutions. Original Residue Conservative Substitution Ala Gly, Ser
Arg His, Lys
Asn Asp, Gin, His
Asp Asn, Glu
Cys Ala, Ser Gin Asn, Glu, His
Glu Asp, Gin, His
Gly Ala
His Asn, Arg, Gin, Glu
He Leu, Val Leu He, Val
Lys Arg, Gin, Glu
Met Leu, He
Phe His, Met, Leu, Trp, Tyr
Ser Cys, Thr Thr Ser, Val
Trp Phe, Tyr
Tyr His, Phe, Trp VaT lie, Leu, Thr
Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.
A "deletion" refers to a change in the amino acid or nucleotide sequence that results in the absence of one or more amino acid residues or nucleotides.
The term "derivative" refers to a chemically modified polynucleotide or polypeptide. Chemical modifications of a polynucleotide can include, for example, replacement of hydrogen by an alkyl, acyl, hydroxyl, or amino group. A derivative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule. A derivative polypeptide is one modified by glycosylation, pegylation, or any similar process that retains at least one biological or immunological function of the polypeptide from which it was derived.
A "detectable label" refers to a reporter molecule or enzyme that is capable of generating a measurable signal and is covalently or noncovalently joined to a polynucleotide or polypeptide.
"Differential expression" refers to increased or upregulated; or decreased, downregulated, or absent gene or protein expression, determined by comparing at least two different samples. Such comparisons may be carried out between, for example, a treated and an untreated sample, or a diseased and a normal sample.
"Exon shuffling" refers to the recombination of different coding regions (exons). Since an exon may represent a structural or functional domain of the encoded protein, new proteins may be assembled through the novel reassortment of stable substructures,°thus allowing acceleration of the evolution of new protein functions.
A "fragment" is a unique portion of INTSIG or the polynucleotide encoding INTSIG which is identical in sequence to but shorter in length than the parent sequence. A fragment may comprise up to the entire length of the defined sequence, minus one nucleotide/amino acid residue. For example, a fragment may comprise from 5 to 1000 contiguous nucleotides or amino acid residues. A fragment used as a probe, primer, antigen, therapeutic molecule, or for other purposes, may be at least 5, 10, 15, 16, 20, 25, 30, 40, 50, 60, 75, 100, 150, 250 or at least 500 contiguous nucleotides or amino acid residues in length. Fragments may be preferentially selected from certain regions of a molecule. For example, a polypeptide fragment may comprise a certain length of contiguous amino acids selected from the first 250 or 500 amino acids (or first 25% or 50%) of a polypeptide as shown in a certain defined sequence. Clearly these lengths are exemplary, and any length that is supported by the specification, including the Sequence Listing, tables, and figures, may be encompassed by the present embodiments.
A fragment of SEQ DD NO :21-40 comprises a region of unique polynucleotide sequence that specifically identifies SEQ DD NO:21-40, for example, as distinct from any other sequence in the genome from which the fragment was obtained. A fragment of SEQ DD NO:21-40 is useful, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ DD NO:21-40 from related polynucleotide sequences. The precise length of a fragment of SEQ DD NO:21-40 and the region of SEQ DD NO:21-40 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment. A fragment of SEQ DD NO: 1-20 is encoded by a fragment of SEQ DD NO:21-40. A fragment of SEQ DD NO: 1-20 comprises a region of unique amino acid sequence that specifically identifies SEQ DD NO: 1-20. For example, a fragment of SEQ DD NO: 1-20 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ DD NO: 1-20. The precise length of a fragment of SEQ DD NO: 1-20 and the region of SEQ DD NO: 1-20 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.
A "full length" polynucleotide sequence is one containing at least a translation initiation codon (e.g., methionine) followed by an open reading frame and a translation termination codon. A "full length" polynucleotide sequence encodes a "full length" polypeptide sequence. "Homology" refers to sequence similarity or, interchangeably, sequence identity, between two or more polynucleotide sequences or two or more polypeptide sequences.
The terms "percent identity" and "% identity," as applied to polynucleotide sequences, refer to the percentage of residue matches between at least two polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
Percent identity between polynucleotide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN version 3.12e sequence alignment program. This program is part of the LASERGENE software package, a suite of molecular biological analysis programs (DNASTAR, Madison WI). CLUSTAL V is described in Higgins, D.G. and P.M. Sharp (1989) CABIOS 5:151-153 and in Higgins, D.G. et al. (1992) CABIOS 8:189-191. For pairwise alignments of polynucleotide sequences, the default parameters are set as follows: Ktuple=2, gap penalty=5, window=4, and "diagonals saved"=4. The "weighted" residue weight table is selected as the default. Percent identity is reported by CLUSTAL V as the "percent similarity" between aligned polynucleotide sequences.
Alternatively, a suite of commonly used and freely available sequence comparison algorithms is provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) (Altschul, S.F. et al. (1990) J. Mol. Biol. 215:403-410), which is available from several sources, including the NCBI, Bethesda, MD, and on the Internet at http://www.ncbi.nlm.nih.gov/BLAST/. The BLAST software suite includes various sequence analysis programs including "blastn," that is used to align a known polynucleotide sequence with other polynucleotide sequences from a variety of databases. Also available is a tool called "BLAST 2 Sequences" that is used for direct pairwise comparison of two nucleotide sequences. "BLAST 2 Sequences" can be accessed and used interactively at http://www.ncbi.nlm.nih.gov/gorf/bl2.html. The "BLAST 2 Sequences" tool can be used for both blastn and blastp (discussed below). BLAST programs are commonly used with gap and other parameters set to default settings. For example, to compare two nucleotide sequences, one may use blastn with the "BLAST 2 Sequences" tool Version 2.0.12 (April-21-2000) set at default parameters. Such default parameters may be, for example: 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
Filter: on
Percent identity may be measured over the length of an entire defined sequence, for example, as defined by a particular SEQ DD number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined sequence, for instance, a fragment of at least 20, at least 30, at least 40, at least 50, at least 70, at least 100, or at least 200 contiguous nucleotides. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures, or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
Nucleic acid sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein. The phrases "percent identity" and "% identity," as applied to polypeptide sequences, refer to the percentage of residue matches between at least two polypeptide sequences aligned using a standardized algorithm. Methods of polypeptide sequence alignment are well-known. Some alignment methods take into account conservative amino acid substitutions. Such conservative substitutions, explained in more detail above, generally preserve the charge and_hydrophobicity at the site of substitution, thus preserving the structure (and therefore function) of the polypeptide.
Percent identity between polypeptide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN version 3.12e sequence alignment program (described and referenced above). For pairwise alignments of polypeptide sequences using CLUSTAL V, the default parameters are set as follows: Ktuple=l, gap penalty=3, window=5, and "diagonals saved"=5. The PAM250 matrix is selected as the default residue weight table. As with polynucleotide alignments, the percent identity is reported by CLUSTAL V as the "percent similarity" between aligned polypeptide sequence pairs.
Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise comparison of two polypeptide sequences, one may use the "BLAST 2 Sequences" tool Version 2.0.12 (April-21-2000) with blastp set at default parameters. Such default parameters may be, for example:
Matrix: BLOSUM62
Open Gap: 11 and Extension Gap: 1 penalties
Gap x drop-off: 50 Expect: 10
Word Size: 3
Filter: on
Percent identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ DD number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
"Human artificial chromosomes" (HACs) are linear rmcrocrrromosomes which may contain DNA sequences of about 6 kb to 10 Mb in size and which contain all of the elements required for chromosome replication, segregation and maintenance.
The term "humanized antibody" refers to an antibody molecule in which the amino acid sequence in the non-antigen binding regions has been altered so that the antibody more closely resembles a human antibody, and still retains its original binding ability.
"Hybridization" refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of complementarity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after the "washing" step(s). The washing step(s) is particularly important in determining the stringency of the hybridization process, with more stringent conditions allowing less non-specific binding, i.e., binding between pairs of nucleic acid strands that are not perfectly matched. Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve the desired stringency, and therefore hybridization specificity. Permissive annealing conditions occur, for example, at 68°C in the presence of about 6 x SSC, about 1% (w/v) SDS, and about 100 μg/ml sheared, denatured salmon sperm DNA.
Generally, stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thennal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. An equation for calculating Tm and conditions for nucleic acid hybridization are well known and can be found in Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual. 2nd ed., vol. 1-3, Cold Spring Harbor Press, Plainview NY; specifically see volume 2, chapter 9.
High stringency conditions for hybridization between polynucleotides of the present invention include wash conditions of 68°C in the presence of about 0.2 x SSC and about 0.1% SDS, for 1 hour. Alternatively, temperatures of about 65°C, 60°C, 55°C, or 42°C may be used. SSC concentration may be varied from about 0.1 to 2 x SSC, with SDS being present at about 0.1%. Typically, blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 μg/ml. Organic solvent, such as formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for RNA:DNA hybridizations. Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art. Hybridization, particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is strongly indicative of a similar role for the nucleotides and their encoded polypeptides.
The term "hybridization complex" refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (e.g., C0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e.g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed). The words "insertion" and "addition" refer to changes in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively. "Immune response" can refer to conditions associated with inflammation, trauma, immune disorders, or infectious or genetic disease, etc. These conditions can be characterized by expression of various factors, e.g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems.
An "immunogenic fragment" is a polypeptide or oligopeptide fragment of INTSIG which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal. The term "immunogenic fragment" also includes any polypeptide or oligopeptide fragment of INTSIG which is useful in any of the antibody production methods disclosed herein or known in the art.
The term "microarray" refers to an arrangement of a plurality of polynucleotides, polypeptides, or other chemical compounds on a substrate.
The terms "element" and "array element" refer to a polynucleotide, polypeptide, or other chemical compound having a unique and defined position on a microarray. The term "modulate" refers to a change in the activity of INTSIG. For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of INTSIG.
The phrases "nucleic acid" and "nucleic acid sequence" refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof. These phrases also refer to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA), or to any DNA-like or RNA-like material.
"Operably linked" refers to the situation in which a first nucleic acid sequence is placed in a functional relationship with a second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Operably linked DNA sequences may be in close proximity or contiguous and, where necessary to join two protein coding regions, in the same reading frame.
"Peptide nucleic acid" (PNA) refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition. PNAs preferentially bind complementary single stranded DNA or RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell.
"Post-translational modification" of an INTSIG may involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and other modifications known in the art. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cell type depending on the enzymatic milieu of INTSIG.
"Probe" refers to nucleic acid sequences encoding INTSIG, their complements, or fragments thereof, which are used to detect identical, allelic or related nucleic acid sequences. Probes are isolated oligonucleotides or polynucleotides attached to a detectable label or reporter molecule. Typical labels include radioactive isotopes, ligands, chemiluminescent agents, and enzymes. "Primers" are short nucleic acids, usually DNA oligonucleotides, which may be annealed to a target polynucleotide by complementary base-pairing. The primer may then be extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification (and identification) of a nucleic acid sequence, e.g., by the polymerase chain reaction (PCR). Probes and primers as used in the present invention typically comprise at least 15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and primers may also be employed, such as probes and primers that comprise at least 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, or at least 150 consecutive nucleotides of the disclosed nucleic acid sequences. Probes and primers may be considerably longer than these examples, and it is understood that any length supported by the specification, including the tables, figures, and Sequence Listing, may be used.
Methods for preparing and using probes and primers are described in the references, for example Sambrook, J. et al. (1989) Molecular Cloning: A Laboratory Manual. 2nd ed., vol. 1-3, Cold Spring Harbor Press, Plainview NY; Ausubel, F.M. et al. (1987) Current Protocols in Molecular Biology. Greene Publ. Assoc. & Wiley-Intersciences, New York NY; Innis, M. et al. (1990) PCR Protocols. A Guide to Methods and Applications. Academic Press, San Diego CA. PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5, 1991, Whitehead Institute for Biomedical Research, Cambridge MA). Oligonucleotides for use as primers are selected using software known in the art for such purpose. For example, OLIGO 4.06 software is useful for the selection of PCR primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases. Similar primer selection programs have incorporated additional features for expanded capabilities. For example, the PrimOU primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas TX) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope. The Primer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambridge MA) allows the user to input a "misprinting library," in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarray s. (The source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.) The PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences. Hence, this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments. The oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of oligonucleotide selection are not limited to those described above.
A "recombinant nucleic acid" is a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two or more otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques such as those described in Sambrook, supra. The term recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion of a portion of the nucleic acid. Frequently, a recombinant nucleic acid may include a nucleic acid sequence operably linked to a promoter sequence. Such a recombinant nucleic acid may be part of a vector that is used, for example, to transform a cell. Alternatively, such recombinant nucleic acids may be part of a viral vector, e.g., based on a vaccinia virus, that could be use to vaccinate a mammal wherein the recombinant nucleic acid is expressed, inducing a protective immunological response in the mammal.
A "regulatory element" refers to a nucleic acid sequence usually derived from untranslated regions of a gene and includes enhancers, promoters, introns, and 5' and 3' untranslated regions (UTRs). Regulatory elements interact with host or viral proteins which control transcription, translation, or RNA stability.
"Reporter molecules" are chemical or biochemical moieties used for labeling a nucleic acid, amino acid, or antibody. Reporter molecules include radionuclides; enzymes; fluorescent, chemiluminescent, or chromogenic agents; substrates; cofactors; inhibitors; magnetic particles; and other moieties known in the art.
An "RNA equivalent," in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
The term "sample" is used in its broadest sense. A sample suspected of containing INTSIG, nucleic acids encoding INTSIG, or fragments thereof may comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; a cell; genomic DNA, RNA, or cDNA, in solution or bound to a substrate; a tissue; a tissue print; etc. The terms "specific binding" and "specifically binding" refer to that interaction between a protein or peptide and an agonist, an antibody, an antagonist, a small molecule, or any natural or synthetic binding composition. The interaction is dependent upon the presence of a particular structure of the protein, e.g., the antigenic determinant or epitope, recognized by the binding molecule. For example, if an antibody is specific for epitope "A," the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody.
The term "substantially purified" refers to nucleic acid or amino acid sequences that are removed from their natural environment and are isolated or separated, and are at least 60% free, preferably at least 75% free, and most preferably at least 90% free from other components with which they are naturally associated.
A "substitution" refers to the replacement of one or more amino acid residues or nucleotides by different amino acid residues or nucleotides, respectively.
"Substrate" refers to any suitable rigid or semi-rigid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillaries. The substrate can have a variety of surface forms, such as wells, trenches, pins, channels and pores, to which polynucleotides or polypeptides are bound.
A "transcript image" refers to the collective pattern of gene expression by a particular cell type or tissue under given conditions at a given time. "Transformation" describes a process by which exogenous DNA is introduced into a recipient cell. Transformation may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method for transformation is selected based on the type of host cell being transformed and may include, but is not limited to, bacteriophage or viral infection, electroporation, heat shock, lipofection, and particle bombardment. The term "transformed cells" includes stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome, as well as transiently transformed cells which express the inserted DNA or RNA for limited periods of time.
A "transgenic organism," as used herein, is any organism, including but not limited to animals and plants, in which one or more of the cells of the organism contains heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. The term genetic manipulation does not include classical cross-breeding, or in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. The transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals. The isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation. Techniques for transferring the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook et al. (1989), supra.
A "variant" of a particular nucleic acid sequence is defined as a nucleic acid sequence having at least 40% sequence identity to the particular nucleic acid sequence over a certain length of one of the nucleic acid sequences using blastn with the "BLAST 2 Sequences" tool Version 2.0.9 (May-07- 1999) set at default parameters. Such a pair of nucleic acids may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at'least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length. A variant may be described as, for example, an "allelic" (as defined above), "splice," "species," or "polymorphic" variant. A splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternate splicing of exons during mRNA processing. The corresponding polypeptide may possess additional functional domains or lack domains that are present in the reference molecule. Species variants are polynucleotide sequences that vary from one species to another. The resulting polypeptides will generally have significant amino acid identity relative to each other. A polymorphic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species. Polymorphic variants also may encompass "single nucleotide polymorphisms" (SNPs) in which the polynucleotide sequence varies by one nucleotide base. The presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state. A "variant" of a particular polypeptide sequence is defined as a polypeptide sequence having at least 40% sequence identity to the particular polypeptide sequence over a certain length of one of the polypeptide sequences using blastp with the "BLAST 2 Sequences" tool Version 2.0.9 (May-07- 1999) set at default parameters. Such a pair of polypeptides may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length of one of the polypeptides.
THE INVENTION
The invention is based on the discovery of new human intracellular signaling molecules (INTSIG), the polynucleotides encoding INTSIG, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders.
Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the invention. Each polynucleotide and its corresponding polypeptide are correlated to a single Incyte project identification number (Incyte Project DD). Each polypeptide sequence is denoted by both a polypeptide sequence identification number (Polypeptide SEQ DD NO:) and an Incyte polypeptide sequence number (Incyte Polypeptide DD) as shown. Each polynucleotide sequence is denoted by both a polynucleotide sequence identification number (Polynucleotide SEQ DD NO:) and an Incyte polynucleotide consensus sequence number (Incyte Polynucleotide DD) as shown. Table 2 shows sequences with homology to the polypeptides of the invention as identified by BLAST analysis against the GenBank protein (genpept) database. Columns 1 and 2 show the polypeptide sequence identification number (Polypeptide SEQ DD NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide DD) for polypeptides of the invention. Column 3 shows the GenBank identification number (Genbank DD NO:) of the nearest GenBank homolog. Column 4 shows the probability score for the match between each polypeptide and its GenBank homolog. Column 5 shows the annotation of the GenBank homolog along with relevant citations where applicable, all of which are expressly incorporated by reference herein.
Table 3 shows various structural features of the polypeptides of the invention. Columns 1 and 2 show the polypeptide sequence identification number (SEQ DD NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide DD) for each polypeptide of the invention. Column 3 shows the number of amino acid residues in each polypeptide. Column 4 shows potential phosphorylation sites, and column 5 shows potential glycosylation sites, as determined by the MOTIF'S program of the GCG sequence analysis software package (Genetics Computer Group, Madison WI). Column 6 shows amino acid residues comprising signature sequences, domains, and motifs. Column 7 shows analytical methods for protein structure/function analysis and in some cases, searchable databases to which the analytical methods were applied.
Together, Tables 2 and 3 summarize the properties of polypeptides of the invention, and these properties establish that the claimed polypeptides are intracellular signaling molecules. For example, SEQ DD NO:2 is 37% identical to Schizosaccharomyces pombe beta transducin (GenBank DD g3451308) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 2.1e-146, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:2 also contains a G-beta repeat WD40 domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from MOTIFS analysis provides further corroborative evidence that SEQ DD NO:2 is a transducin.
In an alternative example, SEQ DD NO:6 is 85% identical to murine nedd-1 protein (GenBank DD g286103) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 0.0, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:6 also contains a WD domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS and MOTTFS analyses provide further corroborative evidence that SEQ DD NO: 6 is a protein involved in signal transduction. In an alternative example, SEQ ID NO: 10 is 51% identical to the human rho GTPase activating protein pi 15 (GenBank DD g840786) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 5.2e-211, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO: 10 contains a rhoGAP domain, an SH3 domain, and a Fes/CIP4 actin cytoskeleton regulatory protein domain, as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) The presence of these domains is confirmed by BLIMPS and MOTTFS analyses, providing further corroborative evidence that SEQ DD NO: 10 is a GTPase activating protein. In an alternative example, SEQ DD NO: 16 is 49% identical to the human ras-related tumor suppressor NOEY2 (GenBank DD g4100355) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 3.6e-45, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO: 16 also contains a ras family domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) Data from BLIMPS and MOTIFS analyses provide further corroborative evidence that SEQ DD NO: 16 is a signaling protein of the ras family.
In an alternative example, SEQ DD NO:20 is 95% identical to murine SLM-1 protein (GenBank DD g4426613) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 3.1e-183, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ DD NO:20 also contains a KH domain (E- value is 0.11) as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See Table 3.) SEQ DD NO:l, SEQ DD NO:3-5, SEQ DD NO:7-9, SEQ DD NO: 11-13, SEQ DD NO: 14-15, and SEQ DD NO:17-19 were analyzed and annotated in a similar manner. The algorithms and parameters for the analysis of SEQ DD NO:1-20 are described in Table 7.
As shown in Table 4, the full length polynucleotide sequences of the present invention were assembled using cDNA sequences or coding (exon) sequences derived from genomic DNA, or any combination of these two types of sequences. Columns 1 and 2 list the polynucleotide sequence identification number (Polynucleotide SEQ DD NO:) and the corresponding Incyte polynucleotide consensus sequence number (Incyte Polynucleotide DD) for each polynucleotide of the invention. Column 3 shows the length of each polynucleotide sequence in basepairs. Column 4 lists fragments of the polynucleotide sequences which are useful, for example, in hybridization or amplification technologies that identify SEQ DD NO:21-40 or that distinguish between SEQ DD NO:21-40 and related polynucleotide sequences. Column 5 shows identification numbers corresponding to cDNA sequences, coding sequences (exons) predicted from genomic DNA, and/or sequence assemblages comprised of both cDNA and genomic DNA. These sequences were used to assemble the full length polynucleotide sequences of the invention. Columns 6 and 7 of Table 4 show the nucleotide start (5') and stop (3') positions of the cDNA and/or genomic sequences in column 5 relative to their respective full length sequences.
The identification numbers in Column 5 of Table 4 may refer specifically, for example, to Incyte cDNAs along with their corresponding cDNA libraries. For example, 105283R6 is the identification number of an Incyte cDNA sequence, and BMARNOT02 is the cDNA library from which it is derived. Incyte cDNAs for which cDNA libraries are not indicated were derived from pooled cDNA libraries (e.g., 71206562V1). Alternatively, the identification numbers in column 5 may refer to GenBank cDNAs or ESTs (e.g., g3034305) which contributed to the assembly of the full length polynucleotide sequences. In addition, the identification numbers in column 5 may identify sequences derived from the ENSEMBL (The Sanger Centre, Cambridge, UK) database (i.e., those sequences including the designation "ENST"). Alternatively, the identification numbers in column 5 may be derived from the NCBI RefSeq Nucleotide Sequence Records Database (i.e., those sequences including the designation "NM" or "NT") or the NCBI RefSeq Protein Sequence Records (i.e., those sequences including the designation "NP"). Alternatively, the identification numbers in column 5 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an "exon stitching" algorithm. For example, ¥L_XXXXXXJ!1_N2_YYYYY_N3_N4 represents a "stitched" sequence in which XXXXXX is the identification number of the cluster of sequences to which the algorithm was applied, and YYYYY is the number of the prediction generated by the algorithm, and N1A3_, if present, represent specific exons that may have been manually edited during analysis (See Example V). Alternatively, the identification numbers in column 5 may refer to assemblages of exons brought together by an "exon-stretching" algorithm. For example,
FLXXXXXX_gAAAAA_gBBBBB_l_N is the identification number of a "stretched" sequence, with XXXXXX being the Incyte project identification number, gAAAAA being the GenBank identification number of the human genomic sequence to which the "exon-stretching" algorithm was applied, gBBBBB being the GenBank identification number or NCBI RefSeq identification number of the nearest GenBank protein homolog, and N referring to specific exons (See Example V). In instances where a RefSeq sequence was used as a protein homolog for the "exon-stretching" algorithm, a RefSeq identifier (denoted by "ΝM," "ΝP," or "NT") may be used in place of the GenBank identifier (i.e., gBBBBB).
Alternatively, a prefix identifies component sequences that were hand-edited, predicted from genomic DNA sequences, or derived from a combination of sequence analysis methods. The following Table lists examples of component sequence prefixes and corresponding sequence analysis methods associated with the prefixes (see Example IV and Example V).
Figure imgf000035_0001
In some cases, Incyte cDNA coverage redundant with the sequence coverage shown in column 5 was obtained to confirm the final consensus polynucleotide sequence, but the relevant Incyte cDNA identification numbers are not shown.
Table 5 shows the representative cDNA libraries for those full length polynucleotide sequences which were assembled using Incyte cDNA sequences. The representative cDNA library is the Incyte cDNA library which is most frequently represented by the Incyte cDNA sequences which were used to assemble and confirm the above polynucleotide sequences. The tissues and vectors which were used to construct the cDNA libraries shown in Table 5 are described in Table 6. The invention also encompasses INTSIG variants. A preferred INTSIG variant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% amino acid sequence identity to the INTSIG amino acid sequence, and which contains at least one functional or structural characteristic of INTSIG.
The invention also encompasses polynucleotides which encode INTSIG. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ DD NO:21-40, which encodes INTSIG. The polynucleotide sequences of SEQ DD NO:21-40, as presented in the Sequence Listing, embrace the equivalent RNA sequences, wherein occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.
The invention also encompasses a variant of a polynucleotide sequence encoding INTSIG. In particular, such a variant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding INTSIG. A particular aspect of the invention encompasses a variant of a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ DD NO:21- 40 which has at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to a nucleic acid sequence selected from the group consisting of SEQ DD NO:21-40. Any one of the polynucleotide variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of INTSIG.
It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of polynucleotide sequences encoding INTSIG, some bearing minimal similarity to the polynucleotide sequences of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible variation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide sequence of naturally occurring INTSIG, and all such variations are to be considered as being specifically disclosed.
Although nucleotide sequences which encode INTSIG and its variants are generally capable of hybridizing to the nucleotide sequence of the naturally occurring INTSIG under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding INTSIG or its derivatives possessing a substantially different codon usage, e.g., inclusion of non- naturally occurring codons. Codons may be selected to increase the rate at which expression of the peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding INTSIG and its derivatives without altering the encoded amino acid sequences include the production of RNA transcripts having more desirable properties, such as a greater half-life, than transcripts produced from the naturally occurring sequence.
The invention also encompasses production of DNA sequences which encode INTSIG and INTSIG derivatives, or fragments thereof, entirely by synthetic chemistry. After production, the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents well known in the art. Moreover, synthetic chemistry may be used to introduce mutations into a sequence encoding INTSIG or any fragment thereof.
Also encompassed by the invention are polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ DD NO:21-40 and fragments thereof under various conditions of stringency. (See, e.g., Wahl, G.M. and S.L. Berger (1987) Methods Enzymol. 152:399-407; Kimmel, A.R. (1987) Methods Enzymol. 152:507- 511.) Hybridization conditions, including annealing and wash conditions, are described in "Definitions."
Methods for DNA sequencing are well known in the art and may be used to practice any of the embodiments of the invention. The methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical, Cleveland OH), Taq polymerase (Applied Biosystems), thermostable T7 polymerase (Amersham Pharmacia Biotech, Piscataway NJ), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE amplification system (Life Technologies, Gaithersburg MD). Preferably, sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno NV), PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (Applied Biosystems). Sequencing is then carried out using either the ABI 373 or 377 DNA sequencing system (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (Molecular Dynamics, Sunnyvale CA), or other systems known in the art. The resulting sequences are analyzed using a variety of algorithms which are well known in the art. (See, e.g., Ausubel, F.M. (1997) Short Protocols in Molecular Biology. John Wiley & Sons, New York NY, unit 7.7; Meyers, R.A. (1995) Molecular Biology and Biotechnology. Wiley VCH, New York NY, pp. 856-853.)
The nucleic acid sequences encoding INTSIG may be extended utilizing a partial nucleotide sequence and employing various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements. For example, one method which may be employed, restriction-site PCR, uses universal and nested primers to amplify unknown sequence from genomic DNA within a cloning vector. (See, e.g., Sarkar, G. (1993) PCR Methods Applic. 2:318-322.)
Another method, inverse PCR, uses primers that extend in divergent directions to amplify unknown sequence from a circularized template. The template is derived from restriction fragments comprising a known genomic locus and surrounding sequences. (See, e.g., Triglia, T. et al. (1988) Nucleic Acids Res. 16:8186.) A third method, capture PCR, involves PCR amplification of DNA fragments adjacent to known sequences in human and yeast artificial chromosome DNA. (See, e.g., Lagerstrom, M. et al. (1991) PCR Methods Applic. 1:111-119.) In this method, multiple restriction enzyme digestions and ligations may be used to insert an engineered double-stranded sequence into a region of unknown sequence before performing PCR. Other methods which may be used to retrieve unknown sequences are known in the art. (See, e.g., Parker, J.D. et al. (1991) Nucleic Acids Res. 19:3055-3060). Additionally, one may use PCR, nested primers, and PROMOTERFINDER libraries (Clontech, Palo Alto CA) to walk genomic DNA. This procedure avoids the need to screen libraries and is useful in finding intron/exon junctions. For all PCR-based methods, primers may be designed using commercially available software, such as OLIGO 4.06 primer analysis software (National Biosciences, Plymouth MN) or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the template at temperatures of about 68°C to 72°C
When screening for full length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. In addition, random-primed libraries, which often include sequences containing the 5' regions of genes, are preferable for situations in which an oligo d(T) library does not yield a full-length cDNA. Genomic libraries may be useful for extension of sequence into 5' non-transcribed regulatory regions.
Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products. In particular, capillary sequencing may employ flowable polymers for electrophoretic separation, four different nucleotide- specific, laser-stimulated fluorescent dyes, and a charge coupled device camera for detection of the emitted wavelengths. Output/light intensity may be converted to electrical signal using appropriate' software (e.g., GENOTYPER and SEQUENCE NAVIGATOR, Applied Biosystems), and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled. Capillary electrophoresis is especially preferable for sequencing small DNA fragments which may be present in limited amounts in a particular sample.
In another embodiment of the invention, polynucleotide sequences or fragments thereof which encode INTSIG may be cloned in recombinant DNA molecules that direct expression of INTSIG, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced and used to express INTSIG.
The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter INTSIG-encoding 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 and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. For example, oligonucleotide- mediated site-directed mutagenesis may be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.
The nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA; described in U.S. Patent No.
5,837,458; Chang, C.-C et al. (1999) Nat. Biotechnol. 17:793-797; Christians, F.C et al. (1999) Nat. Biotechnol. 17:259-264; and Crameri, A. et al. (1996) Nat. Biotechnol. 14:315-319) to alter or improve the biological properties of INTSIG, such as its biological or enzymatic activity or its ability to bind to other molecules or compounds. DNA shuffling is a process by which a library of gene variants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene variants with the desired properties. These preferred variants may then be pooled and further subjected to recursive rounds of DNA shuffling and selection/screening. Thus, genetic diversity is created through "artificial" breeding and rapid molecular evolution. For example, fragments of a single gene containing random point mutations may be recombined, screened, and then reshuffled until the desired properties are optimized. Alternatively, fragments of a given gene may be recombined with fragments of homologous genes in the same gene family, either from the same or different species, thereby maximizing the genetic diversity of multiple naturally occurring genes in a directed and controllable manner. In another embodiment, sequences encoding INTSIG may be synthesized, in whole or in part, using chemical methods well known in the art. (See, e.g., Caruthers, M.H. et al. (1980) Nucleic Acids Symp. Ser. 7:215-223; and Horn, T. et al. (1980) Nucleic Acids Symp. Ser. 7:225-232.) Alternatively, INTSIG itself or a fragment thereof may be synthesized using chemical methods. For example, peptide synthesis can be performed using various solution-phase or solid-phase techniques. (See, e.g., Creighton, T. (1984) Proteins. Structures and Molecular Properties. WH Freeman, New York NY, pp. 55-60; and Roberge, J.Y. et al. (1995) Science 269:202-204.) Automated synthesis may be achieved using the ABI 431 A peptide synthesizer (Applied Biosystems). Additionally, the amino acid sequence of INTSIG, or any part thereof, may be altered during direct synthesis and/or combined with sequences from other proteins, or any part thereof, to produce a variant polypeptide or a polypeptide having a sequence of a naturally occurring polypeptide.
The peptide may be substantially purified by preparative high performance liquid chromatography. (See, e.g., Chiez, R.M. and F.Z. Regnier (1990) Methods Enzymol. 182:392-421.) The composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing. (See, e.g., Creighton, supra, pp. 28-53.) In order to express a biologically active INTSIG, the nucleotide sequences encoding INTSIG or derivatives thereof may be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host. These elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5' and 3' untranslated regions in the vector and in polynucleotide sequences encoding INTSIG. Such elements may vary in their strength and specificity. Specific initiation signals may also be used to achieve more efficient translation of sequences encoding INTSIG. Such signals include the ATG initiation codon and adjacent sequences, e.g. the Kozak sequence. In cases where sequences encoding INTSIG and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals including an in-frame ATG initiation codon should be provided by the vector. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular host cell system used. (See, e.g., Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162.)
Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding INTSIG and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. (See, e.g., Sambrook, J. et al. (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview NY, ch. 4, 8, and 16-17; Ausubel, F.M. et al. (1995) Current Protocols in Molecular Biology, John Wiley & Sons, New York NY, ch. 9, 13, and 16.)
A variety of expression vector/host systems may be utilized to contain and express sequences encoding INTSIG. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e.g., baculovirus); plant cell systems transformed with viral expression vectors (e.g., cauliflower mosaic virus, CaMV, or tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems. (See, e.g., Sambrook, supra; Ausubel, supra; Van Heeke, G. and S.M. Schuster (1989) J. Biol. Chem. 264:5503-5509; Engelhard, E.K. et al. (1994) Proc. Natl. Acad. Sci. USA 91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7:1937-1945; Takamatsu, N. (1987) EMBO J. 6:307-311; The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York NY, pp. 191-196; Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA 81:3655-3659; and Harrington, J.J. et al. (1997) Nat. Genet. 15:345-355.) Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids, may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population. (See, e.g., Di Nicola, M. et al. (1998) Cancer Gen. Ther. 5(6):350-356; Yu, M. et al. (1993) Proc. Natl. Acad. Sci. USA 90(13):6340-6344; Buller, R.M. et al. (1985) Nature 317(6040):813-815; McGregor, D.P. et al. (1994) Mol. Immunol. 31(3):219-226; and Verma, I.M. and N. Somia (1997) Nature 389:239-242.) The invention is not limited by the host cell employed.
In bacterial systems, a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding INTSIG. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding INTSIG can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Life Technologies). Ligation of sequences encoding INTSIG into the vector's multiple cloning site disrupts the lacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence. (See, e.g., Van Heeke, G. and S.M. Schuster (1989) J. Biol. Chem. 264:5503-5509.) When large quantities of INTSIG are needed, e.g. for the production of antibodies, vectors which direct high level expression of INTSIG may be used. For example, vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used. Yeast expression systems may be used for production of INTSIG. A number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH promoters, may be used in the yeast Saccharomyces cerevisiae or Pichia pastoris. In addition, such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation. (See, e.g., Ausubel, 1995, supra; Bitter, G.A. et al. (1987) Methods Enzymol. 153:516-544; and Scorer, CA. et al. (1994) Bio/Technology 12:181-184.)
Plant systems may also be used for expression of INTSIG. Transcription of sequences encoding INTSIG may be driven by viral promoters, e.g., the 35S and 19S promoters of CaMV used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 6:307-311). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e.g., Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105.) These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. (See, e.g., The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York NY, pp. 191-196.)
In mammalian cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, sequences encoding INTSIG may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential El or E3 region of the viral genome may be used to obtain infective virus which expresses INTSIG in host cells. (See, e.g., Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA 81:3655-3659.) In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells. SV40 or EBV- based vectors may also be used for high-level protein expression.
Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained in and expressed from a plasmid. HACs of about 6 kb to 10 Mb are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. (See, e.g., Harrington, J.J. et al. (1997) Nat. Genet. 15:345- 355.) For long term production of recombinant proteins in mammalian systems, stable expression of
INTSIG in cell lines is preferred. For example, sequences encoding INTSIG can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media. The purpose of the selectable marker is to confer resistance to a ι selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be propagated using tissue culture techniques appropriate to the cell type.
Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase and adenine phosphoribosyltransferase genes, for use in fk and apr cells, respectively. (See, e.g., Wigler, M. et al. (1977) Cell 11:223-232; Lowy, I. et al. (1980) Cell 22:817-823.) Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate; neo confers resistance to the aminoglycosides neomycin and G-418; and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. (See, e.g., Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. USA 77:3567-3570; Colbere-Garapin, F. et al. (1981) J. Mol. Biol. 150:1-14.) Additional selectable genes have been described, e.g., trpB and hisD, which alter cellular requirements for metabolites. (See, e.g., Hartman, S.C and R.C Mulligan (1988) Proc. Natl. Acad. Sci. USA 85:8047-8051.) Visible markers, e.g., anthocyanins, green fluorescent proteins (GFP; Clontech), β glucuronidase and its substrate β-glucuronide, or luciferase and its substrate luciferin may be used. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system. (See, e.g., Rhodes, CA. (1995) Methods Mol. Biol. 55:121-131.)
Although the presence/absence of marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed. For example, if the sequence encoding INTSIG is inserted within a marker gene sequence, transformed cells containing sequences encoding INTSIG can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding INTSIG under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
In general, host cells that contain the nucleic acid sequence encoding INTSIG and that express INTSIG may 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 protein sequences. Immunological methods for detecting and measuring the expression of INTSIG 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). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on INTSIG is preferred, but a competitive binding assay may be employed. These and other assays are well known in the art. (See, e.g., Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St. Paul MN, Sect. IV; Coligan, J.E. et al. (1997) Current Protocols in Immunology, Greene Pub. Associates and Wiley-Interscience, New York NY; and Pound, J.D. (1998) Immunochemical Protocols, Humana Press, Totowa NJ.)
A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding INTSIG include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.
Alternatively, the sequences encoding INTSIG, or any fragments thereof, may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Amersham Pharmacia Biotech, Promega (Madison WI), and US Biochemical. Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.
Host cells transformed with nucleotide sequences encoding INTSIG may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a transformed cell may 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 polynucleotides which encode INTSIG may be designed to contain signal sequences which direct secretion of INTSIG through a prokaryotic or eukaryotic cell membrane. In addition, a host cell strain may 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 polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a "prepro" or "pro" form of the protein may 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 WI38) are available from the American Type Culture Collection (ATCC, Manassas VA) and may be chosen to ensure the correct modification and processing of the foreign protein. In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences encoding INTSIG may be ligated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems. For example, a chimeric INTSIG protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide libraries for inhibitors of INTSIG activity. Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matrices. Such moieties include, but are not limited to, glutathione S-transf erase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutinin (HA). GST, MBP, Trx, CBP, and 6-His enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively. FLAG, c-myc, and hemagglutinin (HA) enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags. A fusion protein may also be engineered to contain a proteolytic cleavage site located between the INTSIG encoding sequence and the heterologous protein sequence, so that INTSIG may be cleaved away from the heterologous moiety following purification. Methods for fusion protein expression and purification are discussed in Ausubel (1995, supra, ch. 10). A variety of commercially available kits may also be used to facilitate expression and purification of fusion proteins.
In a further embodiment of the invention, synthesis of radiolabeled INTSIG may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcription and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled amino acid precursor, for example, 35S-methionine.
INTSIG of the present invention or fragments thereof may be used to screen for compounds that specifically bind to INTSIG. At least one and up to a plurality of test compounds may be screened for specific binding to INTSIG. Examples of test compounds include antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules.
In one embodiment, the compound thus identified is closely related to the natural ligand of INTSIG, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner. (See, e.g., Coligan, J.E. et al. (1991) Current Protocols in Immunology 1(2): Chapter 5.) Similarly, the compound can be closely related to the natural receptor to which INTSIG binds, or to at least a fragment of the receptor, e.g., the ligand binding site. In either case, the compound can be rationally designed using known techniques. In one embodiment, screening for these compounds involves producing appropriate cells which express INTSIG, either as a secreted protein or on the cell membrane. Preferred cells include cells from mammals, yeast, Drosophila, or K coli. Cells expressing INTSIG or cell membrane fractions which contain INTSIG are then contacted with a test compound and binding, stimulation, or inhibition of activity of either INTSIG or the compound is analyzed.
An assay may simply test binding of a test compound to the polypeptide, wherein binding is detected by a fluorophore, radioisotope, enzyme conjugate, or other detectable label. For example, the assay may comprise the steps of combining at least one test compound with INTSIG, either in solution or affixed to a solid support, and detecting the binding of INTSIG to the compound. Alternatively, the assay may detect or measure binding of a test compound in the presence of a labeled competitor. Additionally, the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support. INTSIG of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of INTSIG. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for INTSIG activity, wherein INTSIG is combined with at least one test compound, and the activity of INTSIG in the presence of a test compound is compared with the activity of INTSIG in the absence of the test compound. A change in the activity of INTSIG in the presence of the test compound is indicative of a compound that modulates the activity of INTSIG. Alternatively, a test compound is combined with an in vitro or cell-free system comprising INTSIG under conditions suitable for INTSIG activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of INTSIG may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened.
In another embodiment, polynucleotides encoding INTSIG or their mammalian homologs may be "knocked out" in an animal model system using homologous recombination in embryonic stem (ES) cells. Such techniques are well known in the art and are useful for the generation of animal models of human disease. (See, e.g., U.S. Patent No. 5,175,383 and U.S. Patent No. 5,767,337.) For example, mouse ES cells, such as the mouse 129/SvJ cell line, are derived from the early mouse embryo and grown in culture. The ES cells are transformed with a vector containing the gene of interest disrupted by a marker gene, e.g., the neomycin phosphotransferase gene (neo; Capecchi, M.R. (1989) Science 244:1288-1292). The vector integrates into the corresponding region of the host genome by homologous recombination. Alternatively, homologous recombination takes place using the Cre-loxP system to knockout a gene of interest in a tissue- or developmental stage-specific manner (Marth, J.D. (1996) Clin. Invest. 97:1999-2002; Wagner, K.U. et al. (1997) Nucleic Acids Res. 25:4323-4330). Transformed ES cells are identified and microinjected into mouse cell blastocysts such as those from the C57BL/6 mouse strain. The blastocysts are surgically transferred to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains. Transgenic animals thus generated may be tested with potential therapeutic or toxic agents.
Polynucleotides encoding INTSIG may also be manipulated in vitro in ES cells derived from human blastocysts. Human ES cells have the potential to differentiate into at least eight separate cell lineages including endoderm, mesoderm, and ectodermal cell types. These cell lineages differentiate into, for example, neural cells, hematopoietic lineages, and cardiomyocytes (Thomson, J.A. et al. (1998) Science 282:1145-1147).
Polynucleotides encoding INTSIG can also be used to create "knockin" humanized animals (pigs) or transgenic animals (mice or rats) to model human disease. With knockin technology, a region of a polynucleotide encoding INTSIG is injected into animal ES cells, and the injected sequence integrates into the animal cell genome. Transformed cells are injected into blastulae, and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease. Alternatively, a mammal inbred to overexpress INTSIG, e.g., by secreting INTSIG in its milk, may also serve as a convenient source of that protein (Janne, J. et al. (1998) Biotechnol. Annu. Rev. 4:55-74). THERAPEUTICS
Chemical and structural similarity, e.g., in the context of sequences and motifs, exists between regions of INTSIG and intracellular signaling molecules. In addition, the expression of INTSIG is closely associated with brain and neurological tissues including thoracic dorsal root ganglion tissue, dermal tissue, reproductive tissue, digestive and hemic/immune tissue, diseased prostate tissue, and tumorous tissues including bladder, tongue, and testicular. Therefore, INTSIG appears to play a role in cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders. In the treatment of disorders associated with increased INTSIG expression or activity, it is desirable to decrease the expression or activity of INTSIG. In the treatment of disorders associated with decreased INTSIG expression or activity, it is desirable to increase the expression or activity of INTSIG.
Therefore, in one embodiment, INTSIG or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (ADDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia,
Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and
Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; a gastrointestinal disorder such as dysphagia, peptic esophagitis, esophageal spasm, esophageal stricture, esophageal carcinoma, dyspepsia, indigestion, gastritis, gastric carcinoma, anorexia, nausea, emesis, gastroparesis, antral or pyloric edema, abdominal angina, pyrosis, gastroenteritis, intestinal obstruction, infections of the intestinal tract, peptic ulcer, cholelithiasis, cholecystitis, cholestasis, pancreatitis, pancreatic carcinoma, biliary tract disease, hepatitis, hyperbilirubinemia, cirrhosis, passive congestion of the liver, hepatoma, infectious colitis, ulcerative colitis, ulcerative proctitis, Crohn's disease, Whipple's disease, Mallory-Weiss syndrome, colonic carcinoma, colonic obstruction, irritable bowel syndrome, short bowel syndrome, diarrhea, constipation, gastrointestinal hemorrhage, acquired immunodeficiency syndrome (ADDS) enteropathy, jaundice, hepatic encephalopathy, hepatorenal syndrome, hepatic steatosis, hemochromatosis, Wilson's disease, alphaj-antitrypsin deficiency, Reye's syndrome, primary sclerosing cholangitis, liver infarction, portal vein obstruction and thrombosis, centrilobular necrosis, peliosis hepatis, hepatic vein thrombosis, veno- occlusive disease, preeclampsia, eclampsia, acute fatty liver of pregnancy, intrahepatic cholestasis of pregnancy, and hepatic tumors including nodular hyperplasias, adenomas, and carcinomas; a reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, autoimmune disorders, ectopic pregnancy, teratogenesis; cancer of the breast, fibrocystic breast disease, galactorrhea; a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puperty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumours; and a developmental disorder such as renal tubular acidosis, anemia, Gushing' s syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith- Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss. In another embodiment, a vector capable of expressing INTSIG or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG including, but not limited to, those described above.
In a further embodiment, a composition comprising a substantially purified INTSIG in conjunction with a suitable pharmaceutical carrier may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG including, but not limited to, those provided above.
In still another embodiment, an agonist which modulates the activity of INTSIG may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of INTSIG including, but not limited to, those listed above. In a further embodiment, an antagonist of INTSIG may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of INTSIG. Examples of such disorders include, but are not limited to, those cell proliferative, autoimmune/inflammatory, neurological, gastrointestinal, reproductive, and developmental disorders described above. In one aspect, an antibody which specifically binds INTSIG may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express INTSIG.
In an additional embodiment, a vector expressing the complement of the polynucleotide encoding INTSIG may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of INTSIG including, but not limited to, those described above. In other embodiments, any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
An antagonist of INTSIG may be produced using methods which are generally known in the art. In particular, purified INTSIG may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind INTSIG. Antibodies to INTSIG may also be generated using methods that are well known in the art. Such antibodies may 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 (i.e., those which inhibit dimer formation) are generally preferred for therapeutic use. For the production of antibodies, various hosts including goats, rabbits, rats, mice, humans, and others may be immunized by injection with INTSIG or with any fragment or oligopeptide thereof which has immunogenic properties. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol. Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are especially preferable.
It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to INTSIG have an amino acid sequence consisting of at least about 5 amino acids, and generally will consist of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein. Short stretches of INTSIG amino acids may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced.
Monoclonal antibodies to INTSIG may 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 technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e.g., Kohler, G. et al. (1975) Nature 256:495-497; Kozbor, D. et al. (1985) J. Immunol. Methods 81:31-42; Cote, R.J. et al. (1983) Proc. Natl. Acad. Sci. USA 80:2026-2030; and Cole, S.P. et al. (1984) Mol. Cell Biol. 62:109-120.)
In addition, techniques developed for the production of "chimeric antibodies," such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. (See, e.g., Morrison, S.L. et al. (1984) Proc. Natl. Acad. Sci. USA 81:6851-6855; Neuberger, M.S. et al. (1984) Nature 312:604-608; and Takeda, S. et al. (1985) Nature 314:452-454.) Alternatively, techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce INTSIG-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (See, e.g., Burton, D.R. (1991) Proc. Natl. Acad. Sci. USA 88:10134-10137.)
Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (See, e.g., Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. USA 86:3833-3837; Winter, G. et al. (1991) Nature 349:293-299.)
Antibody fragments which contain specific binding sites for INTSIG may also be generated. For example, such fragments include, but are not limited to, F(ab')2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e.g., Huse, W.D. et al. (1989) Science 246:1275-1281.)
Various immunoassays may be used for screening 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. Such immunoassays typically involve the measurement of complex formation between INTSIG and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering INTSIG epitopes is generally used, but a competitive binding assay may also be employed (Pound, supra).
Various methods such as Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for INTSIG. Affinity is expressed as an association constant, Ka, which is defined as the molar concentration of INTSIG-antibody complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions. The K, determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple INTSIG epitopes, represents the average affinity, or avidity, of the antibodies for INTSIG. The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular INTSIG epitope, represents a true measure of affinity. High-affinity antibody preparations with K, ranging from about 109 to 1012 L/mole are preferred for use in immunoassays in which the INTSIG- antibody complex must withstand rigorous manipulations. Low-affinity antibody preparations with Ka ranging from about 106 to 107 L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of INTSIG, preferably in active form, from the antibody (Catty, D. (1988) Antibodies, Volume I: A Practical Approach. IRL Press, Washington DC; Liddell, J.E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies, John Wiley & Sons, New York NY).
The titer and avidity of polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications. For example, a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml, preferably 5-10 mg specific antibody/ml, is generally employed in procedures requiring precipitation of INTSIG-antibody complexes. Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available. (See, e.g., Catty, supra, and Coligan et al. supra.)
In another embodiment of the invention, the polynucleotides encoding INTSIG, or any fragment or complement thereof, may be used for therapeutic purposes. In one aspect, modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified oligonucleotides) to the coding or regulatory regions of the gene encoding INTSIG. Such technology is well known in the art, and antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding INTSIG. (See, e.g., Agrawal, S., ed. (1996) Antisense Therapeutics, Humana Press Inc., Totawa NJ.)
In therapeutic use, any gene delivery system suitable for introduction of the antisense sequences into appropriate target cells can be used. Antisense sequences can be delivered intracellularly in the form of an expression plasmid which, upon transcription, produces a sequence complementary to at least a portion of the cellular sequence encoding the target protein. (See, e.g., Slater, J.E. et al. (1998) J. Allergy Clin. Immunol. 102(3):469-475; and Scanlon, K.J. et al. (1995) 9(13): 1288-1296.) Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors. (See, e.g., Miller, A.D. (1990) Blood 76:271; Ausubel, supra; Uckert, W. and W. Walther (1994) Pharmacol. Ther. 63(3):323-347.) Other gene delivery mechanisms include liposome-derived systems, artificial viral envelopes, and other systems known in the art. (See, e.g., Rossi, J.J. (1995) Br. Med. Bull. 51(l):217-225; Boado, R.J. et al. (1998) J. Pharm. Sci. 87(11):1308-1315; and Morris, M.C. et al. (1997) Nucleic Acids Res. 25(14):2730-2736.)
In another embodiment of the invention, polynucleotides encoding INTSIG may be used for somatic or germline gene therapy. Gene therapy may be performed to (i) correct a genetic deficiency (e.g., in the cases of severe combined immunodeficiency (SCDD)-Xl disease characterized by X- linked inheritance (Cavazzana-Calvo, M. et al. (2000) Science 288:669-672), severe combined immunodeficiency syndrome associated with an inherited adenosine deaminase (ADA) deficiency (Blaese, R.M. et al. (1995) Science 270:475-480; Bordignon, C. et al. (1995) Science 270:470-475), cystic fibrosis (Zabner, J. et al. (1993) Cell 75:207-216; Crystal, R.G. et al. (1995) Hum. Gene
Therapy 6:643-666; Crystal, R.G. et al. (1995) Hum. Gene Therapy 6:667-703), thalassamias, familial hypercholesterolemia, and hemophilia resulting from Factor VDI or Factor IX deficiencies (Crystal, R.G. (1995) Science 270:404-410; Verma, I.M. and N. Somia (1997) Nature 389:239-242)), (ii) express a conditionally lethal gene product (e.g., in the case of cancers which result from unregulated cell proliferation), or (iii) express a protein which affords protection against intracellular parasites (e.g., against human retroviruses, such as human immunodeficiency virus (HIV) (Baltimore, D. (1988) Nature 335:395-396; Poeschla, E. et al. (1996) Proc. Natl. Acad. Sci. USA. 93:11395-11399), hepatitis B or C virus (HBV, HCV); fungal parasites, such as Candida albicans and Paracoccidioides brasiliensis; and protozoan parasites such as Plasmodium falciparum and Trvpanosoma cruzi). In the case where a genetic deficiency in INTSIG expression or regulation causes disease, the expression of INTSIG from an appropriate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency.
In a further embodiment of the invention, diseases or disorders caused by deficiencies in INTSIG are treated by constructing mammalian expression vectors encoding INTSIG and introducing these vectors by mechanical means into INTSIG-deficient cells. Mechanical transfer technologies for use with cells in vivo or ex vitro include (i) direct DNA microinjection into individual cells, (ii) ballistic gold particle delivery, (iii) liposome-mediated transfection, (iv) receptor-mediated gene transfer, and (v) the use of DNA transposons (Morgan, R.A. and W.F. Anderson (1993) Annu. Rev. Biochem. 62:191-217; Ivies, Z. (1997) Cell 91:501-510; Boulay, J-L. and H. Recipon (1998) Curr. Opin. Biotechnol. 9:445-450).
Expression vectors that may be effective for the expression of INTSIG include, but are not limited to, the PCDNA 3.1, EPITAG, PRCCMV2, PREP, PVAX, PCR2-TOPOTA vectors (Invitrogen, Carlsbad CA), PCMV-SCRIPT, PCMV-TAG, PEGSH/PERV (Stratagene, La Jolla CA), and PTET-OFF, PTET-ON, PTRE2, PTRE2-LUC, PTK-HYG (Clontech, Palo Alto CA). INTSIG may be expressed using (i) a constitutively active promoter, (e.g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or β-actin genes), (ii) an inducible promoter (e.g., the tetracycline-regulated promoter (Gossen, M. and H. Bujard (1992) Proc. Natl. Acad. Sci. USA 89:5547-5551; Gossen, M. et al. (1995) Science 268:1766-1769; Rossi, F.M.V. and H.M. Blau (1998) Curr. Opin. Biotechnol. 9:451-456), commercially available in the T-REX plasmid (Invitrogen)); the ecdysone-inducible promoter (available in the plasmids PVGRXR and PIND; Invitrogen); the FK506/rapamycin inducible promoter; or the RU486/mifepristone inducible promoter (Rossi, F.M.V. and Blau, H.M. supra)), or (iii) a tissue-specific promoter or the native promoter of the endogenous gene encoding INTSIG from a normal individual. Commercially available liposome transformation kits (e.g., the PERFECT LIPDD
TRANSFECTION KIT, available from Invitrogen) allow one with ordinary skill in the art to deliver polynucleotides to target cells in culture and require minimal effort to optimize experimental parameters. In the alternative, transformation is performed using the calcium phosphate method (Graham, F.L. and A.J. Eb (1973) Virology 52:456-467), or by electroporation (Neumann, E. et al. (1982) EMBO J. 1:841-845). The introduction of DNA to primary cells requires modification of these standardized mammalian transfection protocols.
In another embodiment of the invention, diseases or disorders caused by genetic defects with respect to INTSIG expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding INTSIG under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (ii) appropriate RNA packaging signals, and (iii) a Rev-responsive element (RRE) along with additional retrovirus cz's-acting RNA sequences and coding sequences required for efficient vector propagation. Retrovirus vectors (e.g., PFB and PFBNEO) are commercially available (Stratagene) and are based on published data (Riviere, I. et al. (1995) Proc. Natl. Acad. Sci. USA 92:6733-6737), incorporated by reference herein. The vector is propagated in an appropriate vector producing cell line (VPCL) that expresses an envelope gene with a tropism for receptors on the target cells or a promiscuous envelope protein such as VSVg (Armentano, D. et al. (1987) J. Virol. 61:1647-1650; Bender, M.A. et al. (1987) J. Virol. 61:1639-1646; Adam, M.A. and A.D. Miller (1988) J. Virol. 62:3802-3806; Dull, T. et al. (1998) J. Virol. 72:8463-8471; Zufferey, R. et al. (1998) J. Virol. 72:9873-9880). U.S. Patent No. 5,910,434 to Rigg ("Method for obtaining retrovirus packaging cell lines producing high transducing efficiency retroviral supernatant") discloses a method for obtaining retrovirus packaging cell lines, and is hereby incorporated by reference. Propagation of retrovirus vectors, transduction of a population of cells (e.g., CD4+ T-cells), and the return of transduced cells to a patient are procedures well known to persons skilled in the art of gene therapy and have been well documented (Ranga, U. et al. (1997) J. Virol. 71:7020-7029; Bauer, G. et al. (1997) Blood 89:2259-2267; Bonyhadi, M.L. (1997) J. Virol. 71:4707-4716; Ranga, U. et al. (1998) Proc. Natl. Acad. Sci. USA 95:1201-1206; Su, L. (1997) Blood 89:2283-2290).
In the alternative, an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding INTSIG to cells which have one or more genetic abnormalities with respect to the expression of INTSIG. The construction and packaging of adenovirus-based vectors are well known to those with ordinary skill in the art. Replication defective adenovirus vectors have proven to be versatile for importing genes encoding immunoregulatory proteins into intact islets in the pancreas (Csete, M.E. et al. (1995) Transplantation 27:263-268). Potentially useful adenoviral vectors are described in U.S. Patent No. 5,707,618 to Armentano ("Adenovirus vectors for gene therapy"), hereby incorporated by reference. For adenoviral vectors, see also Antinozzi, P.A. et al. (1999) Annu. Rev. Nutr. 19:511-544 and Verma, I.M. and N. Somia (1997) Nature 18:389:239-242, both incorporated by reference herein.
In another alternative, a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding INTSIG to target cells which have one or more genetic abnormalities with respect to the expression of INTSIG. The use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing INTSIG to cells of the central nervous system, for which HSV has a tropism. The construction and packaging of herpes-based vectors are well known to those with ordinary skill in the art. A replication-competent herpes simplex virus (HSV) type 1 -based vector has been used to deliver a reporter gene to the eyes of primates (Liu, X. et al. (1999) Exp. Eye Res. 169:385-395). The construction of a HSV-1 virus vector has also been disclosed in detail in U.S. Patent No. 5,804,413 to DeLuca ("Herpes simplex virus strains for gene transfer"), which is hereby incorporated by reference. U.S. Patent No. 5,804,413 teaches the use of recombinant HSV d92 which consists of a genome containing at least one exogenous gene to be transferred to a cell under the control of the appropriate promoter for purposes including human gene therapy. Also taught by this patent are the construction and use of recombinant HSV strains deleted for ICP4, ICP27 and ICP22. For HSV vectors, see also Goins, W.F. et al. (1999) J. Virol. 73:519-532 and Xu, H. et al. (1994) Dev. Biol. 163:152-161, hereby incorporated by reference. The manipulation of cloned herpesvirus sequences, the generation of recombinant virus following the transfection of multiple plasmids containing different segments of the large herpesvirus genomes, the growth and propagation of herpesvirus, and the infection of cells with herpesvirus are techniques well known to those of ordinary skill in the art.
In another alternative, an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding INTSIG to target cells. The biology of the prototypic alphavirus, Semliki Forest Virus (SFV), has been studied extensively and gene transfer vectors have been based on the SFV genome (Garoff, H. and K.-J. Li (1998) Curr. Opin. Biotechnol. 9:464-469). During alphavirus RNA replication, a subgenomic RNA is generated that normally encodes the viral capsid proteins. This subgenomic RNA replicates to higher levels than the full length genomic RNA, resulting in the overproduction of capsid proteins relative to the viral proteins with enzymatic activity (e.g., protease and polymerase). Similarly, inserting the coding sequence for INTSIG into the alphavirus genome in place of the capsid-coding region results in the production of a large number of INTSIG-coding RNAs and the synthesis of high levels of INTSIG in vector transduced cells. While alphavirus infection is typically associated with cell lysis within a few days, the ability to establish a persistent infection in hamster normal kidney cells (BHK-21) with a variant of Sindbis virus (SIN) indicates that the lytic replication of alphaviruses can be altered to suit the needs of the gene therapy application (Dryga, S.A. et al. (1997) Virology 228:74-83). The wide host range of alphaviruses will allow the introduction of INTSIG into a variety of cell types. The specific transduction of a subset of cells in a population may require the sorting of cells prior to transduction. The methods of manipulating infectious cDNA clones of alphaviruses, performing alphavirus cDNA and RNA transfections, and performing alphavirus infections, are well known to those with ordinary skill in the art.
Oligonucleotides derived from the transcription initiation site, e.g., between about positions -10 and +10 from the start site, may also be employed to inhibit gene expression. 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, J.E. et al. (1994) in Huber, B.E. and B.I. Carr, Molecular and Immunologic Approaches, Furura Publishing, Mt. Kisco NY, pp. 163-177.) A complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.
Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. For example, engineered hammerhead motif ribozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding INTSIG.
Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, including the following sequences: GUA, GUU, and GUC Once identified, short RNA sequences of between 15 and 20 ribonucleotides, corresponding to the region of the target gene containing the cleavage site, may be evaluated for secondary structural features which may render the oligonucleotide inoperable. The suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.
Complementary ribonucleic acid molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding INTSIG. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6. Alternatively, these cDNA constructs that synthesize complementary RNA, constitutively or inducibly, can be introduced into cell lines, cells, or tissues.
RNA molecules may 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 phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of 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. An additional embodiment of the invention encompasses a method for screening for a compound which is effective in altering expression of a polynucleotide encoding INTSIG. Compounds which may be effective in altering expression of a specific polynucleotide may include, but are not limited to, oligonucleotides, antisense oligonucleotides, triple helix-forming oligonucleotides, transcription factors and other polypeptide transcriptional regulators, and non-macromolecular chemical entities which are capable of interacting with specific polynucleotide sequences. Effective compounds may alter polynucleotide expression by acting as either inhibitors or promoters of polynucleotide expression. Thus, in the treatment of disorders associated with increased INTSIG expression or activity, a compound which specifically inhibits expression of the polynucleotide encoding INTSIG may be therapeutically useful, and in the treatment of disorders associated with decreased INTSIG expression or activity, a compound which specifically promotes expression of the polynucleotide encoding INTSIG may be therapeutically useful.
At least one, and up to a plurality, of test compounds may be screened for effectiveness in altering expression of a specific polynucleotide. A test compound may be obtained by any method commonly known in the art, including chemical modification of a compound known to be effective in altering polynucleotide expression; selection from an existing, commercially-available or proprietary library of naturally-occurring or non-natural chemical compounds; rational design of a compound based on chemical and/or structural properties of the target polynucleotide; and selection from a library of chemical compounds created combinatorially or randomly. A sample comprising a polynucleotide encoding INTSIG is exposed to at least one test compound thus obtained. The sample may comprise, for example, an intact or permeabilized cell, or an in vitro cell-free or reconstituted biochemical system. Alterations in the expression of a polynucleotide encoding INTSIG are assayed by any method commonly known in the art. Typically, the expression of a specific nucleotide is detected by hybridization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding INTSIG. The amount of hybridization may be quantified, thus forming the basis for a comparison of the expression of the polynucleotide both with and without exposure to one or more test compounds. Detection of a change in the expression of a polynucleotide exposed to a test compound indicates that the test compound is effective in altering the expression of the polynucleotide. A screen for a compound effective in altering expression of a specific polynucleotide can be carried out, for example, using a Schizosaccharomvces pombe gene expression system (Atkins, D. et al. (1999) U.S. Patent No. 5,932,435; Arndt, G.M. et al. (2000) Nucleic Acids Res. 28:E15) or a human cell line such as HeLa cell (Clarke, ML. et al. (2000) Biochem. Biophys. Res. Commun. 268:8-13). A particular embodiment of the present invention involves screening a combinatorial library of oligonucleotides (such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides) for antisense activity against a specific polynucleotide sequence (Bruice, T.W. et al. (1997) U.S. Patent No. 5,686,242; Bruice, T.W. et al. (2000) U.S. Patent No. 6,022,691).
Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient. Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See, e.g., Goldman, C.K. et al. (1997) Nat. Biotechnol. 15:462-466.)
Any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as humans, dogs, cats, cows, horses, rabbits, and monkeys.
An additional embodiment of the invention relates to the administration of a composition which generally comprises an active ingredient formulated with a pharmaceutically acceptable excipient. Excipients may include, for example, sugars, starches, celluloses, gums, and proteins. Various formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton PA). Such compositions may consist of INTSIG, antibodies to INTSIG, and mimetics, agonists, antagonists, or inhibitors of INTSIG.
The compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, pulmonary, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.
Compositions for pulmonary administration may be prepared in liquid or dry powder form. These compositions are generally aerosolized immediately prior to inhalation by the patient. In the case of small molecules (e.g. traditional low molecular weight organic drugs), aerosol delivery of fast- acting formulations is well-known in the art. In the case of macromolecules (e.g. larger peptides and proteins), recent developments in the field of pulmonary delivery via the alveolar region of the lung have enabled the practical delivery of drugs such as insulin to blood circulation (see, e.g., Patton, J.S. et al., U.S. Patent No. 5,997,848). Pulmonary delivery has the advantage of administration without needle injection, and obviates the need for potentially toxic penetration enhancers.
Compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is well within the capability of those skilled in the art.
Specialized forms of compositions may be prepared for direct intracellular delivery of macromolecules comprising INTSIG or fragments thereof. For example, liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule. Alternatively, INTSIG or a fragment thereof may be joined to a short cationic N- terminal portion from the HIV Tat-1 protein. Fusion proteins thus generated have been found to transduce into the cells of all tissues, including the brain, in a mouse model system (Schwarze, S.R. et al. (1999) Science 285:1569-1572). For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays, e.g., of neoplastic cells, or in animal models such as mice, rats, rabbits, dogs, monkeys, or pigs. An animal model may also be used to determine the appropriate 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, for example
INTSIG or fragments thereof, antibodies of INTSIG, and agonists, antagonists or inhibitors of ENTTSIG, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the ED30 (the dose therapeutically effective in 50% of the population) or LD50 (the dose lethal to 50% of the population) statistics. The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the LD50/ED50 ratio. Compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used to formulate a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that includes the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.
The exact dosage will be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. Long-acting compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation.
Normal dosage amounts may vary from about 0.1 μg to 100,000 μg, up to a total dose of about 1 gram, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art. Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc. DIAGNOSTICS
In another embodiment, antibodies which specifically bind INTSIG may be used for the diagnosis of disorders characterized by expression of INTSIG, or in assays to monitor patients being treated with INTSIG or agonists, antagonists, or inhibitors of INTSIG. Antibodies useful for diagnostic purposes may be prepared in the same manner as described above for therapeutics. Diagnostic assays for INTSIG include methods which utilize the antibody and a label to detect INTSIG in human body fluids or in extracts of cells or tissues. The antibodies may be used with or without modification, and may be labeled by covalent or non-covalent attachment of a reporter molecule. A wide variety of reporter molecules, several of which are described above, are known in the art and may be used. A variety of protocols for measuring INTSIG, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of INTSIG expression. Normal or standard values for INTSIG expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to INTSIG under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of INTSIG expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.
In another embodiment of the invention, the polynucleotides encoding INTSIG may be used for diagnostic purposes. The polynucleotides which may be used include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs. The polynucleotides may be used to detect and quantify gene expression in biopsied tissues in which expression of INTSIG may be correlated with disease. The diagnostic assay may be used to determine absence, presence, and excess expression of INTSIG, and to monitor regulation of INTSIG levels during therapeutic intervention. In one aspect, hybridization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding INTSIG or closely related molecules may be used to identify nucleic acid sequences which encode INTSIG. The specificity of the probe, whether it is made from a highly specific region, e.g., the 5' regulatory region, or from a less specific region, e.g., a conserved motif, and the stringency of the hybridization or amplification will determine whether the probe identifies only naturally occurring sequences encoding INTSIG, allelic variants, or related sequences.
Probes may also be used for the detection of related sequences, and may have at least 50% sequence identity to any of the INTSIG encoding sequences. The hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ DD NO:21-40 or from genomic sequences including promoters, enhancers, and introns of the INTSIG gene.
Means for producing specific hybridization probes for DNAs encoding ESfTSIG include the cloning of polynucleotide sequences encoding INTSIG or INTSIG derivatives into vectors for the production of mRNA probes. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides. Hybridization probes may be labeled by a variety of reporter groups, for example, by radionuclides such as 32P or 35S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.
Polynucleotide sequences encoding INTSIG may be used for the diagnosis of disorders associated with expression of INTSIG. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (ADDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis- ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjδgren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt- Jakob disease, and Gerstmann-Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; a gastrointestinal disorder such as dysphagia, peptic esophagitis, esophageal spasm, esophageal stricture, esophageal carcinoma, dyspepsia, indigestion, gastritis, gastric carcinoma, anorexia, nausea, emesis, gastroparesis, antral or pyloric edema, abdominal angina, pyrosis, gastroenteritis, intestinal obstruction, infections of the intestinal tract, peptic ulcer, cholelithiasis, cholecystitis, cholestasis, pancreatitis, pancreatic carcinoma, biliary tract disease, hepatitis, hyperbilirubinemia, cirrhosis, passive congestion of the liver, hepatoma, infectious colitis, ulcerative colitis, ulcerative proctitis, Crohn's disease, Whipple's disease, Mallory- Weiss syndrome, colonic carcinoma, colonic obstruction, irritable bowel syndrome, short bowel syndrome, diarrhea, constipation, gastrointestinal hemorrhage, acquired immunodeficiency syndrome (ADDS) enteropathy, jaundice, hepatic encephalopathy, hepatorenal syndrome, hepatic steatosis, hemochromatosis, Wilson's disease, alphaj-antitrypsin deficiency, Reye's syndrome, primary sclerosing cholangitis, liver infarction, portal vein obstruction and thrombosis, centrilobular necrosis, peliosis hepatis, hepatic vein thrombosis, veno-occlusive disease, preeclampsia, eclampsia, acute fatty liver of pregnancy, intrahepatic cholestasis of pregnancy, and hepatic tumors including nodular hyperplasias, adenomas, and carcinomas; a reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, autoimmune disorders, ectopic pregnancy, teratogenesis; cancer of the breast, fibrocystic breast disease, galactorrhea; a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puperty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumours; and a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss. The polynucleotide sequences encoding INTSIG may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and multiforrnat ELISA-like assays; and in microarrays utilizing fluids or tissues from patients to detect altered INTSIG expression. Such qualitative or quantitative methods are well known in the art.
In a particular aspect, the nucleotide sequences encoding INTSIG may be useful in assays that detect the presence of associated disorders, particularly those mentioned above. The nucleotide sequences encoding INTSIG may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes. After a suitable incubation period, the sample is washed and the signal is quantified and compared with a standard value. If the amount of signal in the patient sample is significantly altered in comparison to a control sample then the presence of altered levels of nucleotide sequences encoding INTSIG in the sample indicates the presence of the associated disorder. Such assays may 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 patient.
In order to provide a basis for the diagnosis of a disorder associated with expression of INTSIG, a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, encoding INTSIG, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with values from an experiment in which a known amount of a substantially purified polynucleotide is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder.
Once the presence of a disorder is established and a treatment protocol is initiated, hybridization assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in the normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.
With respect to cancer, the presence of an abnormal amount of transcript (either under- or overexpressed) in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer. Additional diagnostic uses for oligonucleotides designed from the sequences encoding INTSIG may involve the use of PCR. These oligomers may be chemically synthesized, generated enzymatically, or produced in vitro. Oligomers will preferably contain a fragment of a polynucleotide encoding INTSIG, or a fragment of a polynucleotide complementary to the polynucleotide encoding INTSIG, and will be employed under optimized conditions for identification of a specific gene or condition. Oligomers may also be employed under less stringent conditions for detection or quantification of closely related DNA or RNA sequences.
In a particular aspect, oligonucleotide primers derived from the polynucleotide sequences encoding INTSIG may be used to detect single nucleotide polymorphisms (SNPs). SNPs are substitutions, insertions and deletions that are a frequent cause of inherited or acquired genetic disease in humans. Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods. In SSCP, oligonucleotide primers derived from the polynucleotide sequences encoding INTSIG are used to amplify DNA using the polymerase chain reaction (PCR). The DNA may be derived, for example, from diseased or normal tissue, biopsy samples, bodily fluids, and the like. SNPs in the DNA cause differences in the secondary and tertiary structures of PCR products in single-stranded form, and these differences are detectable using gel electrophoresis in non-denaturing gels. In fSCCP, the oligonucleotide primers are fluorescently labeled, which allows detection of the amplimers in high-throughput equipment such as DNA sequencing machines. Additionally, sequence database analysis methods, termed in silico SNP (isSNP), are capable of identifying polymorphisms by comparing the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence. These computer- based methods filter out sequence variations due to laboratory preparation of DNA and sequencing errors using statistical models and automated analyses of DNA sequence chromatograms. In the alternative, SNPs may be detected and characterized by mass spectrometry using, for example, the high throughput MASSARRAY system (Sequenom, Inc., San Diego CA).
Methods which may also be used to quantify the expression of INTSIG include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves. (See, e.g., Melby, P.C et al. (1993) J. Immunol. Methods 159:235-244; Duplaa, C. et al. (1993) Anal. Biochem. 212:229-236.) The speed of quantitation of multiple samples may be accelerated by running the assay in a high-throughput format where the oligomer or polynucleotide of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.
In further embodiments, oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as elements on a microarray. The microarray can be used in transcript imaging techniques which monitor the relative expression levels of large numbers of genes simultaneously as described below. The microarray may also be used to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic basis of a disorder, to diagnose a disorder, to monitor progression/regression of disease as a function of gene expression, and to develop and monitor the activities of therapeutic agents in the treatment of disease. In particular, this information may be used to develop a pharmacogenomic profile of a patient in order to select the most appropriate and effective treatment regimen for that patient. For example, therapeutic agents which are highly effective and display the fewest side effects may be selected for a patient based on his/her pharmacogenomic profile. In another embodiment, INTSIG, fragments of INTSIG, or antibodies specific for INTSIG may be used as elements on a microarray. The microarray may be used to monitor or measure protein-protein interactions, drug-target interactions, and gene expression profiles, as described above.
A particular embodiment relates to the use of the polynucleotides of the present invention to generate a transcript image of a tissue or cell type. A transcript image represents the global pattern of gene expression by a particular tissue or cell type. Global gene expression patterns are analyzed by quantifying the number of expressed genes and their relative abundance under given conditions and at a given time. (See Seilhamer et al., "Comparative Gene Transcript Analysis," U.S. Patent No. 5,840,484, expressly incorporated by reference herein.) Thus a transcript image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcripts or reverse transcripts of a particular tissue or cell type. In one embodiment, the hybridization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements comprise a subset of a plurality of elements on a microarray. The resultant transcript image would provide a profile of gene activity. Transcript images may be generated using transcripts isolated from tissues, cell lines, biopsies, or other biological samples. The transcript image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or in vitro, as in the case of a cell line.
Transcript images which profile the expression of the polynucleotides of the present invention may also be used in conjunction with in vitro model systems and preclinical evaluation of pharmaceuticals, as well as toxicological testing of industrial and naturally-occurring environmental compounds. All compounds induce characteristic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E.F. et al. (1999) Mol. Carcinog. 24:153-159; Steiner, S. and N.L. Anderson (2000) Toxicol. Lett. 112-113:467-471, expressly incorporated by reference herein). If a test compound has a signature similar to that of a compound with known toxicity, it is likely to share those toxic properties. These fingerprints or signatures are most useful and refined when they contain expression information from a large number of genes and gene families. Ideally, a genome-wide measurement of expression provides the highest quality signature. Even genes whose expression is not altered by any tested compounds are important as well, as the levels of expression of these genes are used to normalize the rest of the expression data. The normalization procedure is useful for comparison of expression data after treatment with different compounds. While the assignment of gene function to elements of a toxicant signature aids in interpretation of toxicity mechanisms, knowledge of gene function is not necessary for the statistical matching of signatures which leads to prediction of toxicity. (See, for example, Press Release 00-02 from the National Institute of Environmental Health Sciences, released February 29, 2000, available at http://www.niehs.nih.gov/oc/news/toxchip.htm.) Therefore, it is important and desirable in toxicological screening using toxicant signatures to include all expressed gene sequences.
In one embodiment, the toxicity of a test compound is assessed by treating a biological sample containing nucleic acids with the test compound. Nucleic acids that are expressed in the treated biological sample are hybridized with one or more probes specific to the polynucleotides of the present invention, so that transcript levels corresponding to the polynucleotides of the present invention may be quantified. The transcript levels in the treated biological sample are compared with levels in an untreated biological sample. Differences in the transcript levels between the two samples are indicative of a toxic response caused by the test compound in the treated sample. Another particular embodiment relates to the use of the polypeptide sequences of the present invention to analyze the proteome of a tissue or cell type. The term proteome refers to the global pattern of protein expression in a particular tissue or cell type. Each protein component of a proteome can be subjected individually to further analysis. Proteome expression patterns, or profiles, are analyzed by quantifying the number of expressed proteins and their relative abundance under given conditions and at a given time. A profile of a cell's proteome may thus be generated by separating and analyzing the polypeptides of a particular tissue or cell type. In one embodiment, the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectric focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Steiner and Anderson, supra). The proteins are visualized in the gel as discrete and uniquely positioned spots, typically by staining the gel with an agent such as Coomassie Blue or silver or fluorescent stains. The optical density of each protein spot is generally proportional to the level of the protein in the sample. The optical densities of equivalently positioned protein spots from different samples, for example, from biological samples either treated or untreated with a test compound or therapeutic agent, are compared to identify any changes in protein spot density related to the treatment. The proteins in the spots are partially sequenced using, for , example, standard methods employing chemical or enzymatic cleavage followed by mass spectrometry. The identity of the protein in a spot may be determined by comparing its partial sequence, preferably of at least 5 contiguous amino acid residues, to the polypeptide sequences of the present invention. In some cases, further sequence data may be obtained for definitive protein identification.
A proteomic profile may also be generated using antibodies specific for INTSIG to quantify the levels of INTSIG expression. In one embodiment, the antibodies are used as elements on a microarray, and protein expression levels are quantified by exposing the microarray to the sample and detecting the levels of protein bound to each array element (Lueking, A. et al. (1999) Anal. Biochem. 270:103-111; Mendoze, L.G. et al. (1999) Biotechniques 27:778-788). Detection may be performed by a variety of methods known in the art, for example, by reacting the proteins in the sample with a thiol- or amino-reactive fluorescent compound and detecting the amount of fluorescence bound at each array element. Toxicant signatures at the proteome level are also useful for toxicological screening, and should be analyzed in parallel with toxicant signatures at the transcript level. There is a poor correlation between transcript and protein abundances for some proteins in some tissues (Anderson, N.L. and J. Seilhamer (1997) Electrophoresis 18:533-537), so proteome toxicant signatures may be useful in the analysis of compounds which do not significantly affect the transcript image, but which alter the proteomic profile. In addition, the analysis of transcripts in body fluids is difficult, due to rapid degradation of mRNA, so proteomic profiling may be more reliable and informative in such cases. In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins that are expressed in the treated biological sample are separated so that the amount of each protein can be quantified. The amount of each protein is compared to the amount of the corresponding protein in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample. Individual proteins are identified by sequencing the amino acid residues of the individual proteins and comparing these partial sequences to the polypeptides of the present invention.
In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins from the biological sample are incubated with antibodies specific to the polypeptides of the present invention. The amount of protein recognized by the antibodies is quantified. The amount of protein in the treated biological sample is compared with the amount in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample.
Microarrays may be prepared, used, and analyzed using methods known in the art. (See, e.g., Brennan, T.M. et al. (1995) U.S. Patent No. 5,474,796; Schena, M. et al. (1996) Proc. Natl. Acad. Sci. USA 93:10614-10619; Baldeschweiler et al. (1995) PCT application W095/251116; Shalon, D. et al. (1995) PCT application WO95/35505; Heller, R.A. et al. (1997) Proc. Natl. Acad. Sci. USA 94:2150-2155; and Heller, MJ. et al. (1997) U.S. Patent No. 5,605,662.) Various types of microarrays are well known and thoroughly described in DNA Microarrays: A Practical Approach, M. Schena, ed. (1999) Oxford University Press, London, hereby expressly incorporated by reference. In another embodiment of the invention, nucleic acid sequences encoding INTSIG may be used to generate hybridization probes useful in mapping the naturally occurring genomic sequence. Either coding or noncoding sequences may be used, and in some instances, noncoding sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a multi-gene family may potentially cause undesired cross hybridization during chromosomal mapping. The sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, e.g., human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial PI constructions, or single chromosome cDNA libraries. (See, e.g., Harrington, J.J. et al. (1997) Nat. Genet. 15:345-355; Price, CM. (1993) Blood Rev. 7:127-134; and Trask, B.J. (1991) Trends Genet. 7:149-154.) Once mapped, the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the inheritance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP). (See, for example, Lander, E.S. and D. Botstein (1986) Proc. Natl. Acad. Sci. USA 83:7353-7357.) Fluorescent in situ hybridization (FISH) may be correlated with other physical and genetic map data. (See, e.g., Heinz-Ulrich, et al. (1995) in Meyers, supra, pp. 965-968.) Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMDV1) World Wide Web site. Correlation between the location of the gene encoding INTSIG on a physical map and a specific disorder, or a predisposition to a specific disorder, may help define the region of DNA associated with that disorder and thus may further positional cloning efforts. In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps. Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the exact chromosomal locus is not known. This information is valuable to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the gene or genes responsible for a disease or syndrome have been crudely localized by genetic linkage to a particular genomic region, e.g., ataxia-telangiectasia to 1 lq22-23, any sequences mapping to that area may represent associated or regulatory genes for further investigation. (See, e.g., Gatti, R.A. et al. (1988) Nature 336:577-580.) The nucleotide sequence of the instant invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals.
In another embodiment of the invention, INTSIG, its catalytic or immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds in any of a variety of drug screening techniques. The fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between INTSIG and the agent being tested may be measured.
Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest. (See, e.g., Geysen, et al. (1984) PCT application WO84/03564.) In this method, large numbers of different small test compounds are synthesized on a solid substrate. The test compounds are reacted with INTSIG, or fragments thereof, and washed. Bound INTSIG is then detected by methods well known in the art. Purified INTSIG can also be coated directly onto plates for use in the aforementioned drug screening techniques. Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.
In another embodiment, one may use competitive drug screening assays in which neutralizing antibodies capable of binding INTSIG specifically compete with a test compound for binding INTSIG. In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with INTSIG.
In additional embodiments, the nucleotide sequences which encode INTSIG may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions.
Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.
The disclosures of all patents, applications and publications, mentioned above and below, are expressly incorporated by reference herein: U.S. Ser. No. 60/240,871, U.S. Ser. No. 60/244,723, U.S. Ser. No. 60/249,402, U.S. Ser. No. 60/252,622, and U.S. Ser. No. 60/255,622.
EXAMPLES I. Construction of cDNA Libraries
Incyte cDNAs were derived from cDNA libraries described in the LJ-FESEQ GOLD database (Incyte Genomics, Palo Alto CA) and shown in Table 4, column 5. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL (Life Technologies), a monophasic solution of phenol and guanidine isothiocyanate. The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated from the lysates with either isopropanol or sodium acetate and ethanol, or by other routine methods. Phenol extraction and precipitation of RNA were repeated as necessary to increase RNA purity. In some cases, RNA was treated with DNase. For most libraries, poly(A)+ RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA purification kit (QIAGEN). Alternatively, RNA was isolated directly from tissue lysates using other RNA isolation kits, e.g., the POLY(A)PURE mRNA purification kit (Ambion, Austin TX).
In some cases, Stratagene was provided with RNA and constructed the corresponding cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Life Technologies), using the recommended procedures or similar methods known in the art. (See, e.g., Ausubel, 1997, supra, units 5.1-6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300- 1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Pharmacia Biotech) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e.g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Life Technologies), PCDNA2.1 plasmid (Invitrogen, Carlsbad CA), PBK-CMV plasmid (Stratagene), PCR2-TOPOTA plasmid (Invitrogen), PCMV-ICIS plasmid (Stratagene), pIGEN (Incyte Genomics, Palo Alto CA), or pTNCY (Incyte Genomics), or derivatives thereof. Recombinant plasmids were transformed into competent E. coli cells including XLl-Blue, XLl-BlueMRF, or SOLR from Stratagene or DH5α, DH10B, or ElectroMAX DH10B from Life Technologies.
II. Isolation of cDNA Clones
Plasmids obtained as described in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an AGTC Miniprep purification kit (Edge Biosystems, Gaithersburg MD); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid purification systems or the R.E.A.L. PREP 96 plasmid purification kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4°C
Alternatively, plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao, V.B. (1994) Anal. Biochem. 216:1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).
III. Sequencing and Analysis
Incyte cDNA recovered in plasmids as described in Example II were sequenced as follows. Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (Applied Biosystems) thermal cycler or the PTC-200 thermal cycler (MJ Research) in conjunction with the HYDRA microdispenser (Robbins Scientific) or the MICROLAB 2200 (Hamilton) liquid transfer system. cDNA sequencing reactions were prepared using reagents provided by Amersham Pharmacia Biotech or supplied in ABI sequencing kits such as the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems). Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were carried out using the MEGABACE 1000 DNA sequencing system (Molecular Dynamics); the ABI PRISM 373 or 377 sequencing system (Applied Biosystems) in conjunction with standard ABI protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, 1997, supra, unit 7.7). Some of the cDNA sequences were selected for extension using the techniques disclosed in Example VDI.
The polynucleotide sequences derived from Incyte cDNAs were validated by removing vector, linker, and poly(A) sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programming, and dinucleotide nearest neighbor analysis. The Incyte cDNA sequences or translations thereof were then queried against a selection of public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM, and hidden Markov model (HMM)-based protein family databases such as PFAM. (HMM is a probabilistic approach which analyzes consensus primary structures of gene families. See, for example, Eddy, S.R. (1996) Curr. Opin. Struct. Biol. 6:361-365.) The queries were performed using programs based on BLAST, FASTA, BLIMPS, and HMMER. The Incyte cDNA sequences were assembled to produce full length polynucleotide sequences. Alternatively, GenBank cDNAs, GenBank ESTs, stitched sequences, stretched sequences, or Genscan-predicted coding sequences (see Examples TV and V) were used to extend Incyte cDNA assemblages to full length. Assembly was performed using programs based on Phred, Phrap, and Consed, and cDNA assemblages were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA. The full length polynucleotide sequences were translated to derive the corresponding full length polypeptide sequences. Alternatively, a polypeptide of the invention may begin at any of the methionine residues of the full length translated polypeptide. Full length polypeptide sequences were subsequently analyzed by querying against databases such as the GenBank protein databases (genpept), SwissProt, BLOCKS, PRINTS, DOMO, PRODOM, Prosite, and hidden Markov model (HMM)-based protein family databases such as PFAM. Full length polynucleotide sequences are also analyzed using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA) and LASERGENE software (DNASTAR). Polynucleotide and polypeptide sequence alignments are generated using default parameters specified by the CLUSTAL algorithm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences.
Table 7 summarizes the tools, programs, and algorithms used for the analysis and assembly of Incyte cDNA and full length sequences and provides applicable descriptions, references, and threshold parameters. The first column of Table 7 shows the tools, programs, and algorithms used, the second column provides brief descriptions thereof, the third column presents appropriate references, all of which are incorporated by reference herein in their entirety, and the fourth column presents, where applicable, the scores, probability values, and other parameters used to evaluate the strength of a match between two sequences (the higher the score or the lower the probability value, the greater the identity between two sequences).
The programs described above for the assembly and analysis of full length polynucleotide and polypeptide sequences were also used to identify polynucleotide sequence fragments from SEQ DD NO:21-40. Fragments from about 20 to about 4000 nucleotides which are useful in hybridization and amplification technologies are described in Table 4, column 4.
IV. Identification and Editing of Coding Sequences from Genomic DNA
Putative intracellular signaling molecules were initially identified by running the Genscan gene identification program against public genomic sequence databases (e.g., gbpri and gbhtg). Genscan is a general-purpose gene identification program which analyzes genomic DNA sequences from a variety of organisms (See Burge, C. and S. Karlin (1997) J. Mol. Biol. 268:78-94, and Burge, C. and S. Karlin (1998) Curr. Opin. Struct. Biol. 8:346-354). The program concatenates predicted exons to form an assembled cDNA sequence extending from a methionine to a stop codon. The output of Genscan is a FASTA database of polynucleotide and polypeptide sequences. The maximum range of sequence for Genscan to analyze at once was set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode intracellular signaling molecules, the encoded polypeptides were analyzed by querying against PFAM models for intracellular signaling molecules. Potential intracellular signaling molecules were also identified by homology to Incyte cDNA sequences that had been annotated as intracellular signaling molecules. These selected Genscan-predicted sequences were then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences were then edited by comparison to the top BLAST hit from genpept to correct errors in the sequence predicted by Genscan, such as extra or omitted exons. BLAST analysis was also used to find any Incyte cDNA or public cDNA coverage of the Genscan- predicted sequences, thus providing evidence for transcription. When Incyte cDNA coverage was available, this information was used to correct or confirm the Genscan predicted sequence. Full length polynucleotide sequences were obtained by assembling Genscan-predicted coding sequences with Incyte cDNA sequences and/or public cDNA sequences using the assembly process described in Example III. Alternatively, full length polynucleotide sequences were derived entirely from edited or unedited Genscan-predicted coding sequences.
V. Assembly of Genomic Sequence Data with cDNA Sequence Data "Stitched" Sequences
Partial cDNA sequences were extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example III were mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster was analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that were subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval was present on more than one sequence in the cluster were identified, and intervals thus identified were considered to be equivalent by transitivity. For example, if an interval was present on a cDNA and two genomic sequences, then all three intervals were considered to be equivalent. This process allows unrelated but consecutive genomic sequences to be brought together, bridged by cDNA sequence. Intervals thus identified were then "stitched" together by the stitching algorithm in the order that they appear along their parent sequences to generate the longest possible sequence, as well as sequence variants. Linkages between intervals which proceed along one type of parent sequence (cDNA to cDNA or genomic sequence to genomic sequence) were given preference over linkages which change parent type (cDNA to genomic sequence). The resultant stitched sequences were translated and compared by BLAST analysis to the genpept and gbpri public databases. Incorrect exons predicted by Genscan were corrected by comparison to the top BLAST hit from genpept. Sequences were further extended with additional cDNA sequences, or by inspection of genomic DNA, when necessary. "Stretched" Sequences
Partial DNA sequences were extended to full length with an algorithm based on BLAST analysis. First, partial cDNAs assembled as described in Example πi were queried against public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases using the BLAST program. The nearest GenBank protein homolog was then compared by BLAST analysis to either Incyte cDNA sequences or GenScan exon predicted sequences described in Example IV. A chimeric protein was generated by using the resultant high-scoring segment pairs (HSPs) to map the translated sequences onto the GenBank protein homolog. Insertions or deletions may occur in the chimeric protein with respect to the original GenBank protein homolog. The GenBank protein homolog, the chimeric protein, or both were used as probes to search for homologous genomic sequences from the public human genome databases. Partial DNA sequences were therefore "stretched" or extended by the addition of homologous genomic sequences. The resultant stretched sequences were examined to determine whether it contained a complete gene. VI. Chromosomal Mapping of INTSIG Encoding Polynucleotides The sequences which were used to assemble SEQ DD NO:21-40 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith-Waterman algorithm. Sequences from these databases that matched SEQ DD NO:21-40 were assembled into clusters of contiguous and overlapping sequences using assembly algorithms such as Phrap (Table 7). Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Genethon were used to determine if any of the clustered sequences had been previously mapped. Inclusion of a mapped sequence in a cluster resulted in the assignment of all sequences of that cluster, including its particular SEQ DD NO:, to that map location. Map locations are represented by ranges, or intervals, of human chromosomes. The map position of an interval, in centiMorgans, is measured relative to the terminus of the chromosome's p- arm. (The centiMorgan (cM) is a unit of measurement based on recombination frequencies between chromosomal markers. On average, 1 cM is roughly equivalent to 1 megabase (Mb) of DNA in humans, although this can vary widely due to hot and cold spots of recombination.) The cM distances are based on genetic markers mapped by Genethon which provide boundaries for radiation hybrid markers whose sequences were included in each of the clusters. Human genome maps and other resources available to the public, such as the NCBI "GeneMap'99" World Wide Web site (http://www.ncbi.nlm.nih.gov/genemap/), can be employed to determine if previously identified disease genes map within or in proximity to the intervals indicated above. In this manner, SEQ DD NO:38 was mapped to chromosome 7 within the interval from 112.90 to 113.40 centiMorgans. VII. Analysis of Polynucleotide Expression
Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (See, e.g., Sambrook, supra, ch. 7; Ausubel (1995) supra, ch. 4 and 16.)
Analogous computer techniques applying BLAST were used to search for identical or related molecules in cDNA databases such as GenBank or LIFESEQ (Incyte Genomics). This analysis is much faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or similar. The basis of the search is the product score, which is defined as: BLAST Score x Percent Identity 5 x minimum {length(Seq. 1), length(Seq. 2)}
The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. The product score is a normalized value between 0 and 100, and is calculated as follows: the BLAST score is multiplied by the percent nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences). The BLAST score is calculated by assigning a score of +5 for every base that matches in a high-scoring segment pair (HSP), and -4 for every mismatch. Two sequences may share more than one HSP (separated by gaps). If there is more than one HSP, then the pair with the highest BLAST score is used to calculate the product score. The product score represents a balance between fractional overlap and quality in a BLAST alignment. For example, a product score of 100 is produced only for 100% identity over the entire length of the shorter of the two sequences being compared. A product score of 70 is produced either by 100% identity and 70% overlap at one end, or by 88% identity and 100% overlap at the other. A product score of 50 is produced either by 100% identity and 50% overlap at one end, or 79% identity and 100% overlap.
Alternatively, polynucleotide sequences encoding INTSIG are analyzed with respect to the tissue sources from which they were derived. For example, some full length sequences are assembled, at least in part, with overlapping Incyte cDNA sequences (see Example ID). Each cDNA sequence is derived from a cDNA library constructed from a human tissue. Each human tissue is classified into one of the following organ/tissue categories: cardiovascular system; connective tissue; digestive system; embryonic structures; endocrine system; exocrine glands; genitalia, female; genitalia, male; germ cells; hemic and immune system; liver; musculoskeletal system; nervous system; pancreas; respiratory system; sense organs; skin; stomatognathic system; unclassified/mixed; or urinary tract. The number of libraries in each category is counted and divided by the total number of libraries across all categories. Similarly, each human tissue is classified into one of the following disease/condition categories: cancer, cell line, developmental, inflammation, neurological, trauma, cardiovascular, pooled, and other, and the number of libraries in each category is counted and divided by the total number of libraries across all categories. The resulting percentages reflect the tissue- and disease-specific expression of cDNA encoding INTSIG. cDNA sequences and cDNA library/tissue information are found in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA). In particular, SEQ DD NO:30 shows a strong association with neurological tissues. 1292 libraries present in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA) isolated from 20 tissue types were examined. SEQ ID NO:30 was found in 73 libraries, 43 (59%) of which were isolated from neurological tissues. Of 113 incidences of SEQ DD NO:30 in all libraries, 75 were in nervous system libraries. SEQ IN NO:30 is useful for distinguishing between nervous tissues and, for example, cardiovascular or endocrine tissues. VIII. Extension of INTSIG Encoding Polynucleotides Full length polynucleotide sequences were also produced by extension of an appropriate fragment of the full length molecule using oligonucleotide primers designed from this fragment. One primer was synthesized to initiate 5' extension of the known fragment, and the other primer was synthesized to initiate 3' extension of the known fragment. The initial primers were designed using OLIGO 4.06 software (National Biosciences), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68 °C to about 72 °C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed. High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the PTC-200 thermal cycler (MJ Research, Inc.). The reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg2+, (NH4)2S04, and 2-mercaptoethanol, Taq DNA polymerase (Amersham Pharmacia Biotech), ELONGASE enzyme (Life Technologies), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B: Step 1 : 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 68 °C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68 °C, 5 min; Step 7: storage at 4°C In the alternative, the parameters for primer pair T7 and SK+ were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C The concentration of DNA in each well was determined by dispensing 100 μl PICOGREEN quantitation reagent (0.25% (v/v) PICOGREEN; Molecular Probes, Eugene OR) dissolved in IX TE and 0.5 μl of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton MA), allowing the DNA to bind to the reagent. The plate was scanned in a Fluoroskan II (Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 μl to 10 μl aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose gel to determine which reactions were successful in extending the sequence.
The extended nucleotides were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC 18 vector (Amersham Pharmacia Biotech). For shotgun sequencing, the digested nucleotides were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with Agar ACE (Promega). Extended clones were religated using T4 ligase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Pharmacia Biotech), treated with Pfu DNA polymerase (Stratagene) to fill-in restriction site overhangs, and transfected into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37 °C in 384- well plates in LB/2x carb liquid media.
The cells were lysed, and DNA was amplified by PCR using Taq DNA polymerase (Amersham Pharmacia Biotech) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 72°C, 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6: 72°C, 5 min; Step 7: storage at 4°C DNA was quantified by PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions as described above. Samples were diluted with 20% dimethysulfoxide (1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT kit (Amersham Pharmacia Biotech) or the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems).
In like manner, full length polynucleotide sequences are verified using the above procedure or are used to obtain 5 'regulatory sequences using the above procedure along with oligonucleotides designed for such extension, and an appropriate genomic library. IX. Labeling and Use of Individual Hybridization Probes
Hybridization probes derived from SEQ DD NO:21-40 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments. Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 software (National Biosciences) and labeled by combining 50 p ol of each oligomer, 250 Ci of [γ-32P] adenosine rriphosphate (Amersham Pharmacia Biotech), and T4 polynucleotide kinase (DuPont NEN, Boston MA). The labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Pharmacia Biotech). An aliquot containing 107 counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl D, Eco RI, Pst I, Xba I, or Pvu II (DuPont NEN).
The DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham NH). Hybridization is carried out for 16 hours at 40 °C. To remove nonspecific signals, blots are sequentially washed at room temperature under conditions of up to, for example, 0.1 x saline sodium citrate and 0.5% sodium dόdecyl sulfate. Hybridization patterns are visualized using autoradiography or an alternative imaging means and compared. X. Microarrays
The linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectric printing (ink-jet printing, See, e.g., Baldeschweiler, supra.), mechanical microspotting technologies, and derivatives thereof. The substrate in each of the aforementioned technologies should be uniform and solid with a non-porous surface (Schena (1999), supra). Suggested substrates include silicon, silica, glass slides, glass chips, and silicon wafers. Alternatively, a procedure analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures. A typical array may be produced using available methods and machines well known to those of ordinary skill in the art and may contain any appropriate number of elements. (See, e.g., Schena, M. et al. (1995) Science 270:467-470; Shalon, D. et al. (1996) Genome Res. 6:639-645; Marshall, A. and J. Hodgson (1998) Nat. Biotechnol. 16:27-31.)
Full length cDNAs, Expressed Sequence Tags (ESTs), or fragments or oligomers thereof may comprise the elements of the microarray. Fragments or oligomers suitable for hybridization can be selected using software well known in the art such as LASERGENE software (DNASTAR). The array elements are hybridized with polynucleotides in a biological sample. The polynucleotides in the biological sample are conjugated to a fluorescent label or other molecular tag for ease of detection. After hybridization, nonhybridized nucleotides from the biological sample are removed, and a fluorescence scanner is used to detect hybridization at each array element. Alternatively, laser desorbtion and mass spectrometry may be used for detection of hybridization. The degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed. In one embodiment, microarray preparation and usage is described in detail below. Tissue or Cell Sample Preparation
Total RNA is isolated from tissue samples using the guanidinium thiocyanate method and poly(A)+ RNA is purified using the oligo-(dT) cellulose method. Each poly(A)+ RNA sample is reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/μl oligo-(dT) primer (21mer), IX first strand buffer, 0.03 units/μl RNase inhibitor, 500 μM dATP, 500 μM dGTP, 500 μM dTTP, 40 μM dCTP, 40 μM dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Pharmacia Biotech). The reverse transcription reaction is performed in a 25 ml volume containing 200 ng poly(A)+ RNA with GEMBRIGHT kits (Incyte). Specific control poly(A)+ RNAs are synthesized by in vitro transcription from non-coding yeast genomic DNA. After incubation at 37° C for 2 hr, each reaction sample (one with Cy3 and another with Cy5 labeling) is treated with 2.5 ml of 0.5M sodium hydroxide and incubated for 20 minutes at 85° C to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (CLONTECH Laboratories, Inc. (CLONTECH), Palo Alto CA) and after combining, both reaction samples are ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The sample is then dried to completion using a SpeedVAC (Savant Instruments Inc., Holbrook NY) and resuspended in 14 μl 5X SSC/0.2% SDS. Microarray Preparation
Sequences of the present invention are used to generate array elements. Each array element is amplified from bacterial cells containing vectors with cloned cDNA inserts. PCR amplification uses primers complementary to the vector sequences flanking the cDNA insert. Array elements are amplified in thirty cycles of PCR from an initial quantity of 1-2 ng to a final quantity greater than 5 μg. Amplified array elements are then purified using SEPHACRYL-400 (Amersham Pharmacia Biotech). Purified array elements are immobilized on polymer-coated glass slides. Glass microscope slides (Corning) are cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments. Glass slides are etched in 4% hydrofluoric acid (VWR Scientific Products Corporation (VWR), West Chester PA), washed extensively in distilled water, and coated with 0.05% aminopropyl silane (Sigma) in 95% ethanol. Coated slides are cured in a 110°C oven.
Array elements are applied to the coated glass substrate using a procedure described in U.S. Patent No. 5,807,522, incorporated herein by reference. 1 μl of the array element DNA, at an average concentration of 100 ng/μl, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 nl of array element sample per slide.
Microarrays are UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene). Microarrays are washed at room temperature once in 0.2% SDS and three times in distilled water. Non-specific binding sites are blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (PBS) (Tropix, Inc., Bedford MA) for 30 minutes at 60° C followed by washes in 0.2% SDS and distilled water as before. Hybridization
Hybridization reactions contain 9 μl of sample mixture consisting of 0.2 μg each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer. The sample mixture is heated to 65° C for 5 minutes and is aliquoted onto the microarray surface and covered with an 1.8 cm2 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 5X SSC in a corner of the chamber. The chamber containing the arrays is incubated for about 6.5 hours at 60° C. The arrays are washed for 10 min at 45° C in a first wash buffer (IX SSC, 0.1% SDS), three times for 10 minutes each at 45° C in a second wash buffer (0.1X SSC), and dried. Detection
Reporter-labeled hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Inc., Santa Clara CA) 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 20X microscope objective (Nikon, Inc., Melville NY). The slide containing the array is placed on a computer-controlled X-Y stage on the microscope and raster- scanned past the objective. The 1.8 cm x 1.8 cm array used in the present example is scanned with a resolution of 20 micrometers.
In two separate scans, a mixed gas multiline laser excites the two fluorophores sequentially. Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477,
Hamamatsu Photonics Systems, Bridgewater NJ) 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. Each array is typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus is capable of recording the spectra from both fluorophores simultaneously. The sensitivity of the scans is typically calibrated using the signal intensity generated by a cDNA control species added to the sample mixture at a known concentration. 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. When two samples from different sources (e.g., representing test and control cells), each labeled with a different fluorophore, are hybridized to a single array for the purpose of identifying genes that are differentially expressed, the calibration is done by labeling samples of the calibrating cDNA with the two fluorophores and adding identical amounts of each to the hybridization mixture.
The output of the photomultiplier tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Inc., Norwood MA) 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 each fluorophore's emission spectrum.
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 gene expression analysis program (Incyte). XL Complementary Polynucleotides
Sequences complementary to the INTSIG-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring INTSIG. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of INTSIG. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the INTSIG-encoding transcript. XII. Expression of INTSIG
Expression and purification of INTSIG is achieved using bacterial or virus-based expression systems. For expression of INTSIG in bacteria, cDNA is subcloned into an appropriate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcription. Examples of such promoters include, but are not limited to, the trp-lac (tac) hybrid promoter and the T5 or T7 bacteriophage promoter in conjunction with the lac operator regulatory element. Recombinant vectors are transformed into suitable bacterial hosts, e.g., BL21(DE3). Antibiotic resistant bacteria express INTSIG upon induction with isopropyl beta-D- thiogalactopyranoside (IPTG). Expression of INTSIG in eukaryotic cells is achieved by infecting insect or mammalian cell lines with recombinant Autographica calif ornica nuclear polyhedrosis virus (AcMNPV), commonly known as baculovirus. The nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding INTSIG by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates. Viral infectivity is maintained and the strong polyhedrin promoter drives high levels of cDNA transcription. Recombinant baculovirus is used to infect Spodoptera frugiperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases. Infection of the latter requires additional genetic modifications to baculovirus. (See Engelhard, E.K. et al. (1994) Proc. Natl. Acad. Sci. USA 91:3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7:1937-1945.)
In most expression systems, INTSIG is synthesized as a fusion protein with, e.g., glutathione S-transferase (GST) or a peptide epitope tag, such as FLAG or 6-His, permitting rapid, single-step, affinity-based purification of recombinant fusion protein from crude cell lysates. GST, a 26-kilodalton enzyme from Schistosoma iaponicum, enables the purification of fusion proteins on immobilized glutathione under conditions that maintain protein activity and antigenicity (Amersham Pharmacia Biotech). Following purification, the GST moiety can be proteolytically cleaved from INTSIG at specifically engineered sites. FLAG, an 8-amino acid peptide, enables immunoaffinity purification using commercially available monoclonal and polyclonal anti-FLAG antibodies (Eastman Kodak). 6- His, a stretch of six consecutive histidine residues, enables purification on metal-chelate resins (QIAGEN). Methods for protein expression and purification are discussed in Ausubel (1995, supra, ch. 10 and 16). Purified INTSIG obtained by these methods can be used directly in the assays shown in Examples XVI, XVϋ, and XVDI, where applicable. XIII. Functional Assays
INTSIG function is assessed by expressing the sequences encoding INTSIG at physiologically elevated levels in mammalian cell culture systems. cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA expression. Vectors of choice include PCMV SPORT (Life Technologies) and PCR3.1 (Invitrogen, Carlsbad CA), both of which contain the cytomegalovirus promoter. 5-10 μg of recombinant vector are transiently transfected into a human cell line, for example, an endothelial or hematopoietic cell line, using either liposome formulations or elecrroporation. 1-2 μg of an additional plasmid containing sequences encoding a marker protein are co-transfected. Expression of a marker protein provides a means to distinguish transfected cells from nontransfected cells and is a reliable predictor of cDNA expression from the recombinant vector. Marker proteins of choice include, e.g., Green Fluorescent Protein (GFP; Clontech), CD64, or a CD64-GFP fusion protein. Flow cytometry (FCM), an automated, laser optics- based technique, is used to identify transfected cells expressing GFP or CD64-GFP and to evaluate the apoptotic state of the cells and other cellular properties. FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death. These events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide; changes in cell size and granularity as measured by forward light scatter and 90 degree side light scatter; down-regulation of DNA synthesis as measured by decrease in bromodeoxyuridine uptake; alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell surface. Methods in flow cytometry are discussed in Ormerod, M.G. (1994) Flow Cytometry, Oxford, New York NY.
The influence of INTSIG on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding INTSIG and either CD64 or CD64-GFP. CD64 and CD64-GFP are expressed on the surface of transfected cells and bind to conserved regions of human immunoglobulin G (IgG). Transfected cells are efficiently separated from nontransfected cells using magnetic beads coated with either human IgG or antibody against CD64 (DYNAL, Lake Success NY). mRNA can be purified from the cells using methods well known by those of skill in the art. Expression of mRNA encoding INTSIG and other genes of interest can be analyzed by northern analysis or microarray techniques.
XIV. Production of INTSIG Specific Antibodies
INTSIG substantially purified using polyacrylamide gel electrophoresis (PAGE; see, e.g., Harrington, M.G. (1990) Methods Enzymol. 182:488-495), or other purification techniques, is used to immunize rabbits and to produce antibodies using standard protocols.
Alternatively, the INTSIG amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Methods for selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions are well described in the art. (See, e.g., Ausubel, 1995, supra, ch. 11.)
Typically, oligopeptides of about 15 residues in length are synthesized using an ABI 431 A peptide synthesizer (Applied Biosystems) using FMOC chemistry and coupled to KLH (Sigma- Aldrich, St. Louis MO) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) to increase immunogenicity. (See, e.g., Ausubel, 1995, supra.) Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide and anti-INTSIG activity by, for example, binding the peptide or INTSIG to a substrate, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG.
XV. Purification of Naturally Occurring INTSIG Using Specific Antibodies Naturally occurring or recombinant INTSIG is substantially purified by immunoaffinity chromatography using antibodies specific for INTSIG. An immunoaffinity column is constructed by covalently coupling anti-INTSIG antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions. Media containing INTSIG are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of INTSIG (e.g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/TNTSIG binding (e.g., a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and INTSIG is collected. XVI. Identification of Molecules Which Interact with INTSIG
INTSIG, or biologically active fragments thereof, are labeled with 125I Bolton-Hunter reagent. (See, e.g., Bolton, A.E. and W.M. Hunter (1973) Biochem. J. 133:529-539.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled INTSIG, washed, and any wells with labeled INTSIG complex are assayed. Data obtained using different concentrations of INTSIG are used to calculate values for the number, affinity, and association of INTSIG with the candidate molecules.
Alternatively, molecules interacting with INTSIG are analyzed using the yeast two-hybrid system as described in Fields, S. and O. Song (1989) Nature 340:245-246, or using commercially available kits based on the two-hybrid system, such as the MATCHMAKER system (Clontech).
INTSIG may also be used in the PATHCALLING process (CuraGen Corp., New Haven CT) which employs the yeast two-hybrid system in a high-throughput manner to determine all interactions between the proteins encoded by two large libraries of genes (Nandabalan, K. et al. (2000) U.S. Patent No. 6,057,101). XVII. Demonstration of INTSIG Activity
INTSIG activity is associated with its ability to form protein-protein complexes and is measured by its ability to regulate growth characteristics of NDT3T3 mouse fibroblast cells. A cDNA encoding INTSIG is subcloned into an appropriate eukaryotic expression vector. This vector is transfected into NIH3T3 cells using methods known in the art. Transfected cells are compared with non-transfected cells for the following quantifiable properties: growth in culture to high density, reduced attachment of cells to the substrate, altered cell morphology, and ability to induce tumors when injected into immunodeficient mice. The activity of INTSIG is proportional to the extent of increased growth or frequency of altered cell morphology in NIH3T3 cells transfected with INTSIG. Alternatively, INTSIG activity is measured by binding of INTSIG to radiolabeled for in polypeptides containing the proline-rich region that specifically binds to SH3 containing proteins (Chan, D.C et al. (1996) EMBO J. 15:1045-1054). Samples of INTSIG are run on SDS-PAGE gels, and transferred onto nitrocellulose by electroblotting. The blots are blocked for 1 hr at room temperature in TBST (137 mM NaCl, 2.7 mM KC1, 25 mM Tris (pH 8.0) and 0.1% Tween-20) containing non-fat dry milk. Blots are then incubated with TBST containing the radioactive formin polypeptide for 4 hrs to overnight. After washing the blots four times with TBST, the blots are exposed to autoradiographic film. Radioactivity is quantitated by cutting out the radioactive spots and counting them in a radioisotope counter. The amount of radioactivity recovered is proportional to the activity of INTSIG in the assay.
Alternatively, INTSIG protein kinase activity is measured by quantifying the phosphorylation of an appropriate substrate in the presence of gamma-labeled 32P-ATP. INTSIG is incubated with the substrate, 32P-ATP, and an appropriate kinase buffer. The 32P incorporated into the product is separated from free 32P-ATP by electrophoresis, and the incorporated 32P is quantified using a beta radioisotope counter. The amount of incorporated 3 P is proportional to the protein kinase activity of INTSIG in the assay. A determination of the specific amino acid residue phosphorylated by protein kinase activity is made by phosphoamino acid analysis of the hydrolyzed protein.
Alternatively, an assay for INTSIG protein phosphatase activity measures the hydrolysis of para-nitrophenyl phosphate (PNPP). INTSIG is incubated together with PNPP in HEPES buffer pH 7.5, in the presence of 0.1% β-mercaptoethanol at 37°C for 60 min. The reaction is stopped by the addition of 6 ml of 10 N NaOH, and the increase in light absorbance of the reaction mixture at 410 nm resulting from the hydrolysis of PNPP is measured using a spectrophotometer. The increase in light absorbance is proportional to the activity of INTSIG in the assay (Diamond, R.H. et al. (1994) Mol. Cell Biol. 14:3752-3762).
An alternative assay measures INTSIG-mediated G-protein signaling activity by monitoring the mobilization of Ca2+ as an indicator of the signal transduction pathway stimulation. (See, e.g., Grynkiewicz, G. et al. (1985) J. Biol. Chem. 260:3440; McColl, S. et al. (1993) J. Immunol. 150:4550-4555; and Aussel, C et al. (1988) J. Immunol. 140:215-220). The assay requires preloading neutrophils or T cells with a fluorescent dye such as FURA-2 or BCECF (Universal Imaging Corp, Westchester PA) whose emission characteristics are altered by Ca++ binding. When the cells are exposed to one or more activating stimuli artificially (e.g., anti-CD3 antibody ligation of the T cell receptor) or physiologically (e.g., by allogeneic stimulation), Ca++ flux takes place. This flux can be observed and quantified by assaying the cells in a fluorometer or fluorescent activated cell sorter. Measurements of Ca** flux are compared between cells in their normal state and those transfected with INTSIG. Increased Ca++ mobilization attributable to increased INTSIG concentration is proportional to INTSIG activity.
Alternatively, INTSIG activity is measured by binding of INTSIG to a substrate which recognizes WD-40 repeats, such as ElonginB, by coimmunoprecipitation (Kamura, T. et al. (1998) Genes Dev. 12:3872-3881). Briefly, epitope tagged substrate and INTSIG are mixed and immunoprecipitated with commercial antibody against the substrate tag. The reaction solution is run on SDS-PAGE and the presence of INTSIG visualized using an antibody to the INTSIG tag. Substrate binding is proportional to INTSIG activity.
Alternatively, INTSIG activity is measured by measuring oxysterol binding. Epitope-tagged INTSIG is incubated with a radio-labeled oxysterol ligand, such as 3H-25-hydroxycholesterol. INTSIG is collected by immunoprecipitation with a commercial antibody against the epitope, and bound hydroxycholesterol quantitated by scintillation counting. INTSIG activity is proportional to the amount of ligand bound.
XVIII. Assay to Detect INTSIG Binding to RNA
The binding of INTSIG to RNA can be assayed using a solid phase RNA binding assay. Hemagglutinin- (HA) tagged wild type and mutant INTSIG in pcDNA3 are transiently transfected into COS cells using LipofectAMINE reagent (Life Technologies, Inc.) for expression and analysis of RNA binding to multiple, simutaneously purified INTSIG proteins. Anti-HA immunoprecipitated INTSIG bound to protein G-Sepharose is incubated with 30 ng of 32P-labeled G8-5 RNA in 30 μl of RNA binding buffer containing 1 μg/μl poly(C) at room temperature for 20 min. with occasional shaking. The beads are then washed twice with 700 μl of RNA binding buffer and resuspended in 20 μl of SDS-polyacrylamide gel electrophoresis sample buffer. The protein and RNA were separated by 10% SDS-polyacrylamdie gel electrophoresis. The RNA bands ran with a mobility equivalent to 25-35 kDa, and this part of the gel is cut out and dried for autoradiography. The upper part of the gel is transferred to a polyvinylidene difluoride membrane and blotted with anti-HA antibody to detect HA-INTSIG (Lin, et al. (1997) J. Biol. Chem. 272:27274-27280).
Various modifications and variations of the described methods and systems 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 certain 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 which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.
Table 1
Figure imgf000088_0001
Table 2
Figure imgf000089_0001
Table 2 (cont.)
Figure imgf000090_0001
Table 3
Figure imgf000091_0001
Table 3 (cont.)
Figure imgf000092_0001
Table 3 (cont.)
Figure imgf000093_0001
Table 3 (cont.)
Figure imgf000094_0001
Table 3 (cont.)
Figure imgf000095_0001
Table 3 (cont.)
Figure imgf000096_0001
Table 3 (cont.)
Figure imgf000097_0001
Table 3 (cont.)
Figure imgf000098_0001
Figure imgf000098_0002
Table 4
£> 0
Figure imgf000099_0001
Table 4 (cont.)
Figure imgf000100_0001
Table 4 (cont.)
o o
Figure imgf000101_0001
Table 4 (cont.)
Figure imgf000102_0001
Table 4 (cont.)
Figure imgf000103_0001
Table 5
o
Table 6
Figure imgf000105_0001
Table 6 (cont.)
Figure imgf000106_0001
Table 6 (cont.)
Figure imgf000107_0001
Table 6 (cont.)
Figure imgf000108_0001
Table 7
Program Description Reference Parameter Threshold
ABI FACTURA A program that removes vector sequences and Applied Biosystems, Foster City, CA. masks ambiguous bases in nucleic acid sequences.
ABI/PARACEL FDF A Fast Data Finder useful in comparing and Applied Biosystems, Foster City, CA; Mismatch <50% annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA.
ABI AutoAssembler A program that assembles nucleic acid sequences. Applied Biosystems, Foster City, CA.
BLAST A Basic Local Alignment Search Tool useful in Altschul, S.F. et al. (1990) J. Mol. Biol. ESTs: Probability value= 1.0E-8 sequence similarity search for amino acid and 215:403-410; Altschul, S.F. et al. (1997) or less nucleic acid sequences. BLAST includes five Nucleic Acids Res. 25:3389-3402. Full Length sequences: Probabilit functions: blastp, blastn, blasts, tblastn, and tblastx. valuer l.OE-10 or less
FASTA A Pearson and Lipman algorithm that searches for Pearson, W.R. and D J. Lipman (1988) Proc. ESTs: fasta E value=1.06E-6
O oo similarity between a query sequence and a group of Natl. Acad Sci. USA 85:2444-2448; Pearson, Assembled ESTs: fasta Identity= sequences of the same type. FASTA comprises as W.R. (1990) Methods Enzymol. 183:63-98; 95% or greater and least five functions: fasta, tfasta, fastx, tfastx, and and Smith, T.F. and M.S. Waterman (1981) Match length=200 bases or greate ssearch. Adv. Appl. Math. 2:482-489. fastx E value=1.0E-8 or less
Full Length sequences: fastx score=100 or greater
BLIMPS A BLocks IMProved Searcher that matches a Henikoff, S. and J.G. Henikoff (1991) Nucleic Probability value= 1.0E-3 or less sequence against those in BLOCKS, PRINTS, Acids Res. 19:6565-6572; Henikoff, J.G. and DOMO, PRODOM, and PFAM databases to search S. Henikoff (1996) Methods Enzymol. for gene families, sequence homology, and 266:88-105; and Attwood, T.K. et al. (1997) J. structural fingerprint regions. Chem. Inf. Comput. Sci. 37:417-424.
HMMER An algorithm for searching a query sequence against Krogh, A. et al. (1994) J. Mol. Biol. PFAM hits: Probability value= hidden Markov model (HMM)-based databases of 235:1501-1531; Sonnhammer, E.L.L. et al. 1.0E-3 or less protein family consensus sequences, such as PFAM. (1988) Nucleic Acids Res. 26:320-322; Signal peptide hits: Score= 0 or Durbin, R. et al. (1998) Our World View, in a greater Nutshell, Cambridge Univ. Press, pp. 1-350.
Table 7 (cont.)
Program Description Reference Parameter Threshold
ProfileScan An algorithm that searches for structural and sequence Gribskov, M. et al. (1988) CABIOS 4:61-66; Normalized quality score≥GCG motifs in protein sequences that match sequence patterns Gribskov, M. et al. (1989) Methods Enzymol. specified "HIGH" value for that defined in Prosite. 183:146-159; Bairoch, A. et al. (1997) particular Prosite motif. Nucleic Acids Res. 25:217-221. Generally, score=l.4-2.1.
Phred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res. sequencer traces with high sensitivity and probability. 8:175-185; Ewing, B. and P. Green (1998) Genome Res. 8:186-194.
Phrap A Phils Revised Assembly Program including SWAT and Smith, T.F. and M.S. Waterman (1981) Adv. Score= 120 or greater; CrossMatch, programs based on efficient implementation Appl. Math. 2:482-489; Smith, T.F. and M.S. Match length= 56 or greater of the Smith-Waterman algorithm, useful in searching Waterman (1981) J. Mol. Biol. 147:195-197; sequence homology and assembling DNA sequences. and Green, P., University of Washington, Seattle, WA.
Consed A graphical tool for viewing and editing Phrap Gordon, D. et al. (1998) Genome Res. 8:195-202. assemblies. SPScan A weight matrix analysis program that scans protein Nielson, H. et al. (1997) Protein Engineering Score=3.5 or greater sequences for the presence of secretory signal peptides. 10:1-6; Claverie, J.M. and S. Audic (1997) CABIOS 12:431-439.
TMAP A program that uses weight matrices to delineate Persson, B. and P. Argos (1994) J. Mol. Biol. transmembrane segments on protein sequences and 237:182-192; Persson, B. and P. Argos (1996) determine orientation. Protein Sci. 5:363-371.
TMHMMER A program that uses a hidden Markov model (HMM) to Sonnhammer, E.L. et al. (1998) Proc. Sixth Intl. delineate transmembrane segments on protein sequences Conf. on Intelligent Systems for Mol. Biol., and determine orientation. Glasgow et al., eds., The Am. Assoc. for Artificial Intelligence Press, Menlo Park, CA, pp. 175-182.
Motifs A program that searches amino acid sequences for patterns Bairoch, A. et al. (1997) Nucleic Acids Res. 25:217-221; that matched those defined in Prosite. Wisconsin Package Program Manual, version 9, page M51-59, Genetics Computer Group, Madison, WI.

Claims

What is claimed is:
1. An isolated polypeptide selected from the group consisting of: a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
2. An isolated polypeptide of claim 1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
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 comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO.21-40.
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 has an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
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 selected from the group consisting of SEQ ID NO:21-40, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least
90% identical to a polynucleotide sequence selected from the group consisting of SEQ
ID NO:21-40, 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 has an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
19. A method for treating a disease or condition associated with decreased expression of functional INTSIG, 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 INTSIG, 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 INTSIG, 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, the 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 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 INTSIG 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 INTSIG 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 method of diagnosing a condition or disease associated with the expression of INTSIG in a subject, comprising administering to said subject an effective amount of the composition of claim 34.
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 having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, 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 having an amino acid sequence selected from the group consisting of SEQ ID NO:1-20.
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 having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20, 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 having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20.
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 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 having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20 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 having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20 in the sample.
45. A method of purifying a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-20 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 having an amino acid sequence selected from the group consisting of SEQ ID NO:1-20.
46. A microarray wherein at least one element of the microarray is a polynucleotide of claim
13.
47. A method of generating a transcript image 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.
56. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:l.
57. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ED NO:2.
58. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:3.
59. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:4.
60. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:5.
61. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:6.
62. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:7.
63. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 8.
64. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:9.
65. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:10.
66. A polypeptide of claim 1 , comprising the amino acid sequence of SEQ ID NO: 11.
67. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:12.
68. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 13.
69. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:14.
70. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:15.
71. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:16.
72. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 17.
73. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:18.
74. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 19.
75. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO:20.
76. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:21.
77. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:22.
78. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:23.
79. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:24.
80. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:25.
81. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:26.
82. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:27.
83. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:28.
84. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:29.
85. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:30.
86. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:31.
87. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:32.
88. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:33.
89. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:34.
90. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:35.
91. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:36.
92. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:37.
93. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:38.
94. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:39.
95. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO:40.
<110> INCYTE GENOMICS, INC. BAUGHN, Mariah R. DING, Li
ELLIOTT, Vicki S. GANDHI, Ameena R. Gietzen, Kimberly J. GRIFFIN, Jennifer A. GURURAJAN, Ra agopal HAFALIA, April J.A. KEARNEY, Liam KHAN, Farrah A. LAL, Preeti LEE, Ernestine A. LU, Dyung Aina M. LU, Yan
NGUYEN, Danniel B. ARVIZU, Chandra RAMKU AR, Jaya TANG, Y. Tom THANGAVELU, Kavitha THORNTON, Michael WALIA, Narinder K. WARREN, Bridget A. XU, Yuming YAO, Monique G. YUE, Henry
<120> INSTRACELLULAR SIGNALING MOLECULES
<130> PF-0827 PCT
<140> To Be Assigned <141> Herewith
<150> 60/240,871; 60/244,723; 60/249,402; 60/252,622; 60/255,622 <151> 2000-10-13; 2000-10-30; 2000-11-15; 2000-11-22; 2000-12-13
<160> 40
<170> PERL Program
<210> 1
<211> 805
<212> PRT
<213> Ho o sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 105283CD1
<400> 1
Met Ala Ala Ser Gly Val Pro Arg Gly Cys Asp He Leu He Val
1 5 10 15
Tyr Ser Pro Asp Ala Glu Glu Trp Cys Gin Tyr Leu Gin Thr Leu
20 25 30
Phe Leu Ser Ser Arg Gin Val Arg Ser Gin Lys He Leu Thr His
35 40 45
Arg Leu Gly Pro Glu Ala Ser Phe Ser Ala Glu Asp Leu Ser Leu
50 55 60 Phe Leu Ser Thr Arg Cys Val Val Val Leu Leu Ser Ala Glu Leu
65 70 75
Val Gin His Phe His Lys Pro Ala Leu Leu Pro Leu Leu Gin Arg
80 85 90
Ala Phe His Pro Pro His Arg Val Val Arg Leu Leu Cys Gly Val
95 100 105
Arg Asp Ser Glu Glu Phe Leu Asp Phe Phe Pro Asp Trp Ala His
110 115 120
Trp Gin Glu Leu Thr Cys Asp Asp Glu Pro Glu Thr Tyr Val Ala
125 130 135
Ala Val Lys Lys Ala He Ser Glu Asp Ser Gly Cys Asp Ser Val
140 145 150
Thr Asp Thr Glu Pro Glu Asp Glu Lys Val Val Ser Tyr Ser Lys
155 160 165
Gin Gin Asn Leu Pro Thr Val Thr Ser Pro Gly Asn Leu Met Val
170 175 180
Val Gin Pro Asp Arg He Arg Cys Gly Ala Glu Thr Thr Val Tyr
185 190 195
Val He Val Arg Cys Lys Leu Asp Asp Arg Val Ala Thr Glu Ala
200 205 210
Glu Phe Ser Pro Glu Asp Ser Pro Ser Val Arg Met Glu Ala Lys
215 220 225
Val Glu Asn Glu Tyr Thr He Ser Val Lys Ala Pro Asn Leu Ser
230 235 240
Ser Gly Asn Val Ser Leu Lys He Tyr Ser Gly Asp Leu Val Val
245 250 255
Cys Glu Thr Val He Ser Tyr Tyr Thr Asp Met Glu Glu He Gly
260 265 270
Asn Leu Leu Ser Asn Ala Ala Asn Pro Val Glu Phe Met Cys Gin
275 280 285
Ala Phe Lys He Val Pro Tyr Asn Thr Glu Thr Leu Asp Lys Leu
290 295 300
Leu Thr Glu Ser Leu Lys Asn Asn He Pro Ala Ser Gly Leu His
305 310 315
Leu Phe Gly He Asn Gin Leu Glu Glu Glu Asp Met Met Thr Asn
320 325 330
Gin Arg Asp Glu Glu Leu Pro Thr Leu Leu His Phe Ala Ala Lys
335 340 345
Tyr Gly Leu Lys Asn Leu Thr Ala Leu Leu Leu Thr Cys Pro Gly
350 355 360
Ala Leu Gin Ala Tyr Ser Val Ala Asn Lys His Gly His Tyr Pro
365 370 375
Asn Thr He Ala Glu Lys His Gly Phe Arg Asp Leu Arg Gin Phe
380 385 390
He Asp Glu Tyr Val Glu Thr Val Asp Met Leu Lys Ser His He
395 400 405
Lys Glu Glu Leu Met His Gly Glu Glu Ala Asp Ala Val Tyr Glu
410 415 420
Ser Met Ala His Leu Ser Thr Asp Leu Leu Met Lys Cys Ser Leu
425 430 435
Asn Pro Gly Cys Asp Glu Asp Leu Tyr Glu Ser Met Ala Ala Phe
440 445 450
Val Pro Ala Ala Thr Glu Asp Leu Tyr Val Glu Met Leu Gin Ala
455 460 465
Ser Thr Ser Asn Pro He Pro Gly Asp Gly Phe Ser Arg Ala Thr
470 475 480
Lys Asp Ser Met He Arg Lys Phe Leu Glu Gly Asn Ser Met Gly
485 490 495 Met Thr Asn Leu Glu Arg Asp Gin Cys His Leu Gly Gin Glu Glu
500 505 510
Asp Val Tyr His Thr Val Asp Asp Asp Glu Ala Phe Ser Val Asp
515 520 525
Leu Ala Ser Arg Pro Pro Val Pro Val Pro Arg Pro Glu Thr Thr
530 535 540
Ala Pro Gly Ala His Gin Leu Pro Asp Asn Glu Pro Tyr He Phe
545 550 555
Lys Val Phe Ala Glu Lys Ser Gin Glu Arg Pro Gly Asn Phe Tyr
560 565 570
Val Ser Ser Glu Ser He Arg Lys Gly Pro Pro Val Arg Pro Trp
575 580 585
Arg Asp Arg Pro Gin Ser Ser He Tyr Asp Pro Phe Ala Gly Met
590 595 600
Lys Thr Pro Gly Gin Arg Gin Leu He Thr Leu Gin Glu Gin Val
605 610 615
Lys Leu Gly He Val Asn Val Asp Glu Ala Val Leu His Phe Lys
620 625 630
Glu Trp Gin Leu Asn Gin Lys Arg Arg Ser Glu Ser Phe Arg Phe
635 640 645
Gin Gin Glu Asn Leu Lys Arg Leu Arg Asp Ser He Thr Arg Arg
650 655 660
Gin Arg Glu Lys Gin Lys Ser Gly Lys Gin Thr Asp Leu Glu He
665 670 675
Thr Val Pro He Arg His Ser Gin His Leu Pro Ala Lys Val Glu
680 685 690
Phe Gly Val Tyr Glu Ser Gly Pro Arg Lys Ser Val He Pro Pro
695 700 705
Arg Thr Glu Leu Arg Arg Gly Asp Trp Lys Thr Asp Ser Thr Ser
710 715 720
Ser Thr Ala Ser Ser Thr Ser Asn Arg Ser Ser Thr Arg Ser Leu
725 730 735
Leu Ser Val Ser Ser Gly Met Glu Gly Asp Asn Glu Asp Asn Glu
740 745 750
Val Pro Glu Val Thr Arg Ser Arg Ser Pro Gly Pro Pro Gin Val
755 760 765
Asp Gly Thr Pro Thr Met Ser Leu Glu Arg Pro Pro Arg Val Pro
770 775 780
Pro Arg Ala Ala Ser Gin Arg Pro Pro Thr Arg Glu Thr Phe His
785 790 795
Pro Pro Pro Pro Val Pro Pro Arg Gly Arg
800 805
<210> 2
<211> 957
<212> PRT
<213> Homo sapiens .
<220>
<221> misc_feature
<223> Incyte ID No: 3350821CD1
<400> 2
Met Cys Cys Thr Glu Gly Ser Leu Arg Lys Arg Asp Ser Gin Arg
1 5 10 15
Ala Pro Glu Ala Val Leu Cys Leu Gin Leu Trp Gin Arg Thr Val
20 25 30
Pro Leu Asp Thr Leu Lys Gly Leu Gly Thr Cys Phe Pro Ser Gly 35 40 45
Pro Glu Leu Arg Gly Ala Gly He Ala Ala Ala Met Glu Arg Ala
50 55 60
Ser Glu Arg Arg Thr Ala Ser Ala Leu Phe Ala Gly Phe Arg Ala
65 70 75
Leu Gly Leu Phe Ser Asn Asp He Pro His Val Val Arg Phe Ser
80 85 90
Ala Leu Lys Arg Arg Phe Tyr Val Thr Thr Cys Val Gly Lys Ser
95 100 105
Phe His Thr Tyr Asp Val Gin Lys Leu Ser Leu Val Ala Val Ser
110 115 120
Asn Ser Val Pro Gin Asp He Cys Cys Met Ala Ala Asp Gly Arg
125 130 135
Leu Val Phe Ala Ala Tyr Gly Asn Val Phe Ser Ala Phe Ala Arg
140 145 150
Asn Lys Glu He Val His Thr Phe Lys Gly His Lys Ala Glu He
155 160 165
His Phe Leu Gin Pro Phe Gly Asp His He He Ser Val Asp Thr
170 175 180
Asp Gly He Leu He He Trp His He Tyr Ser Glu Glu Glu Tyr
185 190 195
Leu Gin Leu Thr Phe Asp Lys Ser Val Phe Lys He Ser Ala He
200 205 210
Leu His Pro Ser Thr Tyr Leu Asn Lys He Leu Leu Gly Ser Glu
215 220 225
Gin Gly Ser Leu Gin Leu Trp Asn Val Lys Ser Asn Gin Lys Tyr
230 235 240
Pro He Arg Gin Thr Phe He Pro Ala Gly Tyr Leu Leu Gly Ala
245 250 255
His Gly Leu Lys Thr Gin Ala Pro Ala Val Asp Val Val Ala He
260 265 270
Gly Leu Met Ser Gly Gin Val He He His Asn He Lys Phe Asn
275 280 285
Glu Thr Leu Met Lys Phe Arg Gin Asp Trp Gly Pro He Thr Ser
290 295 300
He Ser Phe Arg Thr Asp Gly His Pro Val Met Ala Ala Gly Ser
305 310 315
Pro Cys Gly His He Gly Leu Trp Asp Leu Glu Asp Lys Lys Leu
320 325 330
He Asn Gin Met Arg Asn Ala His Ser Thr Ala He Ala Gly Leu
335 340 345
Thr Phe Leu His Arg Glu Pro Leu Leu Val Thr Asn Gly Ala Asp
350 355 360
Asn Ala Leu Arg He Trp He Phe Asp Gly Pro Thr Gly Glu Gly
365 370 375
Arg Leu Leu Arg Phe Arg Met Gly His Ser Ala Pro Leu Thr Asn
380 385 390
He Arg Tyr Tyr Gly Gin Asn Gly Gin Gin He Leu Ser Ala Ser
395 400 405
Gin Asp Gly Thr Leu Gin Ser Phe Ser Thr Val His Glu Lys Phe
410 415 420
Asn Lys Ser Leu Gly His Gly Leu He Asn Lys Lys Arg Val Lys
425 430 435
Arg Lys Gly Leu Gin Asn Thr Met Ser Val Arg Leu Pro Pro He
440 445 450
Thr Lys Phe Ala Ala Glu Glu Ala Arg Glu Ser Asp Trp Asp Gly
455 460 465
He He Ala Cys His Gin Gly Lys Leu Ser Cys Ser Thr Trp Asn 470 475 480
Tyr Gin Lys Ser Thr He Gly Ala Tyr Phe Leu Lys Pro Lys Glu
485 490 495
Leu Lys Lys Asp Asp He Thr Ala Thr Ala Val Asp He Thr Ser
500 505 510
Cys Gly Asn Phe Ala Val He Gly Leu Ser Ser Gly Thr Val Asp
515 520 525
Val Tyr Asn Met Gin Ser Gly He His Arg Gly Ser Phe Gly Lys
530 535 540
Asp Gin Ala His Lys Gly Ser Val Arg Gly Val Ala Val Asp Gly
545 550 555
Leu Asn Gin Leu Thr Val Thr Thr Gly Ser Glu Gly Leu Leu Lys
560 565 570
Phe Trp Asn Phe Lys Asn Lys He Leu He His Ser Val Ser Leu
575 580 585
Ser Ser Ser Pro Asn He Met Leu Leu His Arg Asp Ser Gly He
590 595 600
Leu Gly Leu Ala Leu Asp Asp Phe Ser He Ser Val Leu Asp He
605 610 615
Glu Ala Arg Lys He Val Arg Glu Phe Ser Gly His Gin Gly Gin
620 625 630
' He Asn Asp Met Ala Phe Ser Pro Asp Gly Arg Trp Leu He Ser
635 640 645
Ala Ala Met Asp Cys Ser He Arg Thr Trp Asp Leu Pro Ser Gly
650 655 660
Cys Leu He Asp Cys Phe Leu Leu Asp Ser Ala Pro Leu Asn Val
665 670 675
Ser Met Ser Pro Thr Gly Asp Phe Leu Ala Thr Ser His Val Asp
680 685 690
His Leu Gly He Tyr Leu Trp Ser Asn He Ser Leu Tyr Ser Val
695 700 705
Val Ser Leu Arg Pro Leu Pro Ala Asp Tyr Val Pro Ser He Val
710 715 720
Met Leu Pro Gly Thr Cys Gin Thr Gin Asp Val Glu Val Ser Glu
725 730 735
Glu Thr Val Glu Pro Ser Asp Glu Leu He Glu Tyr Asp Ser Pro
740 745 750
Glu Gin Leu Asn Glu Gin Leu Val Thr Leu Ser Leu Leu Pro Glu
755 760 765
Ser Arg Trp Lys Asn Leu Leu Asn Leu Asp Val He Lys Lys Lys
770 775 780
Asn Lys Pro Lys Glu Pro Pro Lys Val Pro Lys Ser Ala Pro Phe
785 790 795
Phe He Pro Thr He Pro Gly Leu Val Pro Arg Tyr Ala Ala Pro
800 805 810
Glu Gin Asn Asn Asp Pro Gin Gin Ser Lys Val Val Asn Leu Gly
815 820 825
Val Leu Ala Gin Lys Ser Asp Phe Cys Leu Lys Leu Glu Glu Gly
830 835 840
Leu Val Asn Asn Lys Tyr Asp Thr Ala Leu Asn Leu Leu Lys Glu
845 850 855
Ser Gly Pro Ser Gly He Glu Thr Glu Leu Arg Ser Leu Ser Pro
860 865 870
Asp Cys Gly Gly Ser He Glu Val Met Gin Ser Phe Leu Lys Met
875 880 885 lie Gly Met Met Leu Asp Arg Lys Arg Asp Phe Glu Leu Ala Gin
890 895 900
Ala Tyr Leu Ala Leu Phe Leu Lys Leu His Leu Lys Met Leu Pro 905 910 915
Ser Glu Pro Val Leu Leu Glu Glu He Thr Asn Leu Ser Ser Gin
920 925 930
Val Glu Glu Asn Trp Thr His Leu Gin Ser Leu Phe Asn Gin Ser
935 940 945
Met Cys He Leu Asn Tyr Leu Lys Ser Ala Leu Leu
950 955
<210> 3
<211> 274
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 5876846CD1
<400> 3
Met Val Arg Leu Thr Leu Asp Leu He Ala Arg Asn Ser Asn Leu
1 5 10 15
Lys Pro Arg Lys Glu Glu Thr He Ser Gin Cys Leu Lys Lys He
20 25 30
Thr His He Asn Phe Ser Asp Lys Asn He Asp Ala He Glu Asp
35 40 45
Leu Ser Leu Cys Lys Asn Leu Ser Val Leu Tyr Leu Tyr Asp Asn
50 55 60
Cys He Ser Gin He Thr Asn Leu Asn Tyr Ala Thr Asn Leu Thr
65 70 75
His Leu Tyr Leu Gin Asn Asn Cys He Ser Cys He Glu Asn Leu
80 85 90
Arg Ser Leu Lys Lys Leu Glu Lys Leu Tyr Leu Gly Gly Asn Tyr
95 100 105
He Ala Val He Glu Gly Leu Glu Gly Leu Gly Glu Leu Arg Glu
110 115 120
Leu His Val Glu Asn Gin Arg Leu Pro Leu Gly Glu Lys Leu Leu
125 130 135
Phe Asp Pro Arg Thr Leu His Ser Leu Ala Lys Ser Leu Cys He
140 145 150
Leu Asn He Ser Asn Asn Asn He Asp Asp He Thr Asp Leu Glu
155 160 165
Leu Leu Glu Asn Leu Asn Gin Leu He Ala Val Asp Asn Gin Leu
170 175 180
Leu His Val Lys Asp Leu Glu Phe Leu Leu Asn Lys Leu Met Lys
185 190 195
Leu Trp Lys He Asp Leu Asn Gly Asn Pro Val Cys Leu Lys Pro
200 205 210
Lys Tyr Arg Asp Arg Leu He Leu Val Ser Lys Ser Leu Glu Phe
215 220 225
Leu Asp Gly Lys Glu He Lys Asn He Glu Arg Gin Phe Leu Met
230 235 240
Asn Trp Lys Ala Ser Lys Asp Ala Lys Lys He Ser Lys Lys Arg
245 250 255
Ser Ser Lys Asn Glu Asp Ala Ser Asn Ser Leu He Ser Lys His
260 265 270 Ser Val Thr His
<210> 4 <211> 1144
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 3560269CD1
<400> 4
Met Pro Thr Ala Glu Ser Glu Ala Lys Val Lys Thr Lys Val Arg
1 5 10 15
Phe Glu Glu Leu Leu Lys Thr His Ser Asp Leu Met Arg Glu Lys
20 25 30
Lys Lys Leu Lys Lys Lys Leu Val Arg Ser Glu Glu Asn He Ser
35 40 45
Pro Asp Thr He Arg Ser Asn Leu His Tyr Met Lys Glu Thr Thr
50 55 60
Ser Asp Asp Pro Asp Thr He Arg Ser Asn Leu Pro His He Lys
65 70 75
Glu Thr Thr Ser Asp Asp Val Ser Ala Ala Asn Thr Asn Asn Leu
80 85 90
Lys Lys Ser Thr Arg Val Thr Lys Asn Lys Leu Arg Asn Thr Gin
95 100 105
Leu Ala Thr Glu Asn Pro Asn Gly Asp Ala Ser Val Glu Glu Asp
110 115 120
Lys Gin Gly Lys Pro Asn Lys Lys Val He Lys Thr Val Pro Gin
125 130 135
Leu Thr Thr Gin Asp Leu Lys Pro Glu Thr Pro Glu Asn Lys Val
140 145 150
Asp Ser Thr His Gin Lys Thr His Thr Lys Pro Gin Pro Gly Val
155 160 165
Asp His Gin Lys Ser Glu Lys Ala Asn Glu Gly Arg Glu Glu Thr
170 175 180
Asp Leu Glu Glu Asp Glu Glu Leu Met Gin Ala Tyr Gin Cys His
185 190 195
Val Thr Glu Glu Met Ala Lys Glu He Lys Arg Lys He Arg Lys
200 205 210
Lys Leu Lys Glu Gin Leu Thr Tyr Phe Pro Ser Asp Thr Leu Phe
215 220 225
His Asp Asp Lys Leu Ser Ser Glu Lys Arg Lys Lys Lys Lys Glu
230 235 240
Val Pro Val Phe Ser Lys Ala Glu Thr Ser Thr Leu Thr He Ser
245 250 255
Gly Asp Thr Val Glu Gly Glu Gin Lys Lys Glu Ser Ser Val Arg
260 265 270
Ser Val Ser Ser Asp Ser His Gin Asp Asp Glu He Ser Ser Met
275 280 285
Glu Gin Ser Thr Glu Asp Ser Met Gin Asp Asp Thr Lys Pro Lys
290 295 300
Pro Lys Lys Thr Lys Lys Lys Thr Lys Ala Val Ala Asp Asn Asn
305 310 315
Glu Asp Val Asp Gly Asp Gly Val His Glu He Thr Ser Arg Asp
320 325 330
Ser Pro Val Tyr Pro Lys Cys Leu Leu Asp Asp Asp Leu Val Leu
335 340 345
Gly Val Tyr He His Arg Thr Asp Arg Leu Lys Ser Asp Phe Met
350 355 360
He Ser His Pro Met Val Lys He His Val Val Asp Glu His Thr 365 370 375
Gly Gin Tyr Val Lys Lys Asp Asp Ser Gly Arg Pro Val Ser Ser
380 385 390
Tyr Tyr Glu Lys Glu Asn Val Asp Tyr He Leu Pro He Met Thr
395 400 405
Gin Pro Tyr Asp Phe Lys Gin Leu Lys Ser Arg Leu Pro Glu Trp
410 415 420
Glu Glu Gin He Val Phe Asn Glu Asn Phe Pro Tyr Leu Leu Arg
425 430 435
Gly Ser Asp Glu Ser Pro Lys Val He Leu Phe Phe Glu He Leu
440 445 450
Asp Phe Leu Ser Val Asp Glu He Lys Asn Asn Ser Glu Val Gin
455 460 465
Asn Gin Glu Cys Gly Phe Arg Lys He Ala Trp Ala Phe Leu Lys
470 475 480
Leu Leu Gly Ala Asn Gly Asn Ala Asn He Asn Ser Lys Leu Arg
485 490 495
Leu Gin Leu Tyr Tyr Pro Pro Thr Lys Pro Arg Ser Pro Leu Ser
500 505 510
Val Val Glu Ala Phe Glu Trp Trp Ser Lys Cys Pro Arg Asn His
515 520 525
Tyr Pro Ser Thr Leu Tyr Val Thr Val Arg Gly Leu Lys Val Pro
530 535 540
Asp Cys He Lys Pro Ser Tyr Arg Ser Met Met Ala Leu Gin Glu
545 550 555
Glu Lys Gly Lys Pro Val His Cys Glu Arg His His Glu Ser Ser
560 565 570
Ser Val Asp Thr Glu Pro Gly Leu Glu Glu Ser Lys Glu Val He
575 580 585
Lys Trp Lys Arg Leu Pro Gly Gin Ala Cys Arg He Pro Asn Lys
590 595 600
His Leu Phe Ser Leu Asn Ala Gly Glu Arg Gly Cys Phe Cys Leu
605 610 615
Asp Phe Ser His Asn Gly Arg He Leu Ala Ala Ala Cys Ala Ser
620 625 630
Arg Asp Gly Tyr Pro He He Leu Tyr Glu He Pro Ser Gly Arg
635 640 645
Phe Met Arg Glu Leu Cys Gly His Leu Asn He He Tyr Asp Leu
650 655 660
Ser Trp Ser Lys Asp Asp His Tyr He Leu Thr Ser Ser Ser Asp
665 670 675
Gly Thr Ala Arg He Trp Lys Asn Glu He Asn Asn Thr Asn Thr
680 685 690 Phe Arg Val Leu Pro His Pro Ser Phe Val Tyr Thr Ala Lys Phe
695 700 705
His Pro Ala Val Arg Glu Leu Val Val Thr Gly Cys Tyr Asp Ser
710 715 720 Met He Arg He Trp Lys Val Glu Met Arg Glu Asp Ser Ala He
725 730 735
Leu Val Arg Gin Phe Asp Val His Lys Ser Phe He Asn Ser Leu
740 745 750 Cys Phe Asp Thr Glu Gly His His Met Tyr Ser Gly Asp Cys Thr
755 760 765
Gly Val He Val Val Trp Asn Thr Tyr Val Lys He Asn Asp Leu
770 775 780
Glu His Ser Val His His Trp Thr He Asn Lys Glu He Lys Glu
785 790 795 Thr Glu Phe Lys Gly He Pro He Ser Tyr Leu Glu He His Pro 800 805 810
Asn Gly Lys Arg Leu Leu He His Thr Lys Asp Ser Thr Leu Arg
815 ' 820 825
He Met Asp Leu Arg He Leu Val Ala Arg Lys Phe Val Gly Ala
830 835 840
Ala Asn Tyr Arg Glu Lys He His Ser Thr Leu Thr Pro Cys Gly
845 850 855
Thr Phe Leu Phe Ala Gly Ser Glu Asp Gly He Val Tyr Val Trp
860 865 870
Asn Pro Glu Thr Gly Glu Gin Val Ala Met Tyr Ser Asp Leu Pro
875 880 885
Phe Lys Ser Pro He Arg Asp He Ser Tyr His Pro Phe Glu Asn
890 895 900
Met Val Ala Phe Cys Ala Phe Gly Gin Asn Glu Pro He Leu Leu
905 910 915
Tyr He Tyr Asp Phe His Val Ala Gin Gin Glu Ala Glu Met Phe
920 925 930
Lys Arg Tyr Asn Gly Thr Phe Pro Leu Pro Gly He His Gin Ser
935 940 945
Gin Asp Ala Leu Cys Thr Cys Pro Lys Leu Pro His Gin Gly Ser
950 955 960
Phe Gin He Asp Glu Phe Val His Thr Glu Ser Ser Ser Thr Lys
965 970 975
Met Gin Leu Val Lys Gin Arg Leu Glu Thr Val Thr Glu Val He
980 985 990
Arg Ser Cys Ala Ala Lys Val Asn Lys Asn Leu Ser Phe Thr Ser
995 1000 1005
Pro Pro Ala Val Ser Ser, Gin Gin Ser Lys Leu Lys Gin Ser Asn
1010 1015 1020
Met Leu Thr Ala Gin Glu He Leu His Gin Phe Gly Phe Thr Gin
1025 1030 1035
Thr Gly He He Ser He Glu Arg Lys Pro Cys Asn His Gin Val
1040 1045 1050
Asp Thr Ala Pro Thr Val Val Ala Leu Tyr Asp Tyr Thr Ala Asn
1055 1060 1065
Arg Ser Asp Glu Leu Thr He His Arg Gly Asp He He Arg Val
1070 1075 1080
Phe Phe Lys Asp Asn Glu Asp Trp Trp Tyr Gly Ser He Gly Lys
1085 1090 1095
Gly Gin Glu Gly Tyr Phe Pro Ala Asn His Val Ala Ser Glu Thr
1100 1105 1110
Leu Tyr Gin Glu Leu Pro Pro Glu He Lys Glu Arg Ser Pro Pro
1115 1120 1125
Leu Ser Pro Glu Glu Lys Thr Lys He Glu Lys Ser Pro Ala Pro
1130 1135 1140
Gin Lys Val Lys
<210> 5
<211> 513
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No : 4596874CD1
<400> 5 Met Ala Tyr Gin Val Val Glu Lys Gly Ala Ala Leu Gly Thr Leu
1 5 10 15
Glu Ser Glu Leu Gin Gin Arg Gin Ser Arg Leu Ala Ala Leu Glu
20 25 30
Ala Arg Val Ala Gin Leu Arg Glu Ala Arg Ala Gin Gin Ala Gin
35 40 45
Gin Val Glu Glu Trp Arg Ala Gin Asn Ala Val Gin Arg Ala Ala
50 55 60
Tyr Glu Ala Leu Arg Ala His Val Gly Leu Arg Glu Ala Ala Leu
65 70 75
Arg Arg Leu Gin Glu Glu Ala Arg Asp Leu Leu Glu Arg Leu Val
80 85 90
Gin Arg Lys Ala Arg Ala Ala Ala Glu Arg Asn Leu Arg Asn Glu
95 100 105
Arg Arg Glu Arg Ala Lys Gin Ala Arg Val Ser Gin Glu Leu Lys
110 115 120
Lys Ala Ala Lys Arg Thr Val Ser He Ser Glu Gly Pro Asp Thr
125 130 135
Leu Gly Asp Gly Met Arg Glu Arg Arg Glu Thr Leu Ala Leu Ala
140 145 150
Pro Glu Pro Glu Pro Leu Glu Lys Glu Ala Cys Glu Lys Trp Lys
155 160 165
Arg Pro Phe Arg Ser Ala Ser Ala Thr Ser Leu Thr Leu Ser His
170 175 180
Cys Val Asp Val Val Lys Gly Leu Leu Asp Phe Lys Lys Arg Arg
185 190 195
Gly His Ser He Gly Gly Ala Pro Glu Gin Arg Tyr Gin He He
200 205 210
Pro Val Cys Val Ala Ala Arg Leu Pro Thr Arg Ala Gin Asp Val
215 220 225
Leu Asp Ala His Leu Ser Glu Val Asn Ala Val Arg Phe Gly Pro
230 235 240
Asn Ser Ser Leu Leu Ala Thr Gly Gly Ala Asp Arg Leu He His
245 250 255
Leu Trp Asn Val Val Gly Ser Arg Leu Glu Ala Asn Gin Thr Leu
260 255 270
Glu Gly Ala Gly Gly Ser He Thr Ser Val Asp Phe Asp Pro Ser
275 280 285
Gly Tyr Gin Val Leu Ala Ala Thr Tyr Asn Gin Ala Ala Gin Leu
290 295 300
Trp Lys Val Gly Glu Ala Gin Ser Lys Glu Thr Leu Ser Gly His
305 310 315
Lys Asp Lys Val Thr Ala Ala Lys Phe Lys Leu Thr Arg His Gin
320 325 330
Ala Val Thr Gly Ser Arg Asp Arg Thr Val Lys Glu Trp Asp Leu
335 340 345
Gly Arg Ala Tyr Cys Ser Arg Thr He Asn Val Leu Ser Tyr Cys
350 355 360
Asn Asp Val Val Cys Gly Asp His He He He Ser Gly His Asn
365 370 375
Asp Gin Lys He Arg Phe Trp Asp Ser Arg Gly Pro His Cys Thr
380 385 390
Gin Val He Pro Val Gin Gly Arg Val Thr Ser Leu Ser Leu Ser
395 400 405
His Asp Gin Leu His Leu Leu Ser Cys Ser Arg Asp Asn Thr Leu
410 415 420
Lys Val He Asp Leu Arg Val Ser Asn He Arg Gin Val Phe Arg
425 430 435 Ala Asp Gly Phe Lys Cys Gly Ser Asp Trp Thr Lys Ala Val Phe
440 445 450
Ser Pro Asp Arg Ser Tyr Ala Leu Ala Gly Ser Cys Asp Gly Ala
455 460 465
Leu Tyr He Trp Asp Val Asp Thr Gly Lys Leu Glu Ser Arg Leu
470 475 480
Gin Gly Pro His Cys Ala Ala Val Asn Ala Val Ala Trp Cys Tyr
485 490 495
Ser Gly Ser His Met Val Ser Val Asp Gin Gly Arg Lys Val Val
500 505 510 Leu Trp Gin
<210> 6
<211> 667
<212> PRT
<213> Homo sapiens
<220>
<221> misσ_feature
<223> Incyte ID No: 3594012CD1
<400> 6
Met His Phe Thr Gly Ala Val Met Gin Glu Asn Leu Arg Phe Ala
1 5 10 15
Ser Ser Gly Asp Asp He Lys He Trp Asp Ala Ser Ser Met Thr
20 25 30
Leu Val Asp Lys Phe Asn Pro His Thr Ser Pro His Gly He Ser
35 ■ 40 45
Ser He Cys Trp Ser Ser Asn Asn Asn Phe Leu Val Thr Ala Ser
50 55 60
Ser Ser Gly Asp Lys He Val Val Ser Ser Cys Lys Cys Lys Pro
65 70 75
Val Pro Leu Leu Glu Leu Ala Glu Gly Gin Lys Gin Thr Cys Val
80 . 85 90
Asn Leu Asn Ser Thr Ser Met Tyr Leu Val Ser Gly Gly Leu Asn
95 100 105
Asn Thr Val Asn He Trp Asp Leu Lys Ser Lys Arg Val His Arg
110 115 120
Ser Leu Lys Asp His Lys Asp Gin Val Thr Cys Val Thr Tyr Asn
125 130 135
Trp Asn Asp Cys Tyr He Ala Ser Gly Ser Leu Ser Gly Glu He
140 145 150
He Leu His Ser Val Thr Thr Asn Leu Ser Ser Thr Pro Phe Gly
155 160 165
His Gly Ser Asn Gin Ser Val Arg His Leu Lys Tyr Ser Leu Phe
170 175 180
Lys Lys Ser Leu Leu Gly Ser Val Ser Asp Asn Gly He Val Thr
185 190 195
Leu Trp Asp Val Asn Ser Gin Ser Pro Tyr His Asn Phe Asp Ser
200 205 210
Val His Lys Ala Pro Ala Ser Gly He Cys Phe Ser Pro Val Asn
215 220 225
Glu Leu Leu Phe Val Thr He Gly Leu Asp Lys Arg He He Leu
230 235 240
Tyr Asp Thr Ser Ser Lys Lys Leu Val Lys Thr Leu Val Ala Asp
245 250 255
Thr Pro Leu Thr Ala Val Asp Phe Met Pro Asp Gly Ala Thr Leu 260 265 270
Ala He Gly Ser Ser Arg Gly Lys He Tyr Gin Tyr Asp Leu Arg
275 280 285
Met Leu Lys Ser Pro Val Lys Thr He Ser Ala His Lys Thr Ser
290 295 300
Val Gin Cys He Ala Phe Gin Tyr Ser Thr Val Leu Thr Lys Ser
305 310 315
Ser Leu Asn Lys Gly Cys Ser Asn Lys Pro Thr Thr Val Asn Lys
320 325 330
Arg Ser Val Asn Val Asn Ala Ala Ser Gly Gly Val Gin Asn Ser
335 340 345
Gly He Val Arg Glu Ala Pro Ala Thr Ser He Ala Thr Val Leu
350 355 360
Pro Gin Pro Met Thr Ser Ala Met Gly Lys Gly Thr Val Ala Val
365 370 375
Gin Glu Lys Ala Gly Leu Pro Arg Ser He Asn Thr Asp Thr Leu
380 385 390
Ser Lys Glu Thr Asp Ser Gly Lys Asn Gin Asp Phe Ser Ser Phe
395 400 405
Asp Asp Thr Gly Lys Ser Ser Leu Gly Asp Met Phe Ser Pro He
410 415 420
Arg Asp Asp Ala Val Val Asn Lys Gly Ser Asp Glu Ser He Gly
425 430 435
Lys Gly Asp Gly Phe Asp Phe Leu Pro Gin Leu Asn Ser Val Phe
440 445 450
Pro Pro Arg Lys Asn Pro Val Thr Ser Ser Thr Ser Val Leu His
455 460 465
Ser Ser Pro Leu Asn Val Phe Met Gly Ser Pro Gly Lys Glu Glu
470 475 480
Asn Glu Asn Arg Asp Leu Thr Ala Glu Ser Lys Lys He Tyr Met
485 490 495
Gly Lys Gin Glu Ser Lys Asp Ser Phe Lys Gin Leu Ala Lys Leu
500 505 510
Val Thr Ser Gly Ala Glu Ser Gly Asn Leu Asn Thr Ser Pro Ser
515 520' 525
Ser Asn Gin Thr Arg Asn Ser Glu Lys Phe Glu Lys Pro Glu Asn
530 535 540
Glu He Glu Ala Gin Leu He Cys Glu Pro Pro He Asn Gly Ser
545 550 555
Ser Thr Pro Asn Pro Lys He Ala Ser Ser Val Thr Ala Gly Val
560 565 570
Ala Ser Ser Leu Ser Glu Lys He Ala Asp Ser He Gly Asn Asn
575 580 585
Arg Gin Asn Ala Pro Leu Thr Ser He Gin He Arg Phe He Gin
590 595 600
Asn Met He Gin Glu Thr Leu Asp Asp Phe Arg Glu Ala Cys His
605 610 615
Arg Asp He Val Asn Leu Gin Val Glu Met He Lys Gin Phe His
620 625 630
Met Gin Leu Asn Glu Met His Ser Leu Leu Glu Arg Tyr Ser Val
635 640 645
Asn Glu Gly Leu Val Ala Glu He Glu Arg Leu Arg Glu Glu Asn
650 655 660
Lys Arg Leu Arg Ala His Phe
665
<210> 7 <211> 897 <212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 7482435CD1
<400> 7
Met Ala Glu He Pro Leu Tyr Phe Val Asp Leu Gin Asp Asp Leu
1 5 10 15
Asp Asp Tyr Gly Phe Glu Asp Tyr Gly Thr Asp Cys Asp Asn Met
20 25 30
Arg Val Thr Ala Phe Leu Asp He Pro Gly Gin Asp Asn Leu Pro
35 40 45
Pro Leu Thr Arg Leu Glu Lys Tyr Ala Phe Ser Glu Asn Thr Phe
50 55 60
Asn Arg Gin He He Ala Arg Gly Leu Leu Asp He Phe Arg Asp
65 70 75
Phe Gly Asn Asn Glu Glu Asp Phe Leu Thr Val Met Glu He Val
80 85 90
Val Arg Leu Ser Glu Asp Ala Glu Pro Thr Val Arg Thr Glu Leu
95 100 105
Met Glu Gin He Pro Pro He Ala He Phe Leu Gin Glu Asn Arg
110 115 120
Ser Asn Phe Pro Val Val Leu Ser Glu Tyr Leu He Pro He Val
125 130 135
Met Arg Tyr Leu Thr Asp Pro Asn Asn Gin He He Cys Lys Met
140 145 150
Ala Ser Met Leu Ser Lys Ser Thr Val Glu Arg Leu Leu Leu Pro
155 160 165
Arg Phe Cys Glu Leu Cys Gly Asp Arg Lys Leu Phe Gin Val Arg
170 175 180
Lys Val Cys Ala Ala Asn Phe Gly Asp He Cys His Ala Val Gly
185 190 195
Gin Glu Ala Thr Glu Lys Phe Leu He Pro Lys Phe Phe Glu Leu
200 205 210
Cys Ser Asp Ala Val Trp Gly Met Arg Lys Ala Cys Ala Glu Cys
215 220 225
Phe Thr Ala Val Ser His Ser Ser Ser Pro Gly Val Arg Arg Thr
230 235 240
Gin Leu Phe Pro Leu Phe He Arg Leu Val Ser Asp Pro Cys Arg
245 250 255
Trp Val His Gin Ala Ala Phe Gin Ser Leu Gly Pro Phe He Ser
260 265 270
Thr Phe Ala Asn Pro Ser Arg Ala Gly Leu Tyr Leu Arg Glu Asp
275 280 285
Gly Ala Leu Ser He Trp Pro Leu Thr Gin Asp Leu Asp Ser Gly
290 295 300
Phe Ala Ser Gly Ser Pro Ala Pro Ser Ser Gly Gly Asn He Ser
305 310 315
Pro Ala Ser Leu He Arg Ser Ala Lys Pro Val Arg Ser Glu Pro
320 325 330
Glu Leu Pro Val Glu Gly Thr Ser Ala Lys Thr Ser Asp Cys Pro
335 340 345
His Ser Ser Ser Ser Ser Asp Gly Pro Ala Glu Ser Pro Val Glu
350 355 360
Ser Cys Val Ser Ala Gly Ala Glu Trp Thr Arg Val Ser Pro Glu
365 370 375 Thr Ser Ala Arg Ser Lys Leu Ser Asp Met Asn Asp Leu Pro He
380 385 390
Ser Ser Tyr Pro Gly Ser Asp Ser Trp Ala Cys Pro Gly Asn Thr
395 400 405
Glu Asp Val Phe Ser His Phe Leu Tyr Cys Lys Asp Leu Glu Leu
410 415 420
Leu Leu Ser Glu Ala Gly Pro Gin Glu Asp Asp Cys Ser Arg Pro
425 430 435
Gly Val Val His Asn Ser Cys Val Ala Arg Ser Glu He Gin Lys
440 445 450
Val Leu Asp Ser Leu Gin Glu His Leu Met Asn Asp Pro Asp Val
455 460 465
Gin Ala Gin Val Gin Val Leu Ser Ala Ala Leu Arg Ala Ala Gin
470 475 480
Leu Asp Cys Val Asn Glu Ala Glu Ser Lys Pro Thr Ala Gly Leu
485 490 495
Lys Glu Val Ser lie Ser His Pro Ser Ser Ala Ser Asp Asn Gin
500 505 510
He Ala Leu Ala Ala Ser Ser Ser Gin Asp Glu Leu Phe Val Ala
515 520 525
Arg He Leu Gin Ser Pro Asp Pro Gly Gly Pro Arg Asn Gly Thr
530 535 540
Ser Asp His Leu Glu Thr Asp Gin Arg Gin Asp Pro Thr Pro Leu
545 550 555
Glu Glu Asn Lys Ser Lys Leu Gin Asp Val He Pro Gin Pro Leu
560 565 570
Leu Asp Gin Tyr Val Ser Met Thr Asp Pro Ala Arg Ala Gin Thr
575 580 585
Val Asp Thr Asp He Ala Lys His Cys Ala Tyr Ser Leu Pro Gly
590 595 600
Val Ala Leu Thr Leu Gly Arg Gin Asn Trp His Cys Leu Lys Asp
605 610 615
Thr Tyr Glu Thr Leu Ala Ser Asp Val Gin Trp Lys Val Arg Arg
620 625 630
' Ala Leu Ala Phe Ser He His Glu Leu Ala Val He Leu Gly Asp
635 640 645
Gin Leu Thr Ala Ala Asp Leu Val Pro He Phe Asn Gly Phe Leu
650 655 660
Lys Asp Leu Asp Glu Val Arg He Gly Val Leu Arg His Leu Tyr
665 670 675
Asp Phe Leu Lys Leu Leu His Glu Asp Lys Arg Arg Asp Tyr Leu
680 685 690
Tyr Gin Leu Gin Glu Phe Val Val Thr Asp Asn Ser Arg Asn Trp
695 700 705
Arg Phe Arg Tyr Glu Leu Ala Glu Gin Leu He Leu He Leu Glu
710 715 720
Leu Tyr Ser Pro Asn Asp Val Tyr Asp Tyr Leu Met His He Ala
725 730 735
Leu Lys Leu Cys Ala Asp Gin Val Ser Glu Val Arg Trp He Ser
740 745 750
Phe Lys Leu Val Val Ala He Leu Gin Lys Phe Tyr Ser Asn Ser
755 760 765
Glu Ser Ala Leu Gly Leu Asn Phe He Asn Glu Leu He He Arg
770 775 780
Phe Arg His Cys Ser Lys Trp Val Gly Arg Gin Ala Phe Ala Phe
785 790 795
He Cys Gin Ala Val Val Ser Lys Glu Cys Val Pro Val Asp Gin
800 805 810 Phe Met Glu His Leu Leu Pro Ser Leu Leu Ser Leu Ala Ser Asp
815 820 825
Pro Val Pro Asn Val Arg Val Leu Leu Ala Lys Ala Leu Arg Gin
830 835 840
Met Leu Leu Glu Lys Ala Tyr Phe Arg Asn Ala Gly Asn Pro His
845 850 855
Leu Glu Val He Glu Glu Thr He Leu Ala Leu Gin Ser Asp Arg
860 865 870
Asp Gin Asp Val Ser Phe Phe Ala Ala Leu Glu Pro Lys Arg Arg
875 880 885
Asn He He Asp Thr Ala Val Leu Glu Lys Gin Asn
890 895
<210> 8
<211> 454
<212> PRT
<213> Homo sapiens
<220>
<221> misc_£eature
<223> Incyte ID No : 3882333CD1
<400> 8
Met Leu Lys Gin He Leu Ser Glu Met Tyr He Asp Pro Asp Leu
1 5 10 15
Leu Ala Glu Leu Ser Glu Glu Gin Lys Gin He Leu Phe Phe Lys
20 25 30
Met Arg Glu Glu Gin He Arg Arg Trp Lys Glu Arg Glu Ala Ala
35 40 45
Met Glu Arg Lys Glu Ser Leu Pro Val Lys Pro Arg Pro Lys Lys
50 55 60
Glu Asn Gly Lys Ser Val His Trp Lys Leu Gly Ala Asp Lys Glu
65 70 75
Val Trp Val Trp Val Met Gly Glu His His Leu Asp Lys Pro Tyr
80 85 90
Asp Val Leu Cys Asn Glu He He Ala Glu Arg Ala Arg Leu Lys
95 100 iθ5
Ala Glu Gin Glu Ala Glu Glu Pro Arg Lys Thr His Ser Glu Glu
110 115 120
Phe Thr Asn Ser Leu Lys Thr Lys Ser Gin Tyr His Asp Leu Gin
125 130 135
Ala Pro Asp Asn Gin Gin Thr Lys Asp He Trp Lys Lys Val Ala
140 145 150
Glu Lys Glu Glu Leu Glu Gin Gly Ser Arg Pro Ala Pro Thr Leu
155 160 165
Glu Glu Glu Lys He Arg Ser Leu Ser Ser Ser Ser Arg Asn He
170 175 180
Gin Gin Met Leu Ala Asp Ser He Asn Arg Met Lys Ala Tyr Ala
185 190 195
Phe His Gin Lys Lys Glu Ser Met Lys Lys Lys Gin Asp Gly Glu
200 205 210
He Asn Gin He Glu Gly Glu Arg Thr Lys Gin He Cys Lys Ser
215 220 225
Trp Lys Glu Asp Ser Glu Trp Gin Ala Ser Leu Arg Lys Ser Lys
230 235 240
Ala Ala Asp Glu Lys Arg Arg Ser Leu Ala Lys Gin Ala Arg Glu
245 250 255
Asp Tyr Lys Arg Leu Ser Leu Ala Ala Gin Lys Gly Arg Gly Gly 260 265 270
Glu Arg Leu Gin Ser Pro Leu Arg Val Pro Gin Lys Pro Glu Arg
275 280 285
Pro Pro Leu Pro Pro Lys Pro Gin Phe Leu Asn Ser Gly Ala Tyr
290 295 300
Pro Gin Lys Pro Leu Arg Asn Gin Gly Val Val Arg Thr Leu Ser
305 310 315
Ser Ser Ala Gin Glu Asp He He Arg Trp Phe Lys Glu Glu Gin
320 325 330
Leu Pro Leu Arg Ala Gly Tyr Gin Lys Thr Ser Asp Thr He Ala
335 340 345
Pro Trp Phe His Gly He Leu Thr Leu Lys Lys Ala Asn Glu Leu
350 355 360
Leu Leu Ser Thr Gly Met Pro Gly Ser Phe Leu He Arg Val Ser
365 370 375
Glu Arg He Lys Gly Tyr Ala Leu Ser Tyr Leu Ser Glu Asp Gly
380 385 390
Cys Lys His Phe Leu He Asp Ala Ser Ala Asp Ala Tyr Ser Phe
395 400 405
Leu Gly Val Asp Gin Leu Gin His Ala Thr Leu Ala Asp Leu Val
410 415 420
Glu Tyr His Lys Glu Glu Pro He Thr Ser Leu Gly Lys Glu Leu
425 430 435
Leu Leu Tyr Pro Cys Gly Gin Gin Asp Gin Leu Pro Asp Tyr Leu
440 , 445 450 Glu Leu Phe Glu
<210> 9
<211> 344
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> incyte ID No: 7482809CD1
<400> 9 i
Met Ala Thr Pro Tyr Val Pro Val Pro Met Pro He Gly Asn' Ser
1 5 10 15
Ala Ser Ser Phe Thr Thr Asn Arg Asn Gin Arg Ser Ser Ser Phe
20 25 30
Gly Ser Val Ser Thr Ser Ser Asn Ser Ser Lys Gly Gin Leu Glu
35 40 45
Asp Ser Asn Met Gly Asn Phe Lys Gin Thr Ser Val Pro Asp Gin
50 55 60
Met Asp Asn Thr Ser Ser Val Cys Ser Ser Pro Leu He Arg Thr
65 70 75
Lys Phe Thr Gly Thr Ala Ser Ser He Glu Tyr- Ser Thr Arg Pro
80 85 90
Arg Asp Thr Glu Glu Gin Asn Pro Glu Thr Val Asn Trp Glu Asp
95 100 105
Arg Pro Ser Thr Pro Thr He Leu Gly Tyr Glu Val Met Glu Glu
110 115 120
Arg Ala Lys Phe Thr Val Tyr Lys He Leu Val Lys Lys Thr Pro
125 130 135
Glu Glu Ser Trp Val Val Phe Arg Arg Tyr Thr Asp Phe Ser Arg
140 145 150 Leu Asn Asp Lys Leu Lys Glu Met Phe Pro Gly Phe Arg Leu Ala
155 160 165
Leu Pro Pro Lys Arg Trp Phe Lys Asp Asn Tyr Asn Ala Asp Phe
170 175 180
Leu Glu Asp Arg Gin Leu Gly Leu Gin Ala Phe Leu Gin Asn Leu
185 190 195
Val Ala His Lys Asp He Ala Asn Cys Leu Ala Val Arg Glu Phe
200 205 210
Leu Cys Leu Asp Asp Pro Pro Gly Pro Phe Asp Ser Leu Glu Glu
215 220 225
Ser Arg Ala Phe Cys Glu Thr Leu Glu Glu Thr Asn Tyr Arg Leu
230 235 240
Gin Lys Glu Leu Leu Glu Lys Gin Lys Glu Met Glu Ser Leu Lys
245 250 255
Lys Leu Leu Ser Glu Lys Gin Leu His He Asp Thr Leu Glu Asn
260 265 270
Arg He Arg Thr Leu Ser Leu Glu Pro Glu Glu Ser Leu Asp Val
275 280 285
Ser Glu Thr Glu Gly Glu Gin He Leu Lys Val Glu Ser Ser Ala
290 295 300
Leu Glu Val, Asp Gin Asp Val Leu Asp Glu Glu Ser Arg Ala Asp
305 310 315
Asn Lys Pro Cys Leu Ser Phe Ser Glu Pro Glu Asn Ala Val Ser
320 325 330
Glu He Glu Val Ala Glu Val Ala Tyr Asp Ala Glu Glu Asp
335 340
<210> 10
<211> 1115
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1739178CD1 *
<400> 10
Met Ala Lys He Val Val Val Thr Cys Ser Asp Ser Ser Phe Gly
1 5 10 15
Asn Phe Trp Leu Asp Gin Trp Gin Lys Arg Ala Arg Glu Lys Ser
20 25 30
Leu Cys Gin Cys Ser Ala Lys Gin Glu He Arg Thr Gin Leu Val
35 40 45
Glu Gin Phe Lys Cys Leu Glu Gin Gin Ser Glu Ser Arg Leu Gin
50 55 60
Leu Leu Gin Asp Leu Gin Glu Phe Phe Arg Arg Lys Ala Glu He
65 70 75
Glu Leu Glu Tyr Ser Arg Ser Leu Glu Lys Leu Ala Glu Arg Phe
80 85 90
Ser Ser Lys He Arg Ser Ser Arg Glu His Gin Phe Lys Lys Asp
95 100 105
Gin Tyr Leu Leu Ser Pro Val Asn Cys Trp Tyr Leu Val Leu His
110 115 120
Gin Thr Arg Arg Glu Ser Arg Asp His Ala Thr Leu Asn Asp He
125 130 135
Phe Met Asn Asn Val He Val Arg Leu Ser Gin He Ser Glu Asp
140 145 150
Val He Arg Leu Phe Lys Lys Ser Lys Glu He Gly Leu Gin Met 155 160 165
His Glu Glu Leu Leu Lys Val Thr Asn Glu Leu Tyr Thr Val Met
170 175 180
Lys Thr Tyr His Met Tyr His Ala Glu Ser He Ser Ala Glu Ser
185 190 195
Lys Leu Lys Glu Ala Glu Lys Gin Glu Glu Lys Gin Phe Asn Lys
200 205 210
Ser Gly Asp Leu Ser Met Asn Leu Leu Arg His Glu Asp Arg Pro
215 220 225
Gin Arg Arg Ser Ser Val Lys Lys He Glu Lys Met Lys Glu Lys
230 235 240
Arg Gin Ala Lys Tyr Ser Glu Asn Lys Leu Lys Cys Thr Lys Ala
245 250 255
Arg Asn Asp Tyr Leu Leu Asn Leu Ala Ala Thr Asn Ala Ala He
260 265 • 270
Ser Lys Tyr Tyr He His Asp Val Ser Asp Leu He Asp Cys Cys
275 280 285
Asp Leu Gly Phe His Ala Ser Leu Ala Arg Thr Phe Arg Thr Tyr
290 295 300
Leu Ser Ala Glu Tyr Asn Leu Glu Thr Ser Arg His Glu Gly Leu
305 310 315
Asp Val He Glu Asn Ala Val Asp Asn Leu Asp Ser Arg Ser Asp
320 325 330
Lys His Thr Val Met Asp Met Cys Asn Gin Val Phe Cys Pro Pro
335 340 345
Leu Lys Phe Glu Phe Gin Pro His Met Gly Asp Glu Val Cys Gin
350 355 360
Val Ser Ala Gin Gin Pro Val Gin Thr Glu Leu Leu Met Arg Tyr
365 370 375
His Gin Leu Gin Ser Arg Leu Ala Thr Leu Lys He Glu Asn Glu
380 385 390
Glu Val Arg Lys Thr Leu Asp Ala Thr Met Gin Thr Leu Gin Asp
395 400 405
Met Leu Thr Val Glu Asp Phe Asp Val Ser Asp Ala Phe Gin His
410 415 420
Ser Arg Ser Thr Glu Ser Val Lys Ser Ala Ala Ser Glu Thr Tyr
425 430 435
Met Ser Lys He Asn He Ala Lys Arg Arg Ala Asn Gin Gin Glu
440 445 450
Thr Glu Met Phe Tyr Phe Thr Lys Phe Lys Glu Tyr Val Asn Gly
455 460 465
Ser Asn Leu He Thr Lys Leu Gin Ala Lys His Asp Leu Leu Lys
470 475 480
Gin Thr Leu Gly Glu Gly Glu Arg Ala Glu Cys Gly Thr Thr Arg
485 490 495
Pro Pro Cys Leu Pro Pro Lys Pro Gin Lys Met Arg Arg Pro Arg
500 505 510
Pro Leu Ser Val Tyr Ser His Lys Leu Phe Asn Gly Ser Met Glu
515 520 525
Ala Phe He Lys Asp Ser Gly Gin Ala He Pro Leu Val Val Glu
530 535 540
Ser Cys He Arg Tyr He Asn Leu Tyr Gly Leu Gin Gin Gin Gly
545 550 555
He Phe Arg Val Pro Gly Ser Gin Val Glu Val Asn Asp He Lys
560 565 570
Asn Ser Phe Glu Arg Gly Glu Asp Pro Leu Val Asp Asp Gin Asn
575 580 585
Glu Arg Asp He Asn Ser Val Ala Gly Val Leu Lys Leu Tyr Phe 590 595 600
Arg Gly Leu Glu Asn Pro Leu Phe Pro Lys Glu Arg Phe Gin Asp
605 610 615
Leu He Ser Thr He Lys Leu Glu Asn Pro Ala Glu Arg Val His
620 625 630
Gin He Gin Gin He Leu Val Thr Leu Pro Arg Val Val He Val
635 640 645
Val Met Arg Tyr Leu Phe Ala Phe Leu Asn His Leu Ser Gin Tyr
650 655 660
Ser Asp Glu Asn Met Met Asp Pro Tyr Asn Leu Ala He Cys Phe
665 670 675
Gly Pro Thr Leu Met His He Pro Asp Gly Gin Asp Pro Val Ser
680 685 690
Cys Gin Ala His He Asn Glu Val He Lys Thr He He He His
695 700 705
His Glu Ala He Phe Pro Ser Pro Arg Glu Leu Glu Gly Pro Val
710 715 720
Tyr Glu Lys Cys Met Ala Gly Gly Glu Glu Tyr Cys Asp Ser Pro
725 730 735
His Ser Glu Pro Gly Ala He Asp Glu Val Asp His Asp Asn Gly
740 745 750
Thr Glu Pro His Thr Ser Asp Glu Glu Val Glu Gin He Glu Ala
755 760 765
He Ala Lys Phe Asp Tyr Met Gly Arg Ser Pro Arg Glu Leu Ser
770 775 780
Phe Lys Lys Gly Ala Ser Leu Leu Leu Tyr His Arg Ala Ser Glu
785 790 795
Asp Trp Trp Glu Gly Arg His Asn Gly Val Asp Gly Leu He Pro
800 805 810
His Gin Tyr He Val Val Gin Asp Met Asp Asp Ala Phe Ser Asp
815 820 825
Ser Leu Ser Gin Lys Ala Asp Ser Glu Ala Ser Ser Gly Pro Leu
830 835 840
Leu Asp Asp Lys Ala Ser Ser Lys Asn Asp Leu Gin Ser Pro Thr
845 850 855
Glu His He Ser Asp Tyr Gly Phe Gly Gly Val Met Gly Arg Val
860 865 870
Arg Leu Arg Ser Asp Gly Ala Ala He Pro Arg Arg Arg Ser Gly
875 880 885
Gly Asp Thr His Ser Pro Pro Arg Gly Leu Gly Pro Ser He Asp
890 895 900
Thr Pro Pro Arg Ala Ala Ala Cys Pro Ser Ser Pro His Lys He
905 910 915
Pro Leu Thr Arg Gly Arg He Glu Ser Pro Glu Lys Arg Arg Met
920 925 930
Ala Thr Phe Gly Ser Ala Gly Ser He Asn Tyr Pro Asp Lys Lys
935 940 945
Ala Leu Ser Glu Gly His Ser Met Arg Ser Thr Cys Gly Ser Thr
950 955 960
Arg His Ser Ser Leu Gly Asp His Lys Ser Leu Glu Ala Glu Ala
965 970 975
Leu Ala Glu Asp He Glu Lys Thr Met Ser Thr Ala Leu His Glu
980 985 990
Leu Arg Glu Leu Glu Arg Gin Asn Thr Val Lys Gin Ala Pro Asp
995 1000 1005
Val Val Leu Asp Thr Leu Glu Pro Leu Lys Asn Pro Pro Gly Pro
1010 1015 1020
Val Ser Ser Glu Pro Ala Ser Pro Leu His Thr He Val He Arg 1025 1030 1035
Asp Pro Asp Ala Ala Met Arg Arg Ser Ser Ser Ser Ser Thr Glu
1040 1045 1050
Met Met Thr Thr Phe Lys Pro Ala Leu Ser Ala Arg Leu Ala Gly
1055 1060 1065
Ala Gin Leu Arg Pro Pro Pro Met Arg Pro Val Arg Pro Val Val
1070 1075 1080
Gin His Arg Ser Ser Ser Ser Ser Ser Ser Gly Val Gly Ser Pro
1085 1090 1095
Ala Val Thr Pro Thr Glu Lys Met Phe Pro Asn Ser Ser Ala Asp
1100 1105 1110 Lys Ser Gly Thr Met
1115
<210> 11
<211> 839
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 7473630CD1 •
<400> 11
Met Ala Pro Asn Leu Lys Lys Gly Thr Ser Ser Cys Pro Gly Leu
1 5 10 15
Thr Asn Gin Glu Thr His Ser Asp Ser Lys Gly Glu Gly Ala Gly
20 25 30
Pro Asp Gly Lys He Tyr Asp Gly Lys Asp Lys Thr Thr His Leu
35 40 45
Leu Gly Ala Phe Thr Gly Ala Ser Met Arg Gly Leu Thr Leu Ser
50 55 60
Ser Thr Ser Asn Gin Leu Trp Leu Glu Phe Asn Ser Asp Thr Glu
65 70 75
Gly Thr Asp Glu Gly Phe Gin Leu Val Tyr Thr Lys Arg He He
80 85 90
Gly He Ala Glu Glu Val Thr Val Leu Thr Leu Thr Glu Ser Glu
95 100 105
Gin Glu Arg Glu His Leu Ser Arg Glu Asp Gin Val Leu Asn Ser
110 115 120
His Thr Val Lys He Leu Ala Phe His Asn Leu Asp Thr Arg Ser
125 130 135
Val Thr Lys Ala Thr Leu Leu Val Ala Pro Ser Phe Met Asp Ala
140 145 150 lie Gin Ala Thr Leu Ser Thr Glu Val Ala Phe Ser Thr Glu Cys
155 160 165
Gly Gly Arg Phe Lys Gly Glu Ser Ser Gly Arg He Leu Ser Pro
170 175 180
Gly Tyr Pro Phe Pro Tyr Asp Asn Asn Leu Arg Cys Met Trp Met
185 190 195
He Glu Val Asp Pro Gly Asn He Val Ser Leu Gin Phe Leu Ala
200 205 210
Phe Asp Thr Glu Ala Ser His Asp He Leu Arg Val Trp Asp Gly
215 220 225
Pro Pro Glu Asn Asp Met Leu Leu Lys Glu He Ser Gly Ser Leu
230 235 240
He Pro Glu Gly He His Ser Thr Leu Asn He Val Thr He Gin
245 250 255 Phe Asp Thr Asp Phe Tyr He Ser Lys Ser Gly Phe Ala He Gin
260 265 270
Phe Ser Ser Ser Val Ala Thr Ala Cys Arg Asp Pro Gly Val Pro
275 280 285
Met Asn Gly Thr Arg Asn Gly Asp Gly Arg Glu Pro Gly Asp Thr
290 295 300
Val Val Phe Gin Cys Asp Pro Gly Tyr Glu Leu Gin Gly Glu Glu
305 310 315
Arg He Thr Cys He Gin Val Glu Asn Arg Tyr Phe Trp Gin Pro
320 325 330
Ser Pro Pro Val Cys He Ala Pro Cys Gly Gly Asn Leu Thr Gly
335 340 345
Ser Ser Gly Phe He Leu Ser Pro Asn Phe Pro His Pro Tyr Pro
350 355 360
His Ser Arg Asp Cys Asp Trp Thr He Thr Val Asn Ala Asp Tyr
365 370 375
Val He Ser Leu Ala Phe He Ser Phe Ser He Glu Pro Asn Tyr
380 385 390
Asp Phe Leu Tyr He Tyr Asp Gly Pro Asp Ser Asn Ser Pro Leu
395 400 405
He Gly Ser Phe Gin Asp Ser Lys Leu Pro Glu Arg He Glu Ser
410 415 420
Ser Ser Asn Thr Met His Leu Ala Phe Arg Ser Asp Gly Ser Val
425 430 435
Ser Tyr Thr Gly Phe His Leu Glu Tyr Lys Ala Lys Leu Arg Glu
440 445 450
Ser Cys Phe Asp Pro Gly Asn He Met Asn Gly Thr Arg Leu Gly
455 460 465
Met Asp Tyr Lys Leu Gly Ser Thr Val Thr Tyr Tyr Cys Asp Ala
470 475 480
Gly Tyr Val Leu Gin Gly Tyr Ser Thr Leu Thr Cys Phe Met Gly
485 490 495
Asp Asp Gly Arg Pro Gly Trp Asn Arg Ala Leu Pro Ser Cys His
500 505 510
Ala Pro Cys Gly Ser Arg Ser Thr Gly Ser Glu Gly Thr Val Leu
515 520 525
Ser Pro Asn Tyr Pro Lys Asn Tyr Ser Val Gly His Asn Cys Val
530 535 540
Tyr Ser He Ala Val Pro Lys Glu Leu Trp Cys Trp Pro Val Val
545 550 555
Phe Phe Gin Thr Ser Leu His Asp Val Val Glu Val Tyr Asp Gly
560 565 570
Pro Thr Gin Gin Ser Ser Leu Leu Ser Ser Leu Ser Gly Ser His
575 580 585
Ser Gly Glu Ser Leu Pro Leu Ser Ser Gly Asn Gin He Thr He
590 595 600
Arg Phe Thr Ser Val Gly Pro He Thr Ala Lys Gly Phe His Phe
605 610 615
Val Tyr Gin Ala Val Pro Arg Thr Ser Ser Thr Gin Cys Ser Ser
620 625 630
Val Pro Glu Pro Arg Phe Gly Arg Arg He Gly Asn Glu Phe Ala
635 640 645
Val Gly Ser Ser Val Leu Phe Asp Cys Asn Pro Gly Tyr He Leu
650 655 660
His Gly Ser He Ala He Arg Cys Glu Thr Val Pro Asn Ser Leu
665 670 675
Ala Gin Trp Asn Asp Ser Leu Pro Thr Cys He Val Pro Cys Gly
680 685 690 Gly He Leu Thr Lys Arg Lys Gly Thr He Leu Ser Pro Gly Tyr
695 700 705
Pro Glu Pro Tyr Asp Asn Asn Leu Asn Cys Val Trp Lys He Thr
710 715 720
Val Pro Glu Gly Ala Gly He Gin Val Gin Val Val Ser Phe Ala
725 730 735
Thr Glu His Asn Trp Asp Ser Leu Asp Phe Tyr Asp Gly Gly Asp
740 745 750
Asn Asn Ala Pro Arg Leu Gly Ser Tyr Ser Gly Thr Thr He Pro
755 760 765
His Leu Leu Asn Ser Thr Ser Asn Asn Leu Tyr Leu Asn Phe Gin
770 775 780
Ser Asp He Ser Val Ser Ala Ala Gly Phe His Leu Glu Tyr Thr
785 790 795
Ala He Gly Leu Asp Ser Cys Pro Glu Pro Gin Thr Pro Ser Ser
800 805 810
Gly He Lys He Gly Asp Arg Tyr Met Val Gly Asp Val Val Ser
815 820 825
Phe Gin Cys Asp Gin Gly Tyr Ser Leu Gin Val Ser Leu Phe
830 835
<210> 12
<211> 304
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1431520CD1
<400> 12
Met Ser Ser He Lys His Leu Val Tyr Ala Val He Arg Phe Leu
1 5 10 15
Arg Glu Gin Ser Gin Met Asp Thr Tyr Thr Ser Asp Glu Gin Glu
20 25 30
Ser Leu Glu Val Ala He Gin Cys Leu Glu Thr Val Phe Lys He
35 40 45
Ser Pro Glu Asp Thr His Leu Ala Val Ser Gin Pro Leu Thr Glu
50 55 60
Met Phe Thr Ser Ser Phe Cys Lys Asn Asp Val Leu Pro Leu Ser
65 70 75
Asn Ser Val Pro Glu Asp Val Gly Lys Ala Asp Gin Leu Lys Asp
80 85 90
Glu Gly Asn Asn His Met Lys Glu Glu Asn Tyr Ala Ala Ala Val
95 100 105
Asp Cys Tyr Thr Gin Ala He Glu Leu Asp Pro Asn Asn Ala Val
110 115 120
Tyr Tyr Cys Asn Arg Ala Ala Ala Gin Ser Lys Leu Gly His Tyr
125 130 135
Thr Asp Ala He Lys Asp Cys Glu Lys Ala He Ala He Asp Ser
140 145 150
Lys Tyr Ser Lys Ala Tyr Gly Arg Met Gly Leu Ala Leu Thr Ala
155 160 165
Leu Asn Lys Phe Glu Glu Ala Val Thr Ser Tyr Gin Lys Ala Leu
170 175 180
Asp .Leu Asp Pro Glu Asn Asp Ser Tyr Lys Ser Asn Leu Lys He
185 190 195
Ala Glu Gin Lys Leu Arg Glu Val Ser Ser Pro Thr Gly Thr Gly 200 205 210
Leu Ser Phe Asp Met Ala Ser Leu He Asn Asn Pro Ala Phe He
215 220 225
Ser Met Ala Ala Ser Leu Met Gin Asn Pro Gin Val Gin Gin Leu
230 235 240 Met Ser Gly Met Met Thr Asn Ala He Gly Gly Pro Ala Ala Gly
245 250 255
Val Gly Gly Leu Thr Asp Leu Ser Ser Leu He Gin Ala Gly Gin
260 265 270
Gin Phe Ala Gin Gin He Gin Gin Gin Asn Pro Glu Leu He Glu
275 280 285
Gin Leu Arg Asn His He Arg Ser Arg Ser Phe Ser Ser Ser Ala
290 295 300 Glu Glu His Ser
<210> 13
<211> 440
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1916304CD1
<400> 13
Met Pro He Pro Pro Pro Pro Pro Pro Pro Pro Gly Pro Pro Pro
1 5 10 15
Pro Pro Thr Phe His Gin Ala Asn Thr Glu Gin Pro Lys Leu Ser
20 25 30
Arg Asp Glu Gin Arg Gly Arg Gly Ala Leu Leu Gin Asp He Cys
35 40 45
Lys Gly Thr Lys Leu Lys Lys Val Thr Asn He Asn Asp Arg Ser
50 55 60
Ala Pro He Leu Glu Lys Pro Lys Gly Ser Ser Gly Gly Tyr Gly
65 70 75
Ser Gly Gly Ala Ala Leu Gin Pro Lys Gly Gly Leu Phe Gin Gly
80 85 90
Gly Val Leu Lys Leu Arg Pro Val Gly Ala Lys Asp Gly Ser Glu
95 100 105
Asn Leu Ala Gly Lys Pro Ala Leu Gin He Pro Ser Ser Arg Ala
110 115 120
Ala Ala Pro Arg Pro Pro Val Ser Ala Ala Ser Gly Arg Pro Gin
125 ' 130 135
Asp Asp Thr Asp Ser Ser Arg Ala Ser Leu Pro Glu Leu Pro Arg
140 145 150
Met Gin Arg Pro Ser Leu Pro Asp Leu Ser Arg Pro Asn Thr Thr
155 160 165
Ser Ser Thr Gly Met Lys His Ser Ser Ser Ala Pro Pro Pro Pro
170 175 180
Pro Pro Gly Arg Arg Ala Asn Ala Pro Pro Thr Pro Leu Pro Met
185 190 195
His Ser Ser Lys Ala Pro Ala Tyr Asn Arg Glu Lys Pro Leu Pro
200 205 210
Pro Thr Pro Gly Gin Arg Leu His Pro Gly Arg Glu Gly Pro Pro
215 220 225
Ala Pro Pro Pro Val Lys Pro Pro Pro Ser Pro Val Asn He Arg
230 235 240 Thr Gly Pro Ser Gly Gin Ser Leu Ala Pro Pro Pro Pro Pro Tyr
245 250 255
Arg Gin Pro Pro Gly Val Pro Asn Gly Pro Ser Ser Pro Thr Asn
260 265 270
Glu Ser Ala Pro Glu Leu Pro Gin Arg His Asn Ser Leu His Arg
275 280 285
Lys Thr Pro Gly Pro Val Arg Gly Leu Ala Pro Pro Pro Pro Thr
290 295 300
Ser Ala Ser Pro Ser Leu Leu Ser Asn Arg Pro Pro Pro Pro Ala
305 310 315
Arg Asp Pro Pro Ser Arg Gly Ala Ala Pro Pro Pro Pro Pro Pro
320 325 330
Val He Arg Asn Gly Ala Arg Asp Ala Pro Pro Pro Pro Pro Pro
335 340 345
Tyr Arg Met His Gly Ser Glu Pro Pro Ser Arg Gly Lys Pro Pro
350 355 360
Pro Pro Pro Ser Arg Thr Pro Ala Gly Pro Pro Pro Pro Pro Pro
365 370 375
Pro Pro Leu Arg Asn Gly His Arg Asp Ser He Thr Thr Val Arg
380 385 390
Ser Phe Leu Asp Asp Phe Glu Ser Lys Tyr Ser Phe His Pro Val
395 400 405
Glu Asp Phe Pro Ala Pro Glu Glu Tyr Lys His Phe Gin Arg He
410 415 420
Tyr Pro Ser Lys Thr Asn Arg Ala Ala Arg Gly Ala Pro Pro Leu
425 430 435
Pro Pro He Leu Arg
440
<210> 14
<211> 747
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 378504CD1
<400> 14
Met Gin Gly Gly Glu Pro Val Ser Thr Met Lys Val Ser Glu Ser
1 5 10 15
Glu Gly Lys Leu Glu Gly Gin Ala Thr Ala Val Thr Pro Asn Lys
20 25 30
Asn Ser Ser Cys Gly Gly Gly He Ser Ser Ser Ser Ser Ser Arg
35 40 45
Gly Gly Ser Ala Lys Gly Trp Gin Tyr Ser Asp His Met Glu Asn
50 55 60
Val Tyr Gly Tyr Leu Met Lys Tyr Thr Asn Leu Val Thr Gly Trp
65 70 75
Gin Tyr Arg Phe Phe Val Leu Asn Asn Glu Ala Gly Leu Leu Glu
80 85 90
Tyr Phe Val Asn Glu Gin Ser Arg Asn Gin Lys Pro Arg Gly Thr
95 100 105
Leu Gin Leu Ala Gly Ala Val He Ser Pro Ser Asp Glu Asp Ser
110 115 120
His Thr Phe Thr Val Asn Ala Ala Ser Gly Glu Gin Tyr Lys Leu
125 130 135
Arg Ala Thr Asp Ala Lys Glu Arg Gin His Trp Val Ser Arg Leu 140 145 150
Gin He Cys Thr Gin His His Thr Glu Ala He Gly Lys Asn Asn
155 160 165
Pro Pro Leu Lys Ser Arg Ser Phe Ser Leu Ala Ser Ser Ser Asn
170 175 180
Ser Pro He Ser Gin Arg Arg Pro Ser Gin Asn Ala He Ser Phe
185 190 195
Phe Asn Val Gly His Ser Lys Leu Gin Ser Leu Ser Lys Arg Thr
200 205 210
Asn Leu Pro Pro Asp His Leu Val Glu Val Arg Glu Met Met Ser
215 220 225
His Ala Glu Gly GlnOln Arg Asp Leu He Arg Arg He Glu Cys
230 235 240
Leu Pro Thr Ser Gly His Leu Ser Ser Leu Asp Gin Asp Leu Leu
245 250 255
Met Leu Lys Ala Thr Ser Met Ala Thr Met Asn Cys Leu Asn Asp
250 265 270
Cys Phe His He Leu Gin Leu Gin His Ala Ser His Gin Lys Gly
275 280 285
Ser Leu Pro Ser Gly Thr Thr He Glu Trp Leu Glu Pro Lys He
290 295 300
Ser Leu Ser Asn His Tyr Lys Asn Gly Ala Asp Gin Pro Phe Ala
305 310 315
Thr Asp Gin Ser Lys Pro Val Ala Val Pro Glu Glu Gin Pro Val
320 325 330
Ala Glu Ser Gly Leu Leu Ala Arg Glu Pro Glu Glu He Asn Ala
335 340 345
Asp Asp Glu He Glu Asp Thr Cys Asp His Lys Glu Asp Asp Leu
350 355 360
Gly Ala Val Glu Glu Gin Arg Ser Val He Leu His Leu Leu Ser
365 370 375
Gin Leu Lys Leu Gly Met Asp Leu Thr Arg Val Val Leu Pro Thr
380 385 390
Phe He Leu Glu Lys Arg Ser Leu Leu Glu Met Tyr Ala Asp Phe
395 400 405
Met Ser His Pro Asp Leu Phe He Ala He Thr Asn Gly Ala Thr
410 415 420
Ala Glu Asp Arg Met He Arg Phe Phe Glu Tyr Tyr Leu Thr Ser
425 430 435
Phe His Glu Gly Arg Lys Gly Ala He Ala Lys Lys Pro Tyr Asn
440 445 450
Pro He He Gly Glu Thr Phe His Cys Ser Trp Lys Met Pro Lys
455 460 465
Ser Glu Val Ala Ser Ser Val Phe Ser Ser Ser Ser Thr Gin Gly
470 475 480
Val Thr Asn His Ala Pro Leu Ser Gly Glu Ser Leu Thr Gin Val
485 490 495
Gly Ser Asp Cys Tyr Thr Val Arg Phe Val Ala Glu Gin Val Ser
500 505 510
His His Pro Pro Val Ser Gly Phe Tyr Ala Glu Cys Thr Glu Arg
515 520 525
Lys Met Cys Val Asn Ala His Val Trp Thr Lys Ser Lys Phe Leu
530 535 540
Gly Met Ser He Gly Val Thr Met Val Gly Glu Gly He Leu Ser
545 550 555
Leu Leu Glu His Gly Glu Glu Tyr Thr Phe Ser Leu Pro Cys Ala
560 565 570
Tyr Ala Arg Ser He Leu Thr Val Pro Trp Val Glu Leu Gly Gly 575 580 585
Lys Val Ser Val Asn Cys Ala Lys Thr Gly Tyr Ser Ala Ser He
590 595 600
Thr Phe His Thr Lys Pro Phe Tyr Gly Gly Lys Leu His Arg Val
605 610 615
Thr Ala Glu Val Lys His Asn He Thr Asn Thr Val Val Cys Arg
620 625 630
Val Gin Gly Glu Trp Asn Ser Val Leu Glu Phe Thr Tyr Ser Asn
635 640 645
Gly Glu Thr Lys Tyr Val Asp Leu Thr Lys Leu Ala Val Thr Lys
650 655 660
Lys Arg Val Arg Pro Leu Glu Lys Gin Asp Pro Phe Glu Ser Arg
665 670 675
Arg Leu Trp Lys Asn Val Thr Asp Ser Leu Arg Glu Ser Glu He
680 685 590
Asp Lys Ala Thr Glu His Lys His Thr Leu Glu Glu Arg Gin Arg
695 700 705
Thr Glu Glu Arg His Arg Thr Glu Thr Gly Thr Pro Trp Lys Thr
710 715 720
Lys Tyr Phe He Lys Glu Gly Asp Gly Trp Val Tyr His Lys Pro ' 725 730 735
Leu Trp Lys He He Pro Thr Thr Gin Pro Ala Glu
740 745
<210> 15
<211> 770
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 5275371CD1
<400> 15
Met Pro Leu Leu Glu Lys Asn Glu Pro Lys Met Ser Glu Ala Lys
1 5 10 15
Asn Tyr Leu Ser Ser He Leu Asn His Gly Arg Leu Ser Pro Gin
20 25 30
Tyr Met Cys Glu Ala Met Leu He Leu Gly Lys Leu His Tyr Val
35 40 45
Glu Gly Ser Tyr Arg Asp Ala He Ser Met Tyr Ala Arg Ala Gly
50 55 60
He Asp Asp Met Ser Met Glu Asn Lys Pro Leu Tyr Gin Met Arg
65 70 75
Leu Leu Ser Glu Ala Phe Val He Lys Gly Leu Ser Leu Glu Arg
80 85 90
Leu Pro Asn Ser He Ala Ser Arg Phe Arg Leu Thr Glu Arg Glu
95 100 105
Glu Glu Val He Thr Cys Phe Glu Arg Ala Ser Trp He Ala Gin
110 115 120
Val Phe Leu Gin Glu Leu Glu Lys Thr Thr Asn Asn Ser Thr Ser
125 130 135
Arg His Leu Lys Gly Cys His Pro Leu Asp Tyr Glu Leu Thr Tyr
140 145 ■ 150
Phe Leu Glu Ala Ala Leu Gin Ser Ala Tyr Val Lys Asn Leu Lys
155 160 165
Lys Gly Asn He Val Lys Gly Met Arg Glu Leu Arg Glu Val Leu
170 175 180 Arg Thr Val Glu Thr Lys Ala Thr Gin Asn Phe Lys Val Met Ala
185 190 195
Ala Lys His Leu Ala Gly Val Leu Leu His Ser Leu Ser Glu Glu
200 205 210
Cys Tyr Trp Ser Pro Leu Ser His Pro Leu Pro Glu Phe Met Gly
215 220 225
Lys Glu Glu Ser Ser Phe Ala Thr Gin Ala Leu Arg Lys Pro His
230 ' 235 240
Leu Tyr Glu Gly Asp Asn Leu Tyr Cys Pro Lys Asp Asn He Glu
245 250 255
Glu Ala Leu Leu Leu Leu Leu He Ser Glu Ser Met Ala Thr Arg
260 265 270
Asp Val Val Leu Ser Arg Val Pro Glu Gin Glu Glu Asp Arg Thr
275 280 285
Val Ser Leu Gin Asn Ala Ala Ala He Tyr Asp Leu Leu Ser He
290 295 300
Thr Leu Gly Arg Arg Gly Gin Tyr Val Met Leu Ser Glu Cys Leu
305 310 315
Glu Arg Ala Met Lys Phe Ala Phe Gly Glu Phe His Leu Trp Tyr
320 325 330
Gin Val Ala Leu Ser Met Val Ala Cys Gly Lys Ser Ala Tyr Ala
335 340 345
Val Ser Leu Leu Arg Glu Cys Val Lys Leu Arg Pro Ser Asp Pro
350 355 360
Thr Val Pro Leu Met Ala Ala Lys Val Cys He Gly Ser Leu Arg
365 370 375
Trp Leu Glu Glu Ala Glu His Phe Ala Met Met Val He Ser Leu
380 • 385 390
Gly Glu Glu Ala Gly Glu Phe Leu Pro Lys Gly Tyr Leu Ala Leu
395 400 405
Gly Leu Thr Tyr Ser Leu Gin Ala Thr Asp Ala Thr Leu Lys Ser
410 415 420
Lys Gin Asp Glu Leu His Arg Lys Ala Leu Gin Thr Leu Glu Arg
425 430 435
Ala Gin Gin Leu Ala Pro Ser Asp Pro Gin Val He Leu Tyr Val
440 445 450
Ser Leu Gin Leu Ala Leu Val Arg Gin He Ser Ser Ala Met Glu
455 460 465
Gin Leu Gin Glu Ala Leu Lys Val Arg Lys Asp Asp Ala His Ala
470 475 480
Leu His Leu Leu Ala Leu Leu Phe Ser Ala Gin Lys His His Gin
485 490 495
His Ala Leu Asp Val Val Asn Met Ala He Thr Glu His Pro Glu
500 505 510
Asn Phe Asn Leu Met Phe Thr Lys Val Lys Leu Glu Gin Val Leu
515 520 525
Lys Gly Pro Glu Glu Ala Leu Val Thr Cys Arg Gin Val Leu Arg
530 535 540
Leu Trp Gin Thr Leu Tyr Ser Phe Ser Gin Leu Gly Gly Leu Glu
545 550 555
Lys Asp Gly Ser Phe Gly Glu Gly Leu Thr Met Lys Lys Gin Ser
560 565 570
Gly Met His Leu Thr Leu Pro Asp Ala His Asp Ala Asp Ser Gly
575 580 585
Ser Arg Arg Ala Ser Ser He Ala Ala Ser Arg Leu Glu Glu Ala
590 595 600
Met Ser Glu Leu Thr Met Pro Ser Ser Val Leu Lys Gin Gly Pro
605 610 615 Met Gin Leu Trp Thr Thr Leu Glu Gin He Trp Leu Gin Ala Ala
620 625 630
Glu Leu Phe Met Glu Gin Gin His Leu Lys Glu Ala Gly Phe Cys
635 640 645
He Gin Glu Ala Ala Gly Leu Phe Pro Thr Ser His Ser Val Leu
650 655 660
Tyr Met Arg Gly Arg Leu Ala Glu Val Lys Gly Asn Leu Glu Glu
665 670 675
Ala Lys Gin Leu Tyr Lys Glu Ala Leu Thr Val Asn Pro Asp Gly
680 685 690
Val Arg He Met His Ser Leu Gly Leu Met Leu Ser Arg Leu Gly
695 700 705
His Lys Ser Leu Ala Gin Lys Val Leu Arg Asp Ala Val Glu Arg
710 715 720
Gin Ser Thr Cys His Glu Ala Trp Gin Gly Leu Gly Glu Val Leu
725 730 735
Gin Ala Gin Gly Gin Asn Glu Ala Ala Val Asp Cys Phe Leu Thr
740 745 750 Ala Leu Glu Leu Glu Ala Ser Ser Pro Val Leu Pro Phe Ser He
755 760 765 lie Pro Arg Glu Leu
770
<210> 16
<211> 199
<212> PRT
<213> Homo sapiens
<220>
<221> ιrtisc_f eature
<223> Incyte ID No : 490576CD1
<400> 16
Met Pro Glu Gin Ser Asn Asp Tyr Arg Val Ala Val Phe Gly Ala
1 5 10 15
Gly Gly Val Gly Lys Ser Ser Leu Val Leu Arg Phe Val Lys Gly
20 25 30
Thr Phe Arg Glu Ser Tyr He Pro Thr Val Glu Asp Thr Tyr Arg
35 40 45
Gin Val He Ser Cys Asp Lys Ser He Cys Thr Leu Gin He Thr
50 55 60
Asp Thr Thr Gly Ser His Gin Phe Pro Ala Met Gin Arg Leu Ser
65 70 75
He Ser Lys Gly His Ala Phe He Leu Val Tyr Ser He Thr Ser
80 85 90
Arg Gin Ser Leu Glu Glu Leu Lys Pro He Tyr Glu Gin He Cys
95 100 105
Glu He Lys Gly Asp Val Glu Ser He Pro He Met Leu Val Gly
110 115 120
Asn Lys Cys Asp Glu Ser Pro Ser Arg Glu Val Gin Ser Ser Glu
125 130 135
Ala Glu Ala Leu Ala Arg Thr Trp Lys Cys Ala Phe Met Glu Thr
140 145 150
Ser Ala Lys Leu Asn His Asn Val Lys Glu Leu Phe Gin Glu Leu
155 160 165
Leu Asn Leu Glu Lys Arg Arg Thr Val Ser Leu Gin lie Asp Gly
170 175 180
Lys Lys Ser Lys Gin Gin Lys Arg Lys Glu Lys Leu Lys Gly Lys 185 190 195
Cys Val He Met
<210> 17
<211> 790
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1417657CD1
<400> 17
Met Glu Lys Met Ser Arg Val Thr Thr Ala Leu Gly Gly Ser Val
1 5 10 15
Leu Thr Gly Arg Thr Met His Cys His Leu Asp Ala Pro Ala Asn
20 25 30
Ala He Ser Val Cys Arg Asp Ala Ala Gin Val Val Val Ala Gly
35 40 45
Arg Ser He Phe Lys He Tyr Ala He Glu Glu Glu Gin Phe Val
50 55 60
Glu Lys Leu Asn Leu Arg Val Gly Arg Lys Pro Ser Leu Asn Leu
65 70 75
Ser Cys Ala Asp Val Val Trp His Gin Met Asp Glu Asn Leu Leu
80 85 90
Ala Thr Ala Ala Thr Asn Gly Val Val Val Thr Trp Asn Leu Gly
95 100 105
Arg Pro Ser Arg Asn Lys Gin Asp Gin Leu Phe Thr Glu His Lys
110 115 120
Arg Thr Val Asn Lys Val Cys Phe His Pro Thr Glu Ala His Val
125 130 135
Leu Leu Ser Gly Ser Gin Asp Gly Phe Met Lys Cys Phe Asp Leu
140 145 150
Arg Arg Lys Asp Ser Val Ser Thr Phe Ser Gly Gin Ser Glu Ser
155 160 165
Val Arg Asp Val Gin Phe Ser He Arg Asp Tyr Phe Thr Phe Ala
170 175 180
Ser Thr Phe Glu Asn Gly Asn Val Gin Leu Trp Asp He Arg Arg
185 190 195
Pro Asp Arg Cys Glu Arg Met Phe Thr Ala His Asn Gly Pro Val
200 205 210
Phe Cys Cys Asp Trp His Pro Glu Asp Arg Gly Trp Leu Ala Thr
215 220 225
Gly Gly Arg Asp Lys Met Val Lys Val Trp Asp Met Thr Thr His
230 " 235 240
Arg Ala Lys Glu Met His Cys Val Gin Thr He Ala Ser Val Ala
245 250 255
Arg Val Lys Trp Arg Pro Glu Cys Arg His His Leu Ala Thr Cys
260 265 270
Ser Met Met Val Asp His Asn He Tyr Val Trp Asp Val Arg Arg
275 280 285
Pro Phe Val Pro Ala Ala Met Phe Glu Glu His Arg Asp Val Thr
290 295 300
Thr Gly He Ala Trp Arg His Pro His Asp Pro Ser Phe Leu Leu
305 310 315
Ser Gly Ser Lys Asp Ser Ser Leu Cys Gin His Leu Phe Arg Asp
320 325 330 Ala Ser Gin Pro Val Glu Arg Ala Asn Pro Glu Gly Leu Cys Tyr
335 340 345
Gly Leu Phe Gly Asp Leu Ala Phe Ala Ala Lys Glu Ser Leu Val
350 355 360
Ala Ala Glu Ser Gly Arg Lys Pro Tyr Thr Gly Asp Arg Arg His
365 370 375
Pro He Phe Phe Lys Arg Lys Leu Asp Pro Ala Glu Pro Phe Ala
380 385 390
Gly Leu Ala Ser Ser Ala Leu Ser Val Phe Glu Thr Glu Pro Gly
395 400 405
Gly Gly Gly Met Arg Trp Phe Val Asp Thr Ala Glu Arg Tyr Ala
410 415 420
Leu Ala Gly Arg Pro Leu Ala Glu Leu Cys Asp His Asn Ala Lys
425 430 435
Val Ala Arg Glu Leu Gly Arg Asn Gin Val Ala Gin Thr Trp Thr
440 445 450
Met Leu Arg He He Tyr Cys Ser Pro Gly Leu Val Pro Thr Ala
455 460 - 465
Asn Leu Asn His Ser Val Gly Lys Gly Gly Ser Cys Gly Leu Pro
470 475 480
Leu Met Asn Ser Phe Asn Leu Lys Asp Met Ala Pro Gly Leu Gly
485 490 495
Ser Glu Thr Arg Leu Asp Arg Ser Lys Gly Asp Ala Arg Ser Asp
500 505 510
Thr Val Leu Leu Asp Ser Ser Ala Thr Leu He Thr Asn Glu Asp
515 520 525
Asn Glu Glu Thr Glu Gly Ser Asp Val Pro Ala Asp Tyr Leu Leu
530 535 540
Gly Asp Val Glu Gly Glu Glu Asp Glu Leu Tyr Leu Leu Asp Pro
545 550 555
Glu His Ala His Pro Glu Asp Pro Glu Cys Val Leu Pro Gin Glu
560 565 570
Ala Phe Pro Leu Arg His Glu He Val Asp Thr Pro Pro Gly Pro
575 580 585
Glu His Leu Gin Asp Lys Ala Asp Ser Pro His Val Ser Gly Ser
590 595 600
Glu Ala Asp Val Ala Ser Leu Ala Pro Val Asp Ser Ser Phe Ser
605 610 615
Leu Leu Ser Val Ser His Ala Leu Tyr Asp Ser Arg Leu Pro Pro
620 625 630
Asp Phe Phe Gly Val Leu Val Arg Asp Met Leu His Phe Tyr Ala
635 640 645
Glu Gin Gly Asp Val Gin Met Ala Val Ser Val Leu He Val Leu
650 655 660
Gly Glu Arg Val Arg Lys Asp He Asp Glu Gin Thr Gin Glu His
665 670 675
Trp Tyr Thr Ser Tyr He Asp Leu Leu Gin Arg Phe Arg Leu Trp
680 685 690
Asn Val Ser Asn Glu Val Val Lys Leu Ser Thr Ser Arg Ala Val
695 700 705
Ser Cys Leu Asn Gin Ala Ser Thr Thr Leu His Val Asn Cys Ser
710 715 720
His Cys Lys Arg Pro Met Ser Ser Arg Gly Trp Val Cys Asp Arg
725 730 735
Cys His Arg Cys Ala Ser Met Cys Ala Val Cys His His Val Val
740 745 750
Lys Gly Leu Phe Val Trp Cys Gin Gly Cys Ser His Gly Gly His
755 760 765 Leu Gin His He Met Lys Trp Leu Glu Gly Ser Ser His Cys Pro 770 775 780
Ala Gly Cys Gly His Leu Cys Glu Tyr Ser 785 790
<210> 18
<211> 490
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1773215CD1
<400> 18
Met Glu Glu Glu Gly Val Lys Glu Ala Gly Glu Lys Pro Arg Gly
1 5 10 15
Ala Gin Met Val Asp Lys Ala Gly Trp He Lys Lys Ser Ser Gly
20 25 30
Gly Leu Leu Gly Phe Trp Lys Asp Arg Tyr Leu Leu Leu Cys Gin
35 40 45
Ala Gin Leu Leu Val Tyr Glu Asn Glu Asp Asp Gin Lys Cys Val
50 55 60
Glu Thr Val Glu Leu Gly Ser Tyr Glu Lys Cys Gin Asp Leu Arg
65 70 75
Ala Leu Leu Lys Arg Lys His Arg Phe He Leu Leu Arg Ser Pro
80 85 90
Gly Asn Lys Val Ser Asp He Lys Phe Gin Ala Pro Thr Gly Glu
95 100 105
Glu Lys Glu Ser Trp He Lys Ala Leu Asn Glu Gly He Asn Arg
110 115 120
Gly Lys Asn Lys Ala Phe Asp Glu Val Lys Val Asp Lys Ser Cys
125 130 135
Ala Leu Glu His Val Thr Arg Asp Arg Val Arg Gly Gly Gin Arg
140 145 150
Arg Arg Pro Pro Thr Arg Val His Leu Lys Glu Val Ala Ser Ala
155 160 165
Ala Ser Asp Gly Leu Leu Arg Leu Asp Leu Asp Val Pro Asp Ser
170 175 180
Gly Pro Pro Val Phe Ala Pro Ser Asn His Val Ser Glu Ala Gin
185 190 195
Pro Arg Glu Thr Pro Arg Pro Leu Met Pro Pro Thr Lys Pro Phe
200 205 210
Leu Ala Pro Glu Thr Thr Ser Pro Gly Asp Arg Val Glu Thr Pro
215 220 225
Val Gly Glu Arg Ala Pro Thr Pro Val Ser Ala Ser Ser Glu Val
230 235 240
Ser Pro Glu Ser Gin Glu Asp Ser Glu Thr Pro Ala Glu Glu Asp
245 250 255
Ser Gly Ser Glu Gin Pro Pro Asn Ser Val Leu Pro Asp Lys Leu
260 265 270
Lys Val Ser Trp Glu Asn Pro Ser Pro Gin Glu Ala Pro Ala Ala
275 280 285
Glu Ser Ala Glu Pro Ser Gin Ala Pro Cys Ser Glu Thr Ser Glu
290 295 300
Ala Ala Pro Arg Glu Gly Gly Lys Pro Pro Thr Pro Pro Pro Lys
305 310 315
He Leu Ser Glu Lys Leu Lys Ala Ser Met Gly Glu Met Gin Ala 320 325 330
Ser Gly Pro Pro Ala Pro Gly Thr Val Gin Val Ser Val Asn Gly
335 340 345
Met Asp Asp Ser Pro Glu Pro Ala Lys Pro Ser Gin Ala Glu Gly
350 355 360
Thr Pro Gly Thr Pro Pro Lys Asp Ala Thr Thr Ser Thr Ala Leu
365 370 375
Pro Pro Trp Asp Leu Pro Pro Gin Phe His Pro Arg Cys Ser Ser
380 385 390
Leu Gly Asp Leu Leu Gly Glu Gly Pro Arg His Pro Leu Gin Pro
395 400 405
Arg Glu Arg Leu Tyr Arg Ala Gin Leu Glu Val Lys Val Ala Ser
410 415 420
Glu Gin Thr Glu Lys Leu Leu Asn Lys Val Leu Gly Ser Glu Pro
425 430 435
Ala Pro Val Ser Ala Glu Thr Leu Leu Ser Gin Ala Val Glu Gin
440 445 450
Leu Arg Gin Ala Thr Gin Val Leu Gin Glu Met Arg Asp Leu Gly
455 460 465
Glu Leu Ser Gin Glu Ala Pro Gly Leu Arg Glu Lys Arg Lys Glu
470 475 480
Leu Val Thr Leu Tyr Arg Arg Ser Ala Pro
485 490
<210> 19
<211> 914
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 3036986CD1
<400> 19
Met Ala Asn He Asn Leu Lys Glu He Thr 'Leu He Val Gly Val
1 5 10 15
Val Thr Ala Cys Tyr Trp Asn Ser Leu Phe Cys Gly Phe Val Phe
20 25 30
Asp Asp Val Ser Ala He Leu Asp Asn Lys Asp Leu His Pro Ser
35 40 45
Thr Pro Leu Lys Thr Leu Phe Gin Asn Asp Phe Trp Gly Thr Pro
50 55 60
Met Ser Glu Glu Arg Ser His Lys Ser Tyr Arg Pro Leu Thr Val
65 70 75
Leu Thr Phe Arg Leu Asn Tyr Leu Leu Ser Glu Leu Lys Pro Met
80 85 90
Ser Tyr His Leu Leu Asn Met He Phe His Ala Val Val Ser Val
95 100 105
He Phe Leu Lys Val Cys Lys Leu Phe Leu Asp Asn Lys Ser Ser
110 115 120
Val He Ala Ser Leu Leu Phe Ala Val His Pro He His Thr Glu
125 130 135
Ala Val Thr Gly Val Val Gly Arg Ala Glu Leu Leu Ser Ser He
140 145 150
Phe Phe Leu Ala Ala Phe Leu Ser Tyr Thr Arg Ser Lys Gly Pro
155 160 165
Asp Asn Ser He He Trp Thr Pro He Ala Leu Thr Val Phe Leu
170 175 180 Val Ala Val Ala Thr Leu Cys Lys Glu Gin Gly He Thr Val Val
185 190 195
Gly He Cys Cys Val Tyr Glu Val Phe He Ala Gin Gly Tyr Thr
200 205 210
Leu Pro Leu Leu Cys Thr Thr Ala Gly Gin Phe Leu Arg Gly Lys
215 220 225
Gly Ser He Pro Phe Ser Met Leu Gin Thr Leu Val Lys Leu He
230 235 240
Val Leu Met Phe Ser Thr Leu Leu Leu Val Val He Arg Val Gin
245 250 255
Val He Gin Ser Gin Leu Pro Val Phe Thr Arg Phe Asp Asn Pro
260 265 270 Ala Ala Val Ser Pro Thr Pro Thr Arg Gin Leu Thr Phe Asn Tyr
275 280 285
Leu Leu Pro Val Asn Ala Trp Leu Leu Leu Asn Pro Ser Glu Leu
290 295 300
Cys Cys Asp Trp Thr Met Gly Thr He Pro Leu He Glu Ser Leu
305 310 315
Leu Asp He Arg Asn Leu Ala Thr Phe Thr Phe Phe Cys Phe Leu
320 325 330
Gly Met Leu Gly Val Phe Ser He Arg Tyr Ser Gly Asp Ser Ser
335 340 345
Lys Thr Val Leu Met Ala Leu Cys Leu Met Ala Leu Pro Phe He
350 355 ' 360
Pro Ala Ser Asn Leu Phe Phe Pro Val Gly Phe Val Val Ala Glu
365 370 375
Arg Val Leu Tyr Val Pro Ser Met Gly Phe Cys He Leu Val Ala
380 385 390
His Gly Trp Gin Lys He Ser Thr Lys Ser Val Phe Lys Lys Leu
395 400 405
Ser Trp He Cys Leu Ser Met Val He Leu Thr His Ser Leu Lys
410 415 420 Thr Phe His Arg Asn Trp Asp Trp Glu Ser Glu Tyr Thr Leu Phe
425 430 435
Met Ser Ala Leu Lys Val Asn Lys Asn Asn Ala Lys Leu Trp Asn
440 445 450
Asn Val Gly His Ala Leu Glu Asn Glu Lys Asn Phe Glu Arg Ala
455 460 465
Leu Lys Tyr Phe Leu Gin Ala Thr His Val Gin Pro Asp Asp He
470 475 480
Gly Ala His Met Asn Val Gly Arg Thr Tyr Lys Asn Leu Asn Arg
485 490 495
Thr Lys Glu Ala Glu Glu Ser Tyr Met Met Ala Lys Ser Leu Met
500 505 510
Pro Gin He He Pro Gly Lys Lys Tyr Ala Ala Arg He Ala Pro
515 520 525
Asn His Leu Asn Val Tyr He Asn Leu Ala Asn Leu He Arg Ala
530 535 540 Asn Glu Ser Arg Leu Glu Glu Ala Asp Gin Leu Tyr Arg Gin Ala
545 550 555
He Ser Met Arg Pro Asp Phe Lys Gin Ala Tyr He Ser Arg Gly
560 565 570
Glu Leu Leu Leu Lys Met Asn Lys Pro Leu Lys Ala Lys Glu Ala
575 580 585
Tyr Leu Lys Ala Leu Glu Leu Asp Arg Asn Asn Ala Asp Leu Trp
590 595 600
Tyr Asn Leu Ala He Val His He Glu Leu Lys Glu Pro Asn Glu
605 610 615 Ala Leu Lys Asn Phe Asn Arg Ala Leu Glu Leu Asn Pro Lys His
620 625 630
Lys Leu Ala Leu Phe Asn Ser Ala He Val Met Gin Glu Ser Gly
635 640 645
Glu Val Lys Leu Arg Pro Glu Ala Arg Lys Arg Leu Leu Ser Tyr
650 655 660
He Asn Glu Glu Pro Leu Asp Ala Asn Gly Tyr Phe Asn Leu Gly
665 670 675
Met Leu Ala Met Asp Asp Lys Lys Asp Asn Glu Ala Glu He Trp
680 685 690
Met Lys Lys Ala He Lys Leu Gin Ala Asp Phe Arg Ser Ala Leu
695 700 705
Phe Asn Leu Ala Leu Leu Tyr Ser Gin Thr Ala Lys Glu Leu Lys
710 715 720
Ala Leu Pro He Leu Glu Glu Leu Leu Arg Tyr Tyr Pro Asp His
725 730 735
He Lys Gly Leu He Leu Lys Gly Asp He Leu Met Asn Gin Lys
740 745 750
Lys Asp He Leu Gly Ala Lys Lys Cys Phe Glu Arg He Leu Glu
755 760 765
Met Asp Pro Ser Asn Val Gin Gly Lys His Asn Leu Cys Val Val
770 775 780
Tyr Phe Glu Glu Lys Asp Leu Leu Lys Ala Glu Arg Cys Leu Leu
785 790 795
Glu Thr Leu Ala Leu Ala Pro His Glu Glu Tyr He Gin Arg His
800 805 810
Leu Asn He .Val Arg Asp Lys He Ser Ser Ser Ser Phe He Glu
815 820 825
Pro He Phe Pro Thr Ser Lys He Ser Ser Val Glu Gly Lys Lys
830 835 840
He Pro Thr Glu Ser Val Lys Glu He Arg Gly Glu Ser Arg Gin
845 850 855
Thr Gin He Val Lys Thr Ser Asp Asn Lys Ser Gin Ser Lys Ser
860 865 870
Asn Lys Gin Leu Gly Lys Asn Gly Asp Glu Glu Thr Pro His Lys
875 880 885
Thr Thr Lys Asp He Lys Glu He Glu Lys Lys Arg Val Ala Ala
, 890 895 900 Leu Lys Arg Leu Glu Glu He Glu Arg He Leu Asn Gly Glu
905 910
<210> 20
<211> 349
<212> PRT
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 2041080CD1
<400> 20
Met Glu Glu Glu Lys Tyr Leu Pro Glu Leu Met Ala Glu Lys Asp
1 5 10 15
Ser Leu Asp Pro Ser Phe Val His Ala Ser Arg Leu Leu Ala Glu
20 25 30
Glu He Glu Lys Phe Gin Gly Ser Asp Gly Lys Lys Glu Asp Glu
35 40 45
Glu Lys Lys Tyr Leu Asp Val He Ser Asn Lys Asn He Lys Leu 50 55 60
Ser Glu Arg Val Leu He Pro Val Lys Gin Tyr Pro Lys Phe Asn
65 70 75
Phe Val Gly Lys Leu Leu Gly Pro Arg Gly Asn Ser Leu Lys Arg
80 85 90
Leu Gin Glu Glu Thr Gly Ala Lys Met Ser He Leu Gly Lys Gly
95 100 105
Ser Met Arg Asp Lys Ala Lys Glu Glu Glu Leu Arg Lys Ser Gly
110 115 120
Glu Ala Lys Tyr Ala His Leu Ser Asp Glu Leu His Val Leu He
125 130 135
Glu Val Phe Ala Pro Pro Gly Glu Ala Tyr Ser Arg Met Ser His
140 145 150
Ala Leu Glu Glu He Lys Lys Phe Leu Val Pro Asp Tyr Asn Asp
155 160 165
Glu He Arg Gin Glu Gin Leu Arg Glu Leu Ser Tyr Leu Asn Gly
170 175 180
Ser Glu Asp Ser Gly Arg Gly Arg Gly He Arg Gly Arg Gly He
185 190 195
Arg He Ala Pro Thr Ala Pro Ser Arg Gly Arg Gly Gly Ala He
200 205 210
Pro Pro Pro Pro Pro Pro Gly Arg Gly Val Leu Thr Pro Arg Gly
215 220 225
Ser Thr Val Thr Arg Gly Ala Leu Pro Val Pro Pro Val Ala Arg
230 235 240
Gly Val Pro Thr Pro Arg Ala Arg Gly Ala Pro Thr Val Pro Gly
245 250 255
Tyr Arg Ala Pro Pro Pro Pro Ala His Glu Ala Tyr Glu Glu Tyr
260 265 270
Gly Tyr Asp Asp Gly Tyr Gly Gly Glu Tyr Asp Asp Gin Thr Tyr
275 280 285
Glu Thr Tyr Asp Asn Ser Tyr Ala Thr Gin Thr Gin Ser Val Pro
290 295 300
Glu Tyr Tyr Asp Tyr Gly His Gly Val Ser Glu Asp Ala Tyr Asp
305 310 315
Ser Tyr Ala Pro Glu Glu Trp Ala Thr Thr Arg Ser Ser Leu Lys
320 325 330
Ala Pro Pro Gin Arg Ser Ala Arg Gly Gly Tyr Arg Glu His Pro
335 340 345 Tyr Gly Arg Tyr
<210> 21
<211> 2860
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 105283CB1
<400> 21 gccagagcgg cggccggtcc cgcgcggagc ccggcgcccc tccagcccga gccaggacgc 60 cgccggcccc ggtcccggcc ccgggcacgc agcgagccag ggatgtgagc ggcgσcccgσ 120 ggcatggcag cctcaggggt gcccagagga tgcgacatcc tcatcgtcta cagcccggat 180 gccgaggaat ggtgccagta cctgcagacc ctgttcctgt ccagtcggca ggtccgcagc 240 cagaagatac tgactcacag gctgggcccc gaggcctcct tctcggcaga ggacctaagc 300 cttttcctca gcaccσgctg tgtcgtggtg ctgctgtcσg cggagctggt gσagcaσttc 360 cacaagσccg ccttgctgcc cctgctgcag agagctttcc atcctcσgca ccgcgtggtc 420 aggctgctct gcggcgtgσg ggacagcgag gagttcctag acttctttcc agattgggcc 480 cattggcagg agctcacctg tgacgatgag ccagagacct acgtggcagc tgtgaaaaaa 540 gccatttccg aagattσtgg ctgtgactσa gtcactgaca ctgagcctga ggacgagaag 600 gttgtttcct actcgaagca gcagaacctg ccgacggtga cttcacσtgg gaacσtgatg 660 gtggtgσagσ cggaσσgσat tcgctgtggg gσagaaacca ctgtctatgt tattgtgaga 720 tgtaagctgg atgacagggt ggcgacagaa gcagagtttt ctcctgagga ttctccctct 780 gtaaggatgg aagcσaaggt ggagaatgag taσaσσattt σagtgaaggc tcccaacσtt 840 tcatctggga acgtttctct gaagatatat tctggagact tagtggtgtg tgaaaccgtt 900 atcagctatt atactgacat ggaagaaatt gggaatttat tgtccaatgc cgcgaatcct 960 gtggaattca tgtgtcaggc ctttaaaatt gtgccctaca acacagagac ccttgataaa 1020 ctgctaaccg aatcσσtgaa gaacaatatσ cσtgσaagcg gactgcacct σtttggaatc 1080 aaccagctgg aagaagaaga tatgatgaca aatcagaggg atgaagagct gcccaccctg 1140 ttgcattttg ctgσgaagta tggactgaag aacctcactg cσttgttgct cacctgccca 1200 ggagccctgc aggcgtacag cgtggccaac aagcatggcc actaccccaa caccatcgct 1260 gagaaacacg gcttcaggga cctgcggcag ttcatcgacg agtatgtgga aacggtggac 1320 atgctcaaga gtcacattaa agaggaactg atgcacgggg aggaggctga tgctgtgtac 1380 gagtcσatgg cσσaσσtttσ cacagacctg cttatgaaat gctcgctcaa cσcσggσtgt 1440 gacgaggatc tctatgagtc catggctgcc tttgtcccag ctgccactga agacctctat 1500 gttgaaatgc ttσaggσσag taσatσtaaσ σσaatccctg gagatggttt ctctcgggcσ 1560 actaaggact ctatgatccg caagttttta gaaggcaaca gcatgggaat gaccaatctg 1620 gagagagatc agtgccatct tggtσaggaa gaagatgttt atcaσaσggt ggatgaσgat 1680 gaggcctttt ctgtggactt ggccagcagg ccccctgtcc cagtgcccag accagagacc 1740 actgctσσtg gtgσtσaσσa gσtgσctgac aaσgaaσσat acatttttaa agtttttgca 1800 gaaaaaagtc aagagcggcc tgggaatttc tacgtttcct cagagagcat caggaaaggg 1860 cσgcccgtca gaσcatggag ggacaggσσσ σagtcgagta tatatgaccσ ttttgσggga 1920 atgaaaacgc caggccagcg gcagcttatc accctccagg agcaggtgaa gctgggcatt 1980 gtcaacgtgg atgaggctgt gctcσacttc aaagagtggc agctcaacca gaagagaσga 2040 tcggagtcct ttcgtttcca gcaggaaaat cttaaacggc taagagacag catcaσccga 2100 agacagagag agaagcaaaa atcaggaaag cagacagact tggagatcac ggtcccaatt 2160 cggcactcac agcacctgcc tgcaaaagtg gagtttggag tctatgagag tggccccagg 2220 aaaagtgtca ttccccctag gacggagσtg agaσgaggag actggaaaac agacagcacc 2280 tccagcacag caagtagcac aagtaaccgc tccagcaccc ggagcctcct cagtgtgagc 2340 agcgggatgg aaggggacaa cgaggataat gaagtccctg aggttaccag aagtcgcagt 2400 ccaggccccc cacaagtgga tgggacaccc accatgtccc tcgagagacc ccccagggtg 2460 cσtccgagag σtgσσtσaσa gaggcctccg accagggaga ccttccatcc tσctcσaσσt 2520 gttccacσca gaggacgctg attccacctc ctaaaacctg cctacttcag gactttaaga 2580 ctσaσagtct tσagσσtgtt aatgatgtσt tσatgttgag ttttatagca tgactgttga 2640 ccttaagatc cattctcatt gctgataatg ctgcagccct gctggtttgg gcttgcctcg 2700 aagattttat taaggcaσga agaagtgaaa aactaagggc ttcattcaσσ atcaccaagt 2760 atatcgaacc atatacttgt ttgccaaaag gatgaagact taatcgaaat acttacctct 2820 aatttgccat atcagaagσσ taaaaagaat gatσataaat 2860
<210> 22
<211> 3542
<212> DNA
<213> Homo sapiens
<220>
<221> misσ_feature
<223> Incyte ID No : 3350821CB1
<400> 22 atgtgttgca σtgagggσag tctcσggaaa cgσgattσgσ agcgggcgcσ ggaagcggtg 60 ttgtgtctgc agctctggca gaggactgtt ccactagaca cgctgaaggg actgggtacg 120 tgttttcσtt caggaσcaga gctgagagga gctgggatσg σggσggσaat ggaaσgggσσ 180 tcagaaaggc gcacggccag cgcgcttttt gcggggttcc gggccttggg acttttcagc 240 aacgacattc cacaσgtggt gcggttcagσ gσgctcaagc gccggttcta tgtaacaacc 300 tgcgtgggca agagtttcca cacctatgac gttcagaaac ttagtctggt tgcagtaagt 360 aattctgttc cacaggatat ctgctgtatg gcagσtgatg gσagattagt ctttgctgct 420 tatggaaatg ttttctctgc atttgcccgt aataaagaga tagtaσatac ctttaagggt 480 cataaggcag aaatcσattt cttgcaaccc tttggagacc acattatctc tgttgatact 540 gatggcattc ttattatttg gcacatatat tcagaagaag aatacctgca gttgactttt 600 gataaatcag tatttaaaat ttσtgcaatt ttgcatσσaa gtacctactt gaataaaata 660 cttctgggca gtgaacaagg aagcctgcag ttgtggaatg taaaatccaa ccagaaatac 720 ccaatcaggc agacatttat ccctgcgggc tatttgttgg gtgcaσatgg gttgaagaσt 780 caggcaccag ccgtggatgt tgttgctatt ggtcttatgt caggtcaagt tatcattcac 840 aacattaaat ttaatgaaaσ attaatgaag tttσgtσaag actggggacσ σattaσttσa 900 atttcatttc gcacagatgg tcatccagta atggcagctg gaagcccatg tggccatatt 960 ggactctggg atσtagaaga caaaaaatta atcaaccaaa tgagaaatgc acaσtctaca 1020 gcaattgccg gactgacatt tctcσataga gagccacttσ ttgtcacaaa tggcgctgac 1080 aatgctctta ggatatggat atttgatggt cctacaggtg aaggccgact tttgagattc 1140 agaafcgggtc atagtgctcc tcttaccaat atcagatatt atggacagaa tggacagcag 1200 attσtaagtg σaagtcaaga tggaaσtσtt cagtσatttt ccacggtaca tgaaaaattσ 1260 aataagagσt tgggacatgg attaataaat aaaaagagag ttaaaσgtaa aggacttcag 1320 aataccatgt cagtgagact tσσaσσcatσ acaaagtttg σagσagagga agctcgtgaa 1380 agtgactggg atggtatcat tgcttgccat caaggtaagc tatcttgctc aacσtggaat 1440 tatσagaaat σtaσaatagg cgcttacttt ctcaagccaa aagagttgaa gaaagatgac 1500 ataactgcaa cagcagtgga tataacttct tgtggaaact ttgctgtaat tggcctctca 1560 tcaggaactg tagatgtata taacatgcag tσtggσatac atcgaggaag ttttggσaag 1620 gatcaagctc acaagggatc tgttagaggt gtcgcagtgg atggattaaa ccagttgaca 1680 gttaσaactg gtagtgaagg attactσaaa ttctggaact ttaaaaacaa aattttaatc 1740 cattctgtga gcctσagttc atctcσaaat atcatgttgc tacataggga cagtggσatt 1800 ctgggactσg σσttggatga cttctccatt agtgttctgg acatagaagc taggaagatt 1860 gtcagagagt tttctggaca ccaaggccaa ataaatgaca tggcttttag tcctgatggt 1920 cgttggttaa taagtgctgc gatggattgσ tσtattagga cttgggaσσt tccttctggg 1980 tgccttatag actgcttttt gttggactcg gctcctctca atgtttσtat gtctcctact 2040 ggagactttc tggcaacttc ccatgtggac cacσttggaa tttatσtatg gtccaatatt 2100 tccctgtatt cagttgtttc attacggcca cttcctgσag attatgtσσσ ttcaatagtσ 2160 atgσfctσσtg gtacttgtca aacccaagat gtagaagttt cagaagaaac agtagaacca 2220 agtgatgaat tgatagaata tgattcgcca gaacagttga atgagcaatt ggtgactctt 2280 tcaσttcttc ctgaatcacg atggaaaaac cttσttaaσc ttgatgttat taagaaaaag 2340 aataaaccaa aggaaccacc caaagtacσσ aaatcagσac catttttcat tccaacaatt 2400 cσtggccttg tacccagata tgctgσaσσt gaacaaaata atgatσσσσa gcagtctaaa 2460 gtggtaaatc ttggagtttt ggctσaaaaa tcagatttct gcttgaaact tgaagaagga 2520 ctggtaaata ataagtatga σaσtgctctc aaccttctga aagaatcagg cσcatcagga 2580 attgaaacag agctgcgaag cttgtσtσσt gattgtggtg ggtccataga agttatgσag 2640 agσttcttga aaatgattgg gatgatgctg gacagaaagc gtgattttga gttagcσcag 2700 gcataccttg cattgtttct aaagttacac cttaaaatgc ttcσttσaga gσσagtactc 2760 ctagaagaaa taacaaattt gtcatcσσag gtggaagaaa actggaccca tttgcaatσa 2820 ctσttcaatc aaagcatgtg tattttaaat tatctσaaaa gtgctttgtt gtaaaaataa 2880 atttgtgaσt aaaσaaagac tttcatatta aatgggttca attgaactca tttcttattt 2940 tccaagtgtc aatgtgaaaa gaaaataaat gctagcacta ctgactagtc agtatatctc 3000 cactttaaat gctaaatact ttcttgaaat aaaatcgcac ctσccggcca ggcatgatgg 3060 ataatgcctg taattccagc actttcagag gccaaagcgg gaggattgct tgaagccagg 3120' aatttgagcc cagcctgggc aacatagcaa gcaagatσσσ atctctacaa aaagttaaaa 3180 aaataaatta cacctgcσtt tgttttgaag acttaaagaa tctgctcaaa tgatttattt 3240 tctgagtctc ttcaσattaa cttttatatg atttttaaaa aattattacc tgcttatatt 3300 tttgagtatc tgtttcatat aaagagatgc tgσatgttσa σtgtgaagta ttgctgaatt 3360 aaattgaaaa tgtcagataa aaσtgataσa tgatgtagtt ataaaaatgσ tgttatgaag 3420 aaaaaaggaa tgcttttttσ ctcggtggcc tttgataaaa atatgaatga ttgactctca 3480 gagtggagga taaactaatc atcttctagσ aσagccσtgt gtgtgcagga gaggaatcag 3540 gg 3542
<210> 23 <211> 1014 <212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 5876846CB1
<400> 23 ggacccggcg ccgatgaaac cgaccaaact ggagttagct accggactgt cccgcgagcg 60 aaagttggaa ataatacσta aacctacttσ ttgaσatggt gtσaaaaσag agσttttgσt 120 gagagtttga tttataaaga aattatggtt cgactgacct tggatctaat tgccagaaac 180 agcaatctta aaσσσσgaaa agaagaaacc atttcacagt gcctgaagaa aataactcat 240 ataaattttt cagacaaaaa tatagatgca attgaagacc tctctctttg caaaaatctt 300 agtgttttat atttatatga taattgtatt agtcaaatca ctaacctgaa ttatgccaca 360 aatctgaccc acttgtacct acaaaacaat tgtatttcat gtatagagaa cctcaggtca 420 ttaaagaaaσ tggaaaaact gtatσtggga ggσaattaσa ttgσtgtσat agaaggttta 480 gaaggattag gagaactaag agagcttcat gttgagaatc agaggcttcc ccttggggaa 540 aagctfcσtgt ttgatcσaag aactσttσat tσtctggσaa aatccctctg tatattgaat 600 atcagcaata ataatattga tgacattaca gacttagaac tactagagaa tcttaatcag 660 ctcatagσσg ttgaσaacca acttctgσat gtgaaggatt tggagttttt actgaaσaag 720 ttgatgaagc tgtggaaaat tgatctaaat ggaaatcctg tttgtctcaa accaaaatac 780 agggaσagac tgatattggt gtσσaaatσa σtggaatttσ ttgatggaaa agaaattaaa 840 aatatagaaa gacaattcct aatgaattgg aaagcatcca aagatgccaa gaaaatcagc 900 aaaaaaagga gcagtaaaaa tgaggatgσa agcaattσaσ tcataagtaa gσactctgtc 960 actcattagg tgactgctct tctagcaaag gaaatattct aggcaacaaa aaaa 1014
<210> 24
<211> 4040
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 3560269CB1
<400> 24 gcactgcggc acagagccgc ggcttgcttt caggacacgg gtcgctggcg ggttaggtgg 60 ctgσgtggcc gcctσaσttc gcgcaσgcgc cgctaggσtg ggggagttga tttgcactgc 120 tccgggtgtc ggctccggcg ggacgggcgg ttctgcgtac ccctctccag tgtcaacctg 180 gggctgaatc ttcaacctgt σaaagσσttc ttaattatgt ctgagaaaag taσtaaaata 240 ggtgccattt taaattccca gacaaggaaa aaccaaggat tagaaattaa gtagcttgcc 300 , caaggcσaσa σatcagatσσ tgattgagtσ aatgctttgσ attctcagtc ctctgcataa 360 agctgagaga tgcctacagc tgagagtgaa gcaaaagtaa aaaccaaagt tcgctttgaa 420 gaattgctta agacccacag tgatctaatg σgtgaaaaga aaaaaσtgaa gaaaaaaσtt 480 gtcaggtctg aagaaaacat ctcacctgac actattagaa gcaatcttca ctatatgaaa 540 gaaaσtaσaa gtgatgatcσ σgaσaσtatt agaagσaatσ ttσσσσatat taaagaaact 600 acaagtgatg atgtaagtgc tgctaacact aacaacctga agaagagcac gagagtcact 660 aaaaacaaat tgaggaacac aσagttagσa actgaaaatc ctaatggtga tgσtagtgta 720 gaggaagaca aacaaggaaa gccaaataaa aaggtgataa agacggtgcc ccagttgact 780 acacaagacσ tgaaaccgga aaσtσσtgag aataaggttg attσtacaca σσagaaaaca 840 catacaaagc cacagccagg cgttgatcat cagaaaagtg agaaggcaaa tgagggaaga 900 gaagagactg atttagaaga ggatgaagaa ttgatgcaag catatσagtg σσatgtaact 960 gaagaaatgg caaaggagat taagaggaaa ataagaaaga aactgaaaga acagttgact 1020 tactttcσσt σagataσttt attcσatgat gacaaactaa gcagtgaaaa aaggaaaaag 1080 aaaaaggaag ttccagtctt ctctaaagct gaaacaagta cattgaccat ctctggtgac 1140 acagttgaag gtgaacaaaa gaaagaatct tcagttagat cagtttcttσ agattctcat 1200 caagatgatg aaataagctc aatggaacaa agcacagaag acagcatgca agatgataca 1260 aaacctaaac σaaaaaaaaσ aaaaaagaag actaaagcag ttgcagataa taatgaagat 1320 gttgatggtg atggtgttca tgaaataaca agcσgagata gcccggttta tcccaaatgt 1380 ttgcttgatg atgaccttgt cttgggagtt tacattcacc gaactgatag acttaagtca 1440 gattttatga tttctcaccc aatggtaaaa attcatgtgg ttgatgagca taσtggtσaa 1500 tatgtcaaga aagatgatag tggacggcσt gtttcatctt actatgaaaa agagaatgtg 1560 gattatattc ttcctattat gacccagcσa tatgatttta aacagttaaa atcaagactt 1620 ccagagtggg aagaacaaat tgtatttaat gaaaattttc cσtatttgct tcgaggctct 1680 gatgagagtc ctaaagtcat σctgttcttt gagattcttg atttσttaag cgtggatgaa 1740 attaagaata attσtgaggt tcaaaaσcaa gaatgtggσt ttcggaaaat tgcσtgggca 1800 tttσttaagσ ttctgggagc caatggaaat gcaaacatca actcaaaact tcgcttgcag 1860 ctatattacc cacctaσtaa gcctσgatσσ σcattaagtg ttgttgaggσ atttgaatgg 1920 tggtcaaaat gtccaagaaa tcattaccca tcaacactgt acgtaactgt aagaggactg 1980 aaagttccag actgtataaa gccatcttac cgσtctatga tggctcttca ggaggaaaaa 2040 ggtaaaccag tgcattgtga acgtcaccat gagtcaagct cagtagacac agaacctgga 2100 ttagaagagt caaaggaagt aataaagtgg aaacgactσc ctgggσaggσ ttgccgtatc 2160 ccaaacaaac acctcttctc actaaatgca ggagaacgag gatgtttttg tcttgatttc 2220 tcccaσaatg gaagaatatt agσagσagσt tgtgσcagσσ gggatggata tccaattatt 2280 ttatatgaaa ttccttctgg acgtttcatg agagaattgt gtggccacct caatatcatt 2340 tatgatcttt cσtggtσaaa agatgatcac taσatσσtta cttσatcatσ tgatggcact 2400 gccaggatat ggaaaaatga aataaacaat acaaatactt tcagagtttt acctcatcct 2460 tcttttgttt acacggctaa attcσatσσa gctgtaagag agσtagtagt tacaggatgc 2520 tatgattcca tgatacggat atggaaagtt gagatgagag aagattctgc catattggtc 2580 σgacagtttg atgttσaσaa aagttttatc aaσtσacttt gttttgataσ tgaaggtσat 2640 catatgtatt caggagattg tacaggggtg attgttgttt ggaataccta tgtcaagatt 2700 aatgatttgg aacattcagt gσaccactgg aσtataaata aggaaattaa agaaactgag 2760 tttaagggaa ttccaataag ttatttggag attcatccca atggaaaacg tttgttaatc 2820 cataccaaag acagtacttt gagaattatg gatctccgga tattagtagc aaggaagttt 2880 gtaggagcag caaattatcg ggagaagatt catagtactt tgactccafcg tgggactttt 2940 ctgtttgσtg gaagtgagga tggtatagtg tatgtttgga acσσagaaac aggagaacaa 3000 gtagccatgt attctgactt gccattcaag tcacccattc gagacatttc ttatcatcca 3060 tttgaaaata tggttgcatt σtgtgcattt gggcaaaatg agσσaattct tσtgtatatt 3120 tacgatttcc atgttgccca gcaggaggct gaaatgttca aacgctacaa tggaacattt 3180 cσattacctg gaatacaσσa aagtσaagat gccctatgta σσtgtσσaaa actaσσccat 3240 caaggctctt ttcagattga tgaatttgtc cacactgaaa gttσttcaac gaagatgcag 3300 σtagtaaaac agaggcttga aaσtgtσaσa gaggtgataσ gttcctgtgσ tgcaaaagtc 3360 aacaaaaatc tctcatttac ttcaccacca gcagtttcct cacaacagtc taagttaaag 3420 cagtcaaaca tgσtgaσcgσ tσaagagatt ctacatσagt ttggtttcac tσagaσσggg 3480 attatcagca tagaaagaaa gccttgtaac catcaggtag atacagcacc aacggtagtg 3540 gσtσtttatg actacacagc gaatcgatca gatgaaσtaa ccatccatcg cggagaσatt 3600 atccgagtgt ttttcaaaga taatgaagac tggtggtatg gcagcatagg aaagggacag 3660 gaaggttatt ttσσagσtaa tcatgtggct agtgaaacac tgtatcaaga aσtgcctcσt 3720 gagataaagg agcgatcccc tcctttaagσ cctgaggaaa aaaσtaaaat agaaaaatct 3780 ccagσtσctc aaaaggtaaa ataaaacaaa acagσtσaca gagacaσσtc σttσttcσcc 3840 ctgcttctgc ctccatgagt aactacctat atgacgtgct gctgctgaga ccaaggaatg 3900 agtgagtaaa ggtgtttgga agtcaaatafc tggtcctagt taagtaagtg tccttttcaa 3960 gtaagtggta aactttgtta tgtggacccc cttctaactt aggattcata taatttgaac 4020 acaggσtaaa gσtgσtσtgg 4040
<210> 25
<211> 2006
<212> DNA
<213> Homo sapiens
<220>
<221> misσ_feature
<223> Incyte ID No: 4596874CB1
<400> 25 tcσgaσtggc cgccataσat ctctagagaa gctgaσttct ggaσagttct caaagaaσcg 60 cagcσgagcσ aaacaaggca actσcσaσσa ccccccaggc ccctgggagg agtcagagσt 120 tgactccaga cccagtgccc atcactggtc gcactgaggg tgaagtggca cggaggagga 180 ggaggggctc cggctggtct gtggtgagat ggcσtacσag gtggtggaga agggcgcggc 240 cctgggcacg ctggagtcgg agctgcagca gaggcaaagc aggctggcag ccctggaggc 300 ccgcgtggcg cagctgcgag aggcgcgggσ gcagσaggcc cagcaggtgg aggagtggcg 360 ggcgcagaat gcggtgcagc gggcagccta cgaggcgctg cgcgcgcacg tcgggctccg 420 ggaggcggσa ctgcgcaggc tσσaggaaga ggσgcgcgac ctgσtggaga ggctcgtgca 480 gcgcaaggcg cgcgccgcgg ccgagcgcaa cctgcgcaac gagcgccggg agcgggccaa 540 gcaggcgcgg gtgtcccagg agσtgaagaa ggctgccaag cggaccgtga gcatcagσga 600 gggcccggac accctaggcg atgggatgag ggagagaagg gagactctgg ctctggcccc 660 tgagcσagag cσcctggaga aggaagcttg tgagaagtgg aagaggccct tσaggtctgσ 720 ctcagccacc tccctgacgc tgtcccactg tgtggatgtg gtgaaggggc ttctggattt 780 taagaagagg agaggtcact caattggggg agσσcctgag cagcgatacc agatcatσcσ 840 tgtgtgtgtg gctgcccgac ttcctacccg ggctcaggat gtgctggatg cccacctctc 900 tgaggtcaat gσtgttcgtt ttggccccaa cagσagσσtc ctggcσactg gaggggctga 960 ccgcctgatc cacctctgga atgttgtggg aagtcgcctg gaggccaacc agaccctgga 1020 gggagctggt ggcagσatca cσagtgtgga ctttgaσσσσ tσgggctacσ aggttttagσ 1080 agcaacttac aaccaggctg cccagctctg gaaggtgggg gaggcacagt ccaaggagac 1140 actgtctgga cacaaggata aggtgacagc tgcσaaattc aagσtaaσga ggcaccaggc 1200 agtgactggg agccgcgacc ggacagtgaa ggagtgggac ctcggccgtg cctattgctc 1260 caggaccatc aatgtσcttt cctaσtgtaa tgacgtggtg tgtggggaσσ atatσatcat 1320 tagtggccac aatgaccaga agatccggtt ctgggacagc agggggcccc actgcaccca 1380 ggtcatcσσt gtgcagggσσ gggtσaσctc cctgagcσtσ agccacgaσc aactgcacct 1440 gctcagctgt tcccgagaca acacactcaa ggtcatcgac ctgcgtgtca gcaacatccg 1500 ccaggtgttc agggcσgatg gσttcaagtg tggttσtgac tggacσaaag ctgtgttcag 1560 cccggacaga agctatgcac tggcaggctc ctgtgatggg gccctttaca tcfcgggatgt 1620 ggaσaccggg aaactggaga gcagactaca gggaccccat tgσgctgccg tcaaσgσcgt 1680 ggcctggtgc tactccggga gccacatggt gagcgtggac cagggcagga aggttgtgct 1740 ctggσagtag ggσcacgacc tgcctgcσtg ggctggagct σttgcccgaa gcσtgaagct 1800 tccttcggcg ccatgcaggg gttggggttg ggactggagc tggccttggg atttaatggg 1860 gaagaaggcc tggcaggacc tggcctgttt gtttaaaaat gaagtatggg ttgggggatt 1920 acgctagttt ttctttgtat ttttatctct atctatctcc tcactttttc tcσcaaagta 1980 gaaaaaaatg atatctgaaa aaaaaa 2006
<210> 26
<211> 3643
<212> DNA
<213> Homo sapiens
<220>
<221> misσ_feature
<223> Incyte ID No: 3594012CB1
<400> 26 gcggagaggt acttggatgc attttacagg σgcagtcatg caggaaaacc tcagatttgc SO ttcatcagga gatgatatta aaatatggga tgcttcatct atgacattgg tggataaatt 120 caacccacac acatcaccac atggaatcag ctcaatatgt tggagcagca ataataactt 180 tttagtaaca gcatcttcca gtggσgacaa aatagttgtc tcaagttgca aatgtaaacc 240 tgttccactt ttagagcttg ctgaagggca aaagcagaσa tgtgtσaatt taaattσtac 300 atctatgtat ttggtaagcg gaggcctaaa taacactgtt aatatttggg atttaaaatc 360 aaaaagagtt catcgatctc ttaaggatca taaagatσaa gtaacttgtg taacataσaa 420 ttggaatgat tgctacattg cttctggatc tcttagtggt gaaattattt tacacagtgt 480 aaccactaat ttatctagta ctccttttgg σσatggtagt aaccagtσtg ttcggcactt 540 gaagtactcc ttgtttaaga aatcactact gggcagtgtt tcggataatg gaatagtaac 600 tctcfcgggat gtaaatagtσ agagtcσata σcataacttt gaσagtgtac acaaagctcc 660 agcgtcaggc atσtgttttt ctcctgtcaa tgaattgctc tttgtaacσa taggcttgga 720 taaaagaatc atcctctatg acacttcaag taagaagcta gtgaaaaσtt tagtggctga 780 cactcctcta actgcggtag atttcatgcc tgatggagcc actttggcta ttggatcttc 840 ccgggggaaa atatatcaat atgatttaag aatgttgaaa tcaccagtta agaccatcag 900 tgσtcaσaag aσatctgtgc agtgtatagc atttσagtac tcσactgttc ttactaagtc 960 aagtttaaat aaaggctgtt caaataagcc cacaacagtg aacaaacgaa gtgttaatgt 1020 gaatgctgct agtggaggag ttcagaattc cggaattgtc agagaagσaσ σtgccaσgtσ 1080 cattgccaca gttctaccac aacctatgac atcagctatg gggaaaggaa cagttgctgt 1140 tcaagaaaaa gσaggtttgσ σtσgaagcat aaacacagac actttatσta aggaaaσaga 1200 cagtggaaaa aatcaggatt tctccagctt tgatgatact gggaaaagta gtttaggtga 1260 σatgttctca cctatσagag atgatgσtgt agttaaσaag ggaagtgatg agtccatagg 1320 caaaggagat ggctttgact ttctaccgca gttgaactca gtgtttcctc caagaaaaaa 1380 tccagtaact tcaagtactt cagtattgca ttctagtσct cttaatgttt ttatgggatc 1440 tccagggaaa gaggaaaatg aaaaccgtga tctaacagct gagtctaaga aaatatatat 1500 gggaaaacag gaatctaaag aσtccttcaa aσagttagca aagttggtca catσtggtgc 1560 tgaaagtgga aatctaaata cctctccatc atctaaccaa acaagaaatt ctgagaaatt 1620 tgaaaagσσa gagaatgaaa ttgaagccca gttgatatgt gaacσcccaa tcaatggatc 1680 σtcaactcca aatccaaaga tagcatcttc tgtcactgct ggagttgcca gttcactctc 1740 agaaaaaata gccgacagca ttggaaataa ccggcaaaat gcaccattga cttσσattσa 1800 aattcgtttt attcagaaca tgatacagga aacgttggat gactttagag aagcatgσca 1860 tagggacatt gtgaatttgc aagtggagat gattaaaσag tttcatatgσ aactgaatga 1920 aatgcattct ttgctggaaa gatactcagt gaatgaaggt ttagtggctg aaattgaaag 1980 actaσgagaa gaaaacaaaa gattacgggc ccacttttga aatttcagtg aataσcttaa 2040 tgttctgtaa tttgggaagt ttctggcaac acagaactac atagaatcag tattgttfctc 2100 atggcctcσa gggaaaaaat gtttttσaag taagagtaaa agggtgatgg gattttataσ 2160 caacaactgt ttcatcttaa aaatatgtat atttttatat taaaaattgt acagtatgtc 2220 atctaσccaa taggaaagtσ aaσaggatσt ttattttttg aaagctttag ccatσcacta 2280 agtgcccttt ttcataagag aagaaaattg tgcataaaaa ttggttatgt ttgtttttta 2340 gtcatctttt ttaaσatata tttttgattg aσaaattgcc tttcaaattt ttggggσtag 2400 ttgagattta aagagtttga tatgσcttct atttttatgg agaaagtaat tttaaaatgg 2460 caattggtgfc ttctaagcσa ttgaσtaata aaaσataggg ttggctagta attattttgt 2520 fcaacttgatg aagtcaagta tgactattat ttattgtaca tttgataaga caatttttgg 2580 aattttgaat tgcaσaaatt acatgatatc ttttgcattt atgttaσtat attgtaσttσ 2640 tgacaaatct ttattσσtgg gtggtatttt taagatatct ttaσσtataa aaaatgttta 2700 aggttσatag gactcgacaa gagctatctg gtgattttct cattagtaac atgcaaσgtt 2760 gtactgcaaa atttcaatca acatgacaac ttataatgag tggagatttc atattaggta 2820 ctaaatatta tagtattatt tctattttct ttttccaaat aagaagσttg gattatttta 2880 ttttgtggtc tttatcatta actttaattc tttσtgtact gtgtataata tttttatatt 2940 attggcctta ccataaaatt atttagaaag gttgtcaaaa taagttatac ctctttggca 3000 atagatagat gtatacatct acctactatg atctacaatt ttaggttaag tgaagcttgg 3060 gggggctact gacttggtta cσttcttgtc tcttgtccca aagatttaaa σtatgtacct 3120 ttgtatagct cttctgcσcc attttgactt ctgagatgaa agtatttact aaaattaaaa 3180 aaaaaaaaac aaaaaacaaa cctttagctc actaacttta tgggtttctg aagtgatgga 3240 aatttttaag gatatattta ataagcataa acttactaat aattacttcc aaaaataaaa 3300 acaggaatat tacttttaσc cagtgtggtt tatagcatac atttgtactg aagcatatag 3360 ggatgttaat gtgatctttt σctgacagat tatgaaagca ttatgacttg taacaagttt 3420 σcttgtatat caσtaacagg tttagaagaσ ataaatatta gtgtgttttg σctacatggt 3480 gtatttaaat ctattaatat tttσctgttg cttttttaaa aaaataaata caσataatgt 3540 atattaaaag aggtgggatg aaataatttt agtaattatg tgtaσagatg aaacattttt 3600 gtσatggaat ttaaaagcta agtaagtata aaaaataaaa tgt 3643
<210> 27
<211> 2694
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 7482435CB1
<400> 27 atggcggaga tccccσtgta σtttgtggac ttgcaggatg acttagacga σtatggattt 60 gaagactatg gtacagattg cgacaacatg agagtaacgg ccttcttgga cattcσaggc 120 caggataacσ tgcctcσact cactcgσσtg gagaagtatg σtttσagσga gaaσaccttc 180 aaccggcaga ttattgccag agggctgctt gatatcttcc gggacttcgg taacaatgaa 240 gaagaσttcc tcacggtaat ggagattgta gtcagattgt cagaagatgc agagcccaca 300 gtgcggactg agctgatgga acagattcct σσtattgcca tttttttaca agaaaacaga 360 tcaaattttσ cagtggtgct ctctgaatat ctcataccta ttgtaatgag gtacctcaca 420 gatccaaaca atcagataat σtgcaaaatg gcttcaatgσ tgagtaagag tacσgttgaa 480 σgcctgctac tcccccgatt ctgtgaactg tgtggtgaca ggaagctctt tcaagttcgg 540 aaggtttgtg ctgcaaattt tggtgatatt tgtσatgctg ttggaσaaga agccactgaa 600 aaatttttga tcccaaagtt ctttgagctc tgctctgatg ctgtttgggg catgcggaaa 660 gcσtgtgσag aatgcttcac ggctgtgtcg σaσagctcct cccctggggt ccgcagaaσσ 720 cagctcttcc cgctcttcat σagacttgtc agcgacccct gccgatgggt gcaccaggct 780 gccttccagt cσcttggσcc σttcatttσc acσtttgσaa accσctccag ggctggσσtt 840 tatcttcgag aggatggcgc cctgagcatc tggccactca cccaggattt ggactctggt 900 tttgcσtctg gσtcccσtgσ tccσagcagt ggtggtaaca tttccσctgσ σagtttaatt 960 cgttcagcaa agcctgtgcg gagcgagcca gagctacctg tggaagggac ctcagcgaaa 1020 accagtgatt gσσcaσaσag σagtagσtσσ tσtgaσggσc cagσggaaag cσσtgtggaa 1080 agctgtgtgt cggctggagc tgagtggacc agggtttccc cagagaccag cgcccgctca 1140 aagσtttctg aσatgaatga cctσσσcatσ agcagctacc ctggatcaga ttcctgggσσ 1200 tgcccaggga acactgagga tgtgttcagt cattttcttt attgcaagga cttagagctg 1260 cttctgagtg aggctgggcσ tσaggaggaσ gactgσagσa gacσtggggt tgtgσacaac 1320 agctgtgtgg cccggagtga gattcagaaa gtccttgata gcttgcagga gcatctgatg 1380 aatgatccag atgttcaagc tσaagttcag gtattatccg σtgcaσtgag agctgcacag 1440 ctcgactgcg tgaatgaagc tgagagcaag ccaacagcag gcctaaagga agtgtccatt 1500 tcacatσcca gctσtgcσtc tgaσaatcag atσgσtσtgg σggσσtcatσ atctcaggat 1560 gagctctttg tggccaggat attacaaagc ccagatccag gtggacccag aaatggaacc 1620 agtgaσσatc tggagactga ccagaggcag gatσσσaσcσ caσttgaaga gaataaatσt 1680 aaattacagg atgtaatacc tcagccgctg ctagatcagt atgtgtccat gactgaccca 1740 gctcgagccc agactgtcga tactgacata gccaaaσact gtgcσtacag σσtσccaggg 1800 gtggcactga ccctgggcag gcaaaattgg cactgcctga aagatacata tgaaacactg 1860 gcttσtgatg tacagtggaa ggtaσggσga gccσtagcct tctccattca σgagctggct 1920 gtgattcttg gggatcagtt aacagcagct gacctggtgc ctatcttcaa tggattttta 1980 aaggatctgg atgaagtgσg aataggagtt cttagaσacσ tgtatgattt tctaaagttg 2040 cttcatgaag acaagaggag agactatctt tatcagcttc aagaatttgt agtgactgat 2100 aacagcagga attggaggtt tσgatatgaa ctagcagaaσ agctgatact gattctggaa 2160 ctctatagtc ccaatgatgt ttatgattac ctaatgcaca ttgccttaaa gttgtgtgca 2220 gatcaagttt ctgaagttσg gtggatctcσ ttσaaaσtag tcgtggcaat tσtgσagaag 2280 ttctattcca acagtgaaag tgcattgggg ttaaatttca tcaatgagct catcataagg 2340 ttccggcact gttctaagtg ggttggaagg caagσtttcg σtttσatttg tcaggσagtg 2400 gtgagcaagg agtgtgtccc cgtggaccag ttcatggagc acctgcttcc cagσctcctg 2460 agcctσgcat cagatσσtgt gcσσaaσgtg agggttσtgc tagσcaaggσ cσtaaggcag 2520 atgctgttgg aaaaggcgta ttttagaaat gctggtaacc ctcatcttga agtcattgaa 2580 gagaccatct tagσattgca gtcagaσσgg gaσσaagatg tttσσttttt tgcagcσσta 2640 gaaccaaagc ggcggaatat catagacact gctgtactag aaaaacagaa ttaa 2694
<210> 28
<211> 2349
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 3882333CB1
<400> 28 cgccgcttgg gacgcctctg cctttccctc cctcccttcc ccgacggctt ctggcggcca 60 agtggatgtg gcgggtgatσ gagσcaccσt gσσσaggggc gcccagcaσt ggttcagtga 120 aσagσatttt ggaσaggaca tttggtgcca ggtctgagta gccagtttgc tgaattattg 180 tcccagtcag ccaggattgt gagctgtttg ggaagtttσg tggaaacgcc caagtgccag 240 σaσaggtgga gggacacctg gaggccagtt tcaggaactt ttgccacaag tataaaagac 300 ttcagaagtg caaagatgct gaaacagata ctgtcggaga tgtacataga tcctgatcta 360 ctggcagagc tcagcgaaga acagaaacag atcctgttct tcaagatgag agaggaacag 420 atccgaσgat ggaaagaaag agaagσagct atggaaagaa aggagtcσct gccagtgaaa 480 cccagaccaa agaaagagaa tggcaaatcg gttcattgga aacttggagc tgataaggaa 540 gtctgggtat gggtgatggg cgaacaccat σtagataaac cctatgatgt gctctgtaat 600 gaaattattg ctgagagggc ccggctgaaa gcagaacagg aggcagaaga gcccagaaaa 660 actcaσtctg aagaattcac σaatagcttg aaaacaaaat caσagtacca tgatctgcag 720 gctccggata accagcagac taaagacatc tggaagaaag tggcagaaaa ggaggaactg 780 gagcaaggat ctaggccagc aσσaaccctg gaagaagaga aaatccgatc actctccagt 840 tcttcaagaa atattcaaca aatgttggca gattcaatca atcgtatgaa ggcatatgca 900 tttcaccaga agaaagaatc tatgaagaaa aaacaagatg gagaaataaa tσaaatagaa 960 ggagagagaa cgaagcagat ttgtaagagc tggaaagaag actcggaatg gcaggcatct 1020 ctgσgaaaat ccaaagcagσ tgatgagaag agacgctcct tggctaaaca agcaσgagaa 1080 gactacaaga ggttatccct cgcggcccag aaaggaagag gcggtgagag gctgcaaagc 1140 ccσttgσgtg tfcccgcagaa acσagaaaga σσtσσσσttc cacσcaagσσ tcagttσσta 1200 aactcagggg catatcctca aaaaσctctt agaaatcagg gagtggtgag gacactgtcc 1260 agctctgσσσ aagaggaσat catccggtgg tttaaagagg agcagctacc acttcgagcg 1320 ggσtaccaga aaacσtcaga caccatagcc ccctggttcc atggaattct cacactcaag 1380 aaagσaaatg aacttcttct gagcacaggc atgσccggσa gttttctσat ccgagtcagt 1440 gaaaggatca aaggctatgc cctgtcctat ctgtcggagg acggctgtaa acatttcctc 1500 atcgatgσσt σtgσagacgc ctacagcttc ctgggσgtgg accagctaσa gcatgcσacσ 1560 ttggcggatt tggtggaata tcacaaggag gaacccatσa cttccctggg gaaggagσtc 1620 cttctctatσ cctgtggtσa gcaggacσag ctgσctgact acctggagct gtttgagtga 1680 cagcctccat cagggtcatc ctacagcctc caagcgggct ttcccctgga caaatgccac 1740 tgcaaσattt atgtgtgaag σσaaaatσaσ σctgcagcag agcσaatact gatcaactga 1800 aagtaaagta tccatggagt cctcattgac acctcttttc tgcacaaata ctggaattca 1860 atgtcaagag aaaatgaσσt σtgctσaaaa gggagaagag tctσaatttc agcaagtacc 1920 tgtcatgaag ggtatgacct taatgatgta cataaaataa aacaaatgaa gaaatggaaa 1980 acttttagaa attaaggtgt acttgaaaac gaatatσtat σatatgaccc ctgσaσtccc 2040 tctgtatcat ctcaggaggt ttcaggggcc tgttgacatg aagtttcgaa gtttcatgtt 2100 ggctttggaa tggtagσaaa agcσtttσct ggctgagatg atgcttaaaa cacacctcac 2160 ttattgtaca tgttggaacc aggacatgag agacatagaa aaacagaagt catgaatgta 2220 aattgaatga gaggcttaaσ atgcatgaaa ataσagatgg aσσtgcagga aagtgagσaa 2280 acatcgctga gtttgttttc ttgttcggga gaatggggcc ggggctggcc tggcctccct 2340 ggatataσt 2349
<210> 29
<211> 1213
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 7482809CB1
<400> 29 aggcgaaaag ctgσggttaa ggagagtccg gtttaaccgt caσσgggaag σgσgσtcgtt 60 cgggatcgcc gagtgggctg agatagtgaa ttcctaagaa gaaaataatg gattgcatat 120 tagttgttct σtaagtggaσ tcaacagtgt gcaagcttgt tggaaaagcc aaaagaagat 180 ggcaactcct tatgtcccag ttcctatgcc cataggaaac tctgcttcca gttttacaac 240 aaacagaaat caaagaagtt σttσttttgg σagtgtσtσa aσaagσtcaa attcttctaa 300 gggccagtta gaagactcaa atatgggtaa ttttaaacag acaagtgttc ctgatcaaat 360 ggataatact tcatctgtσt gtagcagtcσ σσtσattagg aσtaaattta caggtacagc 420 ttcttccatt gagtattcta ctagaccaag agacactgaa gaacaaaatc cggaaaσagt 480 gaattgggaa gatagaccat ctacacσtaσ tatactgggt tatgaagtga tggaagaaag 540 agctaaattt actgtatata aaatactagt aaagaaaacc ccagaagaaa gctgggtagt 600 tttcagaaga tacactgact tctctaggct taatgaσaaa ttaaaagaga tgtttσcagg 660 ttttcgacfca gcacttcctc caaaacgctg gtttaaagat aattacaatg ctgacttttt 720 agaagaσaga caattaggat tacaagcgtt tcttcaaaat ttagtagctc acaaggacat 780 tgctaactgσ σttgcagtga gagaatttct ttgtttggat gatccaσσgg gtccatttga 840 tagcctagaa gaaagcaggg cattctgtga aactttagaa gagacaaaσt accgcttaca 900 gaaagaacta cttgaaaaac aaaaggagat ggaatcacta aagaaactgc tcagtgagaa 960 gσaacttcat atagacaσtt tagagaacag aatcagaaca ttgtctttag aaσctgaaga 1020 atσactggat gtgtcagaaa cagaaggtga acagatccta aaggtggagt cctctgcact 1080 tgaggttgat σaagatgtcc tggatgaaga atctagagσt gataataaaσ σatgσttaag 1140 ttttagtgaa cctgaaaatg ctgtatcaga gatagaagta gcagaagtgg catatgatgc 1200 tgaagaagac taa 1213
<210> 30
<211> 3465
<212> DNA
<213> Homo sapiens
<220>
<221> misσ_feature
<223> Incyte ID No: 1739178CB1
<400> 30 atggcaaaaa tcgtggtggt gacatgcagt gattcaagtt ttggaaaσtt ttggttagat 60 caatggcaaa aacgagcaag agagaaatct ttgtgccaat gctctgccaa gcaagagatc 120 cgcacgcagc tggtggagca gttcaaatgt ctggagcagσ aatσagagtσ gσgaσtgσag 180 ctgcttcaag acctccagga gtttttccgc cggaaagctg agattgagct cgagtactcc 240 cgcagcctgg agaagctggσ tgagcgcttσ tσσtσcaaaa tσcgcagσtσ σσgggagσaσ 300 cagttcaaga aggaccagta cctcctctcg cctgtgaact gttggtatct ggttctgcat 360 cagacccggc gggagagccg agaccatgcσ aσσσtσaatg aσatσttσat gaaσaatgtσ 420 atcgtccgcc tctcccagat cagtgaggat gtcatcagac tcttcaaaaa gagcaaggag 480 attggcσtgσ agatgcacga ggagctσctg aaggtgacσa atgagσtσta caσagtcatg 540 aaaacctacc acatgtacca tgcagagagc atcagtgcgg aaagcaagct gaaggaggct 600 gagaagcagg aggagaagca gttσaataag tσaggagaσσ tσagσatgaa σctgctσcgg 660 cacgaggacc ggccccagcg ccgcagσtct gtgaagaaga ttgagaagat gaaggagaag 720 aggcaggcca agtaσtσtga gaacaagctg aaatgcacaa aggcσσggaa tgaσtacttg 780 ctcaatctgg cagccaccaa cgcagctata agcaaatact acatccatga tgtctctgat 840 σtgatσgatt gctgtgattt gggσttccat gcσagσσtgg ccσgcacctt ccggaσσtat 900 ctctcagctg aatacaacct ggagacctct cgccacgaag ggctggatgt cattgagaat 960 gcagtggaca aσσtggattσ ccgaagtgac aagcacacag tcatggaσat gtgcaatcaa 1020 gtcttctgcc ctccactcaa gttcgagttc cagccccaca tgggggatga ggtctgσcag 1080 gtcagcgσtσ agσagcccgt cσagacagaa ctgctcatgc gttatcacca gσtgcagtσσ 1140 agactggcca ccctcaagat agagaatgag gaggttagga aaaccctgga tgccaccatg 1200 cagacattac aggaσatgct gactgtggag gaσtttgatg tσtccgatgσ σttσσaaσaσ 1260 agtcgatcga cagagtccgt caagtcggct gcctctgaga cctacatgag caagatσaac 1320 attgccaaga ggagagσσaa σcagσaggaa aσagaaatgt tttattttaσ aaaatttaaa 1380 gagtatgtga atggcagtaa cctcatcacc aagctgcagg ccaagcacga tttactcaag 1440 cagaccctgg gcgaagggga aagagcagaa tgcggcacca ccaggσσσσσ σtgtσttσcc 1500 cctaaaccac agaaaatgag gagacctagg cctctctcag tgtatagcca taaactcttt 1560 aacggσagta tggaagcatt σattaaggat tcaggacaag ctatacσgct tgtagtcgag 1620 agctgcatcc gttacatcaa tttatatgga ctccagcagc aggggatctt cagagtgcca 1680 ggatctcagg tggaagtcaa tgacatcaaa aattcctttg agagaggtga agacccσctt 1740 gtggacgatc aaaatgaacg agatatcaat tcagtcgctg gtgttttaaa actgtatttc 1800 cgaggaσtgg aaaacσσaσt σtttcσtaag gaaaggtttc aagatttgat atctactatt 1860 aaactggaga acccagccga gagggtgcac cagatccaac aaatcctcgt cacccttccc 1920 cgσgtggtca ttgtggtcat gagatacctc ttcgctttσc tcaaσcaσct ctccσagtat 1980 agcgacgaga acatgatgga tccctacaac ctggccatct gcttcgggcc taccctcatg 2040 cacatccctg atgggcagga σσctgtgtcc tgccaggσaσ acatcaatga agtcatcaaa 2100 aσcatcatca tσcatcatga agccatσttσ cccagccccc gggagσtaga gggacctgtg 2160 tatgaaaaat gcatggσtgg aggggaagaa tattgtgaσa gσσcacacag cgagcσaggg 2220 gσσatσgatg aagttgacca tgacaatggc actgagcctc ataccagcga tgaagaagtg 2280 gagcagatcg aggctattgσ σaagtttgac tacatggggc ggtccccgcg tgagσtatσσ 2340 ttσaagaagg gggcctcgct gctcctgtac caccgcgcσt cggaggactg gtgggagggc 2400 cggcacaacg gcgtggatgg actcatσσcc catcagtaca tagttgtaca ggacatggat 2460 gatgccttct cσgacagcct gagccagaag gctgacagcg aggccagcag tgggccattg 2520 ctggatgaca aggcctcttc caaaaacgaσ ctcσagtσcc ccacggagca catctcggat 2580 tacggctttg ggggggtgat gggccgagtg cggttacgat ctgatggagc agccatcccc 2640 agacgcσgaa gσgggggcga σacacacagc ccgccccggg gcσtgggccc cagσatagaσ 2700 acaccacccc gggctgσtgc ctgccccagc agcccccaca aaatccccct cacccggggg 2760 aggatcgaga gσcctgagaa gcggaggatg gσgaσgttcg ggagcgctgg σagσatcaac 2820 taccctgaca agaaggcgct ctccgaaggg cactcgatga ggtcgacctg cggttccacc 2880 aggcacagca gcctagggga ccacaagtσσ ctggaggcσg aggσcctggσ agaagacatσ 2940 gagaagacca tgagcacggc tctgcacgag ttgcgggaac tcgagaggca gaacacggtc 3000 aagσaggσgσ cagatgtggt gctggacacc ctggagcσσσ tgaagaaσσσ gσσaggσσσσ 3060 gtcagctcgg agcccgccag tccccttcac accatcgtca tccgcgaccc cgatgccgcc 3120 atgcgcσgσa gσagcagσtc ctcσaσσgag atgatgaσσa σσttcaagcc agccctgtcσ 3180 gcccgcctgg ctggcgccca gctccgcccg ccccccatgc ggcccgtgcg gccggtggtc 3240 cagcacσggt ccagcagcag cagcagctcg ggσgtgggσa gσσσggcσgt gaσgσσσaσc 3300 gagaagatgt tccccaacag ctcagcggac aagtcgggca ccatgtgacc tgcaggatgg 3360 gccgcgcσgσ cgtggcccgc tgtggσtσaσ σacggcccag ggtggccttg gtggcttσσa 3420 cgtgcttccc agtgaccctg gccttggagg gcagaggtcc gtctg 3465
<210> 31
<211> 2609
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 7473630CB1
<400> 31 gtcggcaaca ggctcagccc taccctcaga agcgccctaa aaaacctaca gtgtagaaga 60 gcaσaccaga cagtgtgtct gσaagaagga tggctcσtaa tctgaaaaaa ggtaσaagtt 120 cctgtccagg gctcaccaat caggaaactc attctgactc caagggtgaa ggagcagggc 180 cagatggcaa gatttatgat ggaaaagata aaaσgaσtσa tσtaσtaggt gcttttactg 240 gtgcatctat gcgcggactg acacttagta gtacttcaaa tcaactctgg ctagaattta 300 attcσgatac tgaagggaca gatgaaggct ttcaacttgt gtataσσaag agaatσatag 360 gtattgcaga ggaggtcaca gtactcacat tgacagaatc agaacaggag cgagaacatt 420 tgtctaggga ggatcaggtt ttgaaσtσtσ aσaσtgtgaa gatσctggca ttσcacaatt 480 tggatacaag atcagtgacc aaggccactt tgctggtagc acσatcattt atggatgcaa 540 tσσaggσtaσ aσtσtccacg gaagtagcct tctσaactga atgtggaggt cgttttaaag 600 gagaatcatc aggaagaatc ttatctcctg gctatccttt tccatatgac aataacctgc 660 gttgcatgtg gatgattgag gtagatσctg gaaatattgt cagcttgcag tttσttgσtt 720 ttgatacgga agcatcacat gatatactcc gagtctggga cggtccacca gaaaatgata 780 tgcttttaaa ggaaattagt ggatσtctta ttcctgaagg aattσatagc acσσtσaata 840 tagtaaccat ccagtttgac acggattttt atattagcaa atctggattt gcaattcagt 900 tttcaagttc tgttgccact gσgtgtcgtg aσccaggggt σcσcatgaat gggactcgaa 960 atggggatgg aagagaacct ggggacactg ttgtttttca atgtgaccca ggafcatgaac 1020 ttcaaggaga ggaaagaata aσσtgcattc aggtagaaaa tσggtaσttc tggcagcσca 1080 gcccaccagt ctgtatagca ccσtgtggag gcaatttaac aggatcttσa ggσtttattc 1140 tttcaccaaa σttσcctcat σσatatccgc atagcagaga ctgtgactgg actatσaσcg 1200 tcaatgcaga ctatgttatc tccttggcgt tcatcagttt tagcatagaa ccaaactatg 1260 acttcctσta tatσtatgat ggaσcagaca gtaatagcσσ aσtgattgga agttttσaag 1320 acagcaagtt accagagaga atagaaagca gctcaaatac aatgcatttg gcttttcgga 1380 gtgatggatc tgttagttac actggatttc atσtagaata caaagcaaaa ctgcgagagt 1440 cctgctttga tcσaggσaat ataatgaatg gcacσagact tggaatggat tataaattag 1500 ggtσaacagt σaσσtattaσ tgtgatgctg gttatgttct tcaaggttat tcaacactσa 1560 cctgtttcat gggagatgat ggaagacctg gatggaatag agcσttgcca agttgtσatg 1620 σgccctgtgg aagtcgttca aσaggttcag aaggcactgt tctatcacca aactatcσaa 1680 aaaattacag tgtgggacat aattgtgttt attctatagc agttccaaag gagttgtggt 1740 gttggσcagt tgtatttttc cagacatcac tccacgatgt tgttgaggtg tatgatgggc 1800 caactcagca atcttctctg ttatcttccc tctcaggatc ccattcagga gaatcacttc 1860 cactgagttσ aggtaatσag atcaσaattσ gatttacttc agttggacσa ataacagcta 1920 agggatttca ctttgtttac caagctgttc ctagaacaag ttctacacaa tgcagttctg 1980 tgcctgaacσ aagattcgga agaagaattg gcaatgaatt tgcagtσggt tcatcggttc 2040 tttttgattg taatccagga tatattctcc atggatccat agcaattagg tgtgaaacag 2100 tgccσaattc tttggcσσag tggaatgatt ccttaσσtac ttgtattgtg ccσtgtggtg 2160 gaattttaaσ taagcgcaaa gggactattt tgtcacctgg ataccctgag ccttatgaca 2220 acaatσtgaa ttgtgtgtgg aagatσacag tgccagaggg agctggcatt caagtgcaag 2280 ttgttagctt tgctacagaa cataattggg attctctgga cttttatgat gggggagaca 2340 acaatgctσσ aagacttgga agctattcag gaacaacaat acσσσatσtt ttgaatagta 2400 cgtctaataa tctgtatcta aattttcaat cagacatcag tgtttctgct gcaggatttc 2460 atcttgaata σacagcaatt ggtttggatt cctgtcσtga aσσaσaaact σσtagcagtg 2520 gaattaaaat tggagacaga tatatggttg gagatgtagt atcctttcag tgtgatcaag 2580 gatattctσt tcaggtaagt σtattttaa 2609
<210> 32
<211> 2580
<212> DNA
<213> Homo sapiens
<220>
<221> misσ_feafcure
<223> Incyte ID No : 1431520CB1
<400> 32 gagagagagσ acagcctggt gggttctggg gtctacggσc taggggσcgg ggaagtttgc 60 gccgccgcga ccagtgctgc gatcccgagc cgggctccag ccccgaggac caggggtcgg 120 gcgggcctgc σtacggaacσ σσgcgggσca gσagcagtcg tctσgσgtcc tcσtgcttgg 180 aaaagtgttt aagcttctaa aatgtcatct atcaagcacc tggtttatgc agttattcgt 240 ttcttaσggg aaσaaagtca gatggaσaσt tacaσσtcgg atgaaσaaga aagtttggaa 300 gttgcaattc agtgcttgga gacagttttt aagatcagcc cagaagatac acacctagca 360 gtttσaσagσ σtttgaσaga aatgtttacc agttσσttσt gtaagaatga σgttctgσcc 420 ctttcaaact cagtgcctga agatgtggga aaagctgacc aattaaaaga tgaaggcaat 480 aaccacatga aagaagaaaa ttatgctgσt gσagtggatt gttacacaca ggcaatagaa 540 ttggatccca ataatgcagt ttactattgc aacagggctg ctgctcagag caaattaggt 600 cactaσacag atgcgataaa ggattgtgaa aaagcaatag caattgattc aaagtacagc 660 aaggcctatg ggagaatggg gctggccctc actgccttga ataaatttga agaagcagtt 720 acaagttatc aaaaggcatt agatcttgaσ σctgaaaatg attcσtataa gtcaaatctg 780 aaaatagcag aacagaagtt aagagaggta tccagtccta caggaactgg actgagcttt 840 gacatggcta gσttgataaa taatccagcσ ttσattagta tggcggcaag tttaatgcag 900 aaccctcaag ttcaacagct aatgtcagga atgatgacaa atgccattgg gggacctgct 960 gctggagttg ggggcσtaaσ tgaσσtgtσa agσctcatσc aagcgggaca gcagtttgct 1020 cagcagatac agcaacaaaa tcctgaactt atagagcaac tgagaaatca catccggagc 1080 agatcattca gcagcagcgc tgaagagcat tσσtgattta aσcaggggct caagσσσaag 1140 atacaaatgg tttatggcta tgaatgaagt atttgttgta gatagtaccc cctccctcct 1200 tcaaaaaacc aaacaσcata tctgatgtgt gaaaataatg gaggaaaaσc tσttgtgtat 1260 attcttaaag aataaatctg tttagaσaat aggtttatca caaacaaact caaaacataa 1320 catgaσtcct tttgaaatga tcaσtagcca tgttacgtgc σaatatagca ctgggataag 1380 gttccatttt caaacttcta tcatatttgt atattagact taatagcaaa gcatatttat 1440
-tgaactctag tagggctatt tgttggtaag ctσtttagσt σσaatttσcc ttaagcaaag 1500 ttttttgaga agggaaatga cccaaaggaa aggtttccta tgctggttga gaagaagtgt 1560 acttgccaσt gagσagtatg tcaggaaaga aggaatatta tσtaggttta gσtttgataa 1620 gtgctattag taatgaatat ataacatggg aaccaatgtt atctttaatg ttgcttgttc 1680 tggtgaaσag agaatcttaa gagctgttag aaagtagcac ttgatgcaag ggatgttttg 1740 aaaagaaaaa attggtaatg cgaatgtata gaaagtaaag gtaggatgct caggttttct 1800- gcatagttσt taaσtaatct tgtσtgσagt ttggtattga taatattagσ atggccactt 1860 atgctaaata cacaataaga tacatttaga aatccttaat gtactggtta ggtcagtggt 1920 acaactgttt gacttaatta tcaσaatttc cccaatggta accttacσtt ggaaactatc 1980 aaatataaat atactttaac taagccagtg acctggtcct aagttaagga ttttctaaat 2040 ggtatttaat tgtgtgccσa taaacactga ttttttttaa aaagaaaact tagtaattat 2100 ttgatggata acaaaaagca tctatcacta acattgcttg ttcatgtaat ctgtgataat 2160 ttggcttggc atgσcagtct tataattaac ataatttaaa tttgatgtaa tttaaatata 2220 acaggagact taaaatattt tattataaat tagatgccat attagttagt ggagaaaagg 2280 ggcattaggt attaattgca tgggtctaac tacattgatg atgagagttt aattccaact 2340 agagtatatt ttttgtttat taataatgaa attggtgctg acatgtaatg aσttcaσaag 2400 ttttacgagg gaaaactgaa aaaggaagta atataaattt tgtggggttt ttttgagatg 2460 gagtcttgct cttttgccag gctggagtgg tgttgcaatg tcagctcact gcaacctcσa 2520 cctcctggat tcaagtgatt ctcgagcσtt aagσtccσaa gtagctggga ttacgggcgc 2580
<210> 33
<211> 2181
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1916304CB1
<400> 33 cggσggσggc gacggcgaga aagagcttgc cggggggcga gcaggacagg acgaagccgg 60 agtgtaggcg gcagaggatt cgctcccaga gcagctgcgg ccaggtcgga aagaggccgg 120 ggcggσtggg tatatgaatg acctaaaggt acaaataaag acggagagag aacagtgcca 180 actgggagca gggcaagaat gccaattcct σσtcccccgc caσσσσσaσσ tggtcctcσt 240 σcacctccca catttcatσa ggcaaacaca gagcagccca agctgagtag agatgagcag 300 cggggtcgag gcgccctctt acaggacatt tgσaaaggga σσaagσtgaa gaaggtgaσσ 360 aacattaatg atcggagtgc tcccatcctσ gagaagccga aaggaagcag tggtggctat 420 ggctctggag gagctgccct gcagcccaag ggaggtctct tccaaggagg agtgctgaag 480 cttcgacσtg tgggagccaa ggatggttca gagaacctag σtggtaagcc agccctgcaa 540 atccccagtt ctcgagctgc tgcσccaagg cσtσσagtat σtgccgcσag σgggσgtσct 600 caggatgata cagacagσag ccgggcσtσa ctcccagaac tgccccggat gcagagaccc 660 tctttaccgg acctctctcg gcctaatacc accagcagta cgggcatgaa gσaσagσtσσ 720 tσtgσσσσtc ccccaσcacc cσσagggσgg σgtgccaacg caccccccac acctctgcct 780 atgcacagca gcaaagcccc cgcσtacaac agagagaaac ccttgcσaσσ gaσgσctgga 840 caaaggcttσ aσcctggtcg agagggaσct cctgctcσac σcccagtcaa accacctcct 900 tcccctgtga atatcagaaσ aggaccaagt ggcσagtσtc tggctcctcc tσctσσgσσt 960 taccgccagc ctcctggggt cccσaatgga ccσtσtagcc ccactaatga gtcagcccct 1020 gagctgccac agagacacaa ttctttgcat aggaagacac cagggcσtgt cagaggccta 1080 gcacctcctc caσccacctc ggσσtcccca tctttactga gtaataggcc acctccccca 1140 gcccgagacc ctcccagtcg gggagcagct cctccaccσc cacσacσtgt gatσcgaaat 1200 ggtgccaggg atgσtσcccc tccσccacca ccataccgaa tgcatgggtc agaacccccg 1260 agccgaggaa agσccccacσ tccaccctca aggacgσcag ctgggccacc σσσtcctcct 1320 ccaccgccσc tgaggaatgg ccacagagat tctatcacca ctgtccggtc tttcttggat 1380 gattttgagt caaagtattc cttccatcca gtagaagact ttcσtgctcc agaagaatat 1440 aaacactttc agaggatata tccσagcaaa aσaaaccgag ctgcccgtgg agccccacct 1500 ctgccaccσa ttσtcaggtg aagσσtggct tggtcccgtt cctσaggaaa aggatggacc 1560 ttctcttctt σtσagatggt cccttccatt cccctgaaac ctgcatgaga gctcctaaca 1620 tgtttctcca atgcaatcaa gcσσtagaσt ccaaatgtcσ tccσagctσa σctσσatσta 1680 tgcatσtσat σtσtggattt ggtgatσaga ctctatattg acagtaggat ctcaaaccct 1740 gcatccatcc ttcctccagc aagccctgct agσcacatga ggaacaagtt tσcgtgtctt 1800 ctgccttcct σttggggaaa ggtgccttgt tgtgatgaat taactcactg ttagggcagg 1860 gtggagaatg gtactccttc cttctcctgt ccactgtggg ggaagcttgg caggtatatt 1920 atatttcatσ atttaggagg ctggcatgac caggaσttat gggtgggagg ggagcatttt 1980 tagtgaagca agaaaggagt ttgccaagaa gtgatctgtt ttaaaggtca tatttggaga 2040 aagggcaagg aattgggtct gctttatttt tgggggtatt ttgtttttgt tctcacctgc 2100 tgccccccca ccσcaccacc ccagggataa attggatata aacactaaat actaatcagt 2160 tgaacttaaσ atttaataaa a 2181
<210> 34
<211> 4149
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 378504CB1
<400> 34 atttgacagt σccgσσccag cσgσσgtgtg atttagσaaσ cggtatcata ttcagatcaa 60 atcttaaaaa aaaaaagfcca cctacagaac tacagaatgg ttctctcccg ctcaagaagg 120 gσaagtggac ttgggcgtta agatgcaggg gggtgaacca gtgtcσaσaa tgaaagtctc 180 ggagagcgaa ggaaagctgg agggccaggc cacagcggtg accccgaaca agaacagcag 240 ctgtggaggt ggaatcagta gσagcagcag cagccgcggt ggcagtgcaa aaggctggσa 300 gtacagtgat cacatggaaa atgtgtatgg ctatttaatg aagtatacca accttgtcac 360 tgggtggσag tacaggtttt ttgttttaaa caatgaagσt gggctgttgg agtactttgt 420 gaatgaacag tctagaaatc agaaacctag aggaactttg cagcttgcag gagctgtaat 480 atcacccagt gatgaggatt ctσacacctt caσtgtaaac gctgσσagtg gggaacaata 540 taaactcaga gctacagatg caaaagagcg acagcactgg gttagcagac ttcagatatg 600 tacaσagσat catactgaag ctattggaaa gaataatcct cctctgaagt cacggagctt 660 ctcacttgca tctagtagta attctcctat atcgcagagg agaccaagtc aaaatgccat 720 ttcttttttt aatgttggac attccaaact gcaatcactg agcaaaagaa ctaatttacc 780 tccagaccat cttgtggaag tcagagaaat gatgtctcat gctgaaggac aacaaagaga 840 cttaattaga cgaattgaat gccttcctac ttctggσcat cttagttσσt tggaccagga 900 tctcttaatg ctcaaagcta cttccatggσ aactatgaaσ tgcttaaatg actgctttca 960 tattctccag ttacagcatg catcacatca gaagggctca ttgccttcag gaacgacaat 1020 cgagtggtta gaaccaaaga tatctttatc aaaccactat aaaaatggag ctgaccagcc 1080 ctttgσaaσt gatσagagta agccggtggc agtcccagaa gagcagcctg ttgcagaatc 1140 tggactatta gcgagggagσ ctgaagaaat aaatgcagat gatgagatag aggatacatg 1200 tgaccacaaa gaggatgacc tgggagctgt agaagaaσaa σgtagtgtca tcctacatσt 1260 cttgtcacag cttaagctgg gcatggattt aacaagagtg gtgσttccta cafcttafccct 1320 agagaagcgt tσσttgctgg aaatgtatgσ agaσtttatg tctcatσσag acctatttat 1380 agccatcact aatggagcca cagctgagga cagaatgatfc cgcttttttg agtactacct 1440 tacctcattt catgaaggcc gtaagggagc σattgctaaa aaaccataca atcctatσat 1500 tggagaaaca tttcactgtt cctggaagat gccaaaaagc gaggtagcat ccagtgtttt 1560 tagcagttσt tcσaccσagg gagtcacaaa tcatgσtσσt ttatcggggg agtctttgac 1620 ccaggtggga tcagactgtt acacagtcag atttgttgct gagcaggttt ctcatcatcc 1680 tcσagtctca ggattttatg cagaatgtac agagaggaag atgtgtgtaa atgcgcatgt 1740 ctggactaag agcaagttct taggcatgtc aataggcgtg acaatggttg gagaaggtat 1800 ccttagtctg ttggagcatg gagaagagta cacattttσt ctaccctgtg catatgctcg 1860 gtcaattttg actgttcσtt gggtagaact gggtggcaaa gtcagtgtca actgtgcaaa 1920 aactggatat tσagcσagca tcaσttttca taccaagσσa ttttatggtg gcaaactgca 1980 tcgggttaσa gctgaagtaa agcaσaaσat caccaacact gtggtatgca gagtgcaagg 2040 ggaatggaat agtgttcttg agttσaσata tagσaatgga gagaσaaagt atgtggaσtt 2100 gactaaattg gcagtgacga agaaaagagt gagacctctg gagaagcagg atccatttga 2160 atσcaggcga ttgtggaaaa atgtgacaga ctσgctgaga gaatctgaaa ttgataaggc 2220 cacagagcat aagcataccc tggaagaacg tcagaggact gaagaaaggc atσgtactga 2280 aacaggcaca cσttggaaaa ccaaatattt tattaaagag ggagatggct gggtttatca 2340 taaaccaσtt tggaaaataa ttccaacaac acaaccagca gagtgacaca tactatctaa 2400 aactσgaσσa aatgaggttc ttctctgttt aσσctaaatc ctσσσagaat ggagtcattg 2460 cactgagtga cctgcttcct gattgcgcag actgaaacta gctaaacctg aatgtaccta 2520 ctagggcaσc ataatactgc agcaagacca aagtggtaaa gaaacaσagt ggacctttta 2580 ccaacctgtt catgtgatgt gagcaatacc atcttaaaac ttgttacctg aatcagtaga 2640 tgaatctttt atcσagttσt tgσtσσtaaa gttaagtttg aatσccctat ttttgσaσag 2700 gggcagcaga tacacacaaσ aatgagaact cagtgacttt gatttctttg tagtgaaaag 2760 tgaagtctσσ gtttσagagt ttgtgtσttt σtttσtgtcc ataactgaag tattcaσtaσ 2820 tcttgtaaac caaccaagag gaggagaaag atgacccaga agtggattca gccattgtgc 2880 ctgaaatcag tgtttaaaaa aaaaaatcaa cσaggttgtg gtaaσaaggσ attσtatttc 2940 ttcaaaaaga ctgtatgcct gtgtctgagg aacttaccta ttatccacct σtgttggaac 3000 tctcttttaa aaagtacatt tatagattga tcagaattat aaσcatggag aattttttct 3060 tctgagcatt ttaatatact tgaaaacaac attgacttga aaaatttcag aacatttttc 3120 agtacctagt tttattaaat attacaσttg agagacaσtt tttaaaaatg tgttaatgtc 3180 aatatgatga gattttagcc tttctccaga actaaggcat taaagaaaat agcaaatatt 3240 aaaaaataaa actgttactt ttttccttct ttσttttcac ctttaggtta atatccagta 3300 ttatgtgtta tccctttgga taagtatgct ttattttacc tctgttaaaa attaaaataa 3360 atgattσtat tcatatttgt cagtaattca aaacttatat gtgtaactga acgcgcatgt 3420 aaggtatggt tttatttatt tttttttttt ttgaggaaat ttaaatgcta aagaaacaac 3480 gaaatgaaaa ggtatcagga aaaaaagatc aggaagttgt attcaggtac aaatσttttt 3540 ttaaataagt attttgttga ggttgaagaa ttgctggcaa ttaaaagaat agagctaatt 3600 atggctttσa tσattσattσ atgtatttat tgagcaccta cttattatgg tgctσaaσaσ 3660 ttgttactgc aagctaσctt aatttcccaa gagtggtgcc ttactctgtt ttttctgata 3720 tggfccttcca atcagtgtgt gtaaσatacσ tgttgtttat cagσσattgt aggtggσtgt 3780 gtctgttgca tcatcataag aagtttaagc tttgtgctct gataaattgt gttctgttaa 3840 agaggttagt aggatgaaaa cagσaaaaσa ataatttttt σaaσaaattg taaattataa 3900 gaaaaagagt tggtttgtgt acaacaattt taatgattcc cttgttcatt tttgctgtga 3960 aatgcactga aaaaaatcct caaaatgagt tatagttcσt gtgttgggaa aattgaσaaa 4020 taataaaact agagaacaac catataacaa aaaacaaaaa acaacaaaac aaaaaaaaca 4080 acacaacaca aacaaaaaca aaaaaacaaa caacσacaaa caaaaaaaaa σagagσσσσσ 4140 catagacca 4149
<210> 35
<211> 3080
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 5275371CB1
<400> 35 gctagaaatg gggaaaacca attatggtta catagaaatt aggagcatgt aaaagtacat 60 tattaaacca cttcatgtat tttttctctt σaatcatttg gggagaattσ ttσttggttt 120 aagaacttat tatccacagc acagcatatg tatgactttg tcccttattt taaattctga 180 aattctggga atttgtcccσ gagtσσtggg aactctgtct tccσaaσcat taaactaσag 240 cagaaaataa atccaagggg caatttgtat tgtgaccata gtagaatgcc tgtgtctttt 300 ctttaaaggc tgttgaaatt tatgaaaaat gtgσctttat tttσtσagac ctgagσσatt 360 catgtactgg gtatgggtgg ttgggtgggt ccaggagtgt gcaactccct ccattatccg 420 ccttgttcgg ggtttgσtgσ tctgaccσσt aσtttgσttt tcagatgact ttgggaaatt 480 gctgctggct gaggccctcc tggagcagtg tttgaaggag aaccatgcca aaataaaaga 540 ctccatgσct ttgctggaga agaatgagσc gaagatgagc gaagccaaaa attatctaag 600 cagtatcctt aaccatggga ggctctcgcc acagtacatg tgtgaggcca tgctgatcct 660 gggcaaactg cattacgtgg agggctcata ccgagatgcc atcagcatgt acgcaσgggσ 720 cgggattgat gacatgtcca tggagaacaa gcccctgtat cagatgcggc tgctgtcgga 780 ggcttttgtc atcaaaggσσ tσtσtctgga acgcctaccc aactccatcg cctcσσgσtt 840 ccgcctgaca gagagggagg aggaagtgat cacσtgtttt gagagggcct cctggatcgc 900 tcaggtgttc ctgσaggaat tggagaagac caσaaataac agcacgtcga ggσatctgaa 960 aggctgtcac ccgcttgact atgagctcac ctacttσσtg gaagσtgσσσ tccagagcgc 1020 ctatgtgaaa aaσctgaaga aggggaacat σgtgaagggσ atgagagagσ tccgggaagt 1080 gctgcggact gtggagacca aagcaactca gaacttcaaa gtgatggcgg ccaagcacct 1140 ggσgggggtσ σtgσtgσaσt σcctgagtga ggagtgctaσ tggagcσσcσ tgtcccacσσ 1200 tσtgcctgag ttcatgggca aggaggagag ttctttcgcc actcaggccc tgcggaaacc 1260 tσaσctσtat gaaggagaσa aσσtσtaσtg σσσcaaggaσ aacatσgagg aagσσσtcσt 1320 gσtcctcctc atcagcgaat ccatggcaac tcgagatgtg gtgctgagcc gggtgccgga 1380 gcaggaggag gaccggacag tgagσttgσa gaatgccgca gccatctatg acσtσσtgag 1440 catcacgttg ggcagaaggg gacagtacgt catgctctcg gagtgcctgg agcgagccat 1500 gaagtttgcg tttggagaat ttcaσctttg gtacσaggtg gσσσtσtcca tggtggcttg 1560 tgggaagtca gcctacgctg tgtccctgct gcgggagtgt gtgaagttgc ggccctcgga 1620 cccσaσσgtg cσσσtgatgg ccgσgaaggt σtgcatcggg tcσcttcgct ggctagagga 1680 agcagagcac tttgccatga tggtgatcag cctcggagag gaagccgggg agttcctccc 1740 caagggctaσ ctggσtσtgg gtσtcaσσta tagcctgcag gccacσgaσg σcacσctgaa 1800 gtccaagcaa gatgaattgc accggaaggc actgcagacg ctggagaggg ctcagcagct 1860 ggcgσσσagt gacccσσagg tσatcσtσta tgtσtcgctg cagctggσσc tcgtccgaca 1920- gatctccagt gccatggagc agctgcagga ggccctgaag gtacgcaagg atgatgccca 1980 cgccctσcac ctgctggcaσ tgctcttctc tgcσσagaag σaσσacσagσ atgσσctgga 2040 tgttgtcaac atggccatca ccgagcaccc tgagaacttσ aacctgatgt tcaccaaggt 2100 gaagctggag caggtgctga aaggcccaga ggaagccctc gtgacctgca gacaagtgct 2160 gaggctgtgg cagaccctgt acagcttctc ccagctggga ggcctagaaa aggatggcag 2220 cttσggtgag ggcctcacca tgaagaagσa gagtggcatg σacctgaσtt tgσσtgatgσ 2280 ccatgatgca gactctggct cccggcgggc ttcgtccatc gccgcctccc ggctggagga 2340 ggcσatgtσa gagσtgacta tgccσtcttσ ggtcσtgaag σagggccσca tgcagσtgtg 2400 gaccacgctg gaacagatct ggctgcaggc tgctgagctg ttcatggagc agcagcacct 2460 caaggaagca ggtttctgσa tσσaggaggc ggcgggcσtc ttccσσaσtt σtcactcagt 2520 actctatatg cggggccggc tggctgaggt gaagggcaac ctggaggagg ccaagcagct 2580 gtaσaaggag gcgctcaσgg tgaaσccaga tggσgtgcgc atcatgcata gcσtgggtσt 2640 gatgctgagt cggctgggcc acaagagctt ggcccagaag gtgcttcgtg atgccgtgga 2700 gaggcagagt acgtgccacg aggcgtggca gggσσtgggc gaggtgctgc aggcσσaggg 2760 ccagaacgag gctgccgttg actgcttcct caccgccctt gagctggagg ccagcagccc 2820 tgtactgσσσ ttσtσcatca tcccσagaga gctσtgaσga cgctgcagcc gcagggaggg 2880 aggggctggc cagagggaga ggcagcaggg aacgtgggtc agggtggggc aacagtggca 2940 tcaggtgcgg ggσctcaggg aaataσatct ttagtgaacg cctctgcagc tgcagccσtσ 3000 gttctcttgg ctgggccaag aggccttcct ggatttcttt gttgtcaatt gggaacaggc 3060 tgσtgagσaσ σgaσtgσcag 3080
<210> 36
<211> 4167
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 490576CB1
<400> 36 ggσggσggga gggagtgcgc tgσgcσtgcc tσσgggagga gccgcatcca caσaσσσtgσ 60 gctgccctgt cctgcgcgag tggagctctg aagaagctct gagcggagtt gtgttcttcc 120 ccaggtgcgt σσtggctgag agttggagct ctσσagσaaσ atgσctgagc agagtaacga 180 ttaccgggtg gccgtgtttg gggctggcgg tgttggcaag agctccctgg tgttgaggtt 240 tgtgaaaggc aσattσcggg agagσtacat cσσgacggtg gaagacaσct accggσaagt 300 gatcagctgt gacaagagca tatgcacatt gcagatcacc gacacgacgg ggagccacca 360 gttcccggcc atgcagcggc tgtcσatctσ σaaagggσac gσcttcatcc tggtgtactσ 420 cattaccagc cgacagtcct tggaggagct caagσccatc tacgaacaaa tctgcgagat 480 caaaggggac gtggagagca tσcccatσat gσtggtgggg aacaagtgtg atgagagσσσ 540 cagccgcgag gtgcagagca gcgaggcgga ggccttggcc cgcacatgga agtgtgσσtt 600 catggagaσc tcagccaagσ tσaaσcataa cgtgaaggag σttttσσagg agσtgctcaa 660 cctggagaag cgcaggaccg tgagtctcca gatcgacggg aaaaagagca agcagcagaa 720 aaggaaagag aagctcaaag gcaagtgσgt gatcatgtga aggcσcttcc tgcgggagga 780 gcagctgtgt gtccccggca cctcactccc ccaaaatgac acccaccgtc gtcagggtag 840 catgtataat gσσcaσgtgt taaaσattgc atttaatcga gatgcgtcct attgtcctta 900 agagggcgtt tcacaccacc aacagtaagc cacccactct ggagtcacag aatctgccag 960 gσggttσaag tgaaaaccaa σaσaσtcagc atσσσtggga aσtgagaggt gccagcaatt 1020 gctgaaggtg gcgatgaaca cccgaaggtg ggagggagga ctggtaccca caaagcaaca 1080 tgtaccgaga ggactaaatg tcatσtaσgt gσatgtgaga gσgtgttaaσ ctagagttaσ 1140 ctgcaccaac cccagacaga agccaatcac atctttgggg gaggggaggg gcaggaagag 1200 gtgagaagat cagatggtcc aaagtggacc aσacttggtσ σattttacac ttttttaaag 1260 gggattaaaa aacacagcct ctcccccaaa gggtgtccgt tcttaattcc cacctggcct 1320 gttaggagσc ttgσtaσcσt gaggggatgt gttσacctta cctagacσta gttaggaagt 1380 atcattttaa gctattagag tatttatctt catgtgcagg gataagtgca ctaacagtgt 1440 gctgctctgt cggaagttct tcagttttta agtgaggata tcgtgacagt attaaaacat 1500 cgcaataatg ttcctgtgtg ttatacatcg agggttttag aaatgtgatt ttcttctttt 1560 gacctgtgag gagtataact tctttσagσc ctcagatttt aaatacaagc aaataaactc 1620 actattttta gacgtttttt tcctccaagg tggttttctt ctcttaaata actcgatctg 1680 tacccagctg ggtagcagσσ agσaaaggcc atcagacaac cagaagcaσa tccatttttg 1740 tagtgtcaσa aacatgtata tgccacactt tgcaccttaa tgaaatactt tgaaacagaa 1800 gttattcact gtgtttttga tgatσtatct gtattggaaa tatgttcσtg gaaaatgcat 1860 ttaaataata gtaaattctc ttgcatgttc cattatacgt gtcttσtaag agctgttcaa 1920 taσagtattσ aσtctagaaa σaattatσtt tttσtcttaa tgattttgtg tgcatcttta 1980 atctttcaag ccaaattaca gctatttcag gtttcctgtg ttagcttggg gataggatgg 2040 tggctggaga σaggσaggσt tctctgσcct gggaagagσσ σactcagctt aattgctσtg 2100 ccatcgtaga gcctggttgg acttggcttc ctgaaaactc ccactgatag tgcctgttag 2160 atctcctgtt tgtttσagtt ggcagaacat ttactggccc caaσtgtggc atcatcσtct 2220 cagcagtctt cctgtcaccc gcctggcagg cagaaggagc tgcagtccca cgtgggcctg 2280 cσtggggggg tgggggctgc atggctgttg ggtggcagtg tcagcacagg gagggcttaa 2340 gttggggatg tttgaccagg ccacσtcctg caactgctgt ttctcctgtc cctcctatgc 2400 agggσttgca gσagσagcag tgtggσcatc tcσatσσσσc aaagcacact tgσtctctca 2460 atatgtccta gttttcttca gccttttctg gttcagttcc cttgtcctga tctcatcctc 2520 tctggtσtcc caataaσtca σccttgggat gtgtttagag cgtgggaggt gcσtttgaga 2580 actgcttgac tccatgatct σctagaacaa aaccgccctg actttacagg gggaacactc 2640 atgctgagct gagaaagcag agaagtggcg tgggagccag ctgggggtga agagcatttg 2700 ggccagtccc gtggccccct tcagattσσt caagcaggat tgttctgttc taaaaagctg 2760 ttgcacagca ttcgσaatga gatσtttagt tggσggattt tctggaacat ttgtttttca 2820 acttgtcccg acattttttt ttctgtttct attctgagag agagatgatc aagttttaat 2880 ttgggtatag gttaaatgga agaagaaaca gaacttσatg gccaaagtag acσtatagat 2940 tttgattggg ttσtttgtta acagtagaat gcgatctttg ccactgactg tagtattaat 3000 aaggttttaa tgtgagatat tcctgcaaac catcccattt σtactgattg taagtcagaa 3060 tttcttttat ccctttcaaa tcagtttcta catgtttaag tgttcagggc ttcatcagca 3120 tgagaagttt gtaattactg aaagtctgat ttcattcagg acacattttt ttccttcata 3180 ttttttctgt gaatttatag gctaggaagg ctattgaagc ctcaattatg ggtcttcatt 3240 ttgagatcgt tttctatgag ctgaaσtgag gatatcaatg gttatctcaa aatσgtσttt 3300 taggagatcc ccaattgact σagagtttga ggagttagta tcacagaatt agattttttt 3360 aaagcatttg taσgtttcca ttcccaaata tgtagctgtg gttcttgaaa acacatccta 3420 cattgcatat gggcatagca gtttttgacc caggcagaat aagttaatat ttaattaaat 3480 attgctttga agatggcgct ctgggcatga gcatggggσt σσatgacttc ccttctatcc 3540 ccatgagccc ctcctccatc cagcgacaag ccatgggcat gcatacaatg cagcaagacc 3600 aacacaagag caatattgaa ttgttcattc tatctaaaat tacatgtata taaaatatat 3660 aatttatctt cctgcatttt tgaagtataa agtcataaat tgtaσatatc tgtaagctag 3720 tatatttgtt tcactgtttg taatatttaa gaaatgctca ttctttgtag aacaaaaatg 3780 tattaaatat tttaaaaatt gctctgtgat acttaatttt tttcccσaaa atttgtaatg 3840 tgttgcttct acataagttc tctggaaata tctaσaaσta ataggacaca tgtaaatcct 3900 tgaagacaca tcσtggaatt cataccccac aaggacagtg tgtataσaaa gtatttgcag 3960 agcatgactt ttatatgtgt gggatatσaa tgtgtatatt tatatttaaa gtgtatttat 4020 tgttacaagt ctattσtcta ttatatttta tttactctgc ggttataaaa atcacccttg 4080
- catacaagtt tctagttgσc agtgatgttc tggaaataat gggagatatt acaataaagc 4140 tacagttatg aσaaaaaaaa aaaaaaa 4167 <210> 37
<211> 3591
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No : 1417657CB1
<400> 37 gggcccctcc tggcctctgσ σaaσgσagct cσσtgacccg gaactttcgt ggtgtcgggc 60 gagagtttgt gcσcacttcc tgagagggat gggtaagaag aggaccactt ccggcgaggg 120 acgggaaaga cagcggcttc cggσgcggcg gttccggaσa aσcgtgcgct tttagtaaaa 180 gattggggtt cgσgcggggg agaagggctg ccccgggccc tctggttctc gtcccgcagc 240 gtccgctcσσ σcgcgccact gcgccgσtσc caggaaccct gtactccggg gtcgecggct 300 tctctcctgc ctccggtccc gccagacacc tcgagctcct taagtagctc ggtccttgac 360 gtcσσtσtgg gσσcttcccg cgtctatσgσ σtgagtcσcc gggcccctct agcσctctgt 420 tccctcccct cttttgttcc tccctagagc cccgccgccc tcagggctga cagtgtggac 480 ggcgggagtc tcctcgσtcc cctgctggga ttgactgacc gagcgtttag tgactgcσσa 540 gatctggctg atgggggtac cgagaggtgg cctgggccgg gaatgtccag ctagagtctt 600 σcgtggaagt σagacatgaa aσtgacaggc ctaagggaag ctaggaagtc ccctσaccgc 660 tcagccaggg tgatgggctg gactgacaga ctccagtgaa tttgagcttg cctgtcaggc 720 tgattggctg atagaσagcc ctggattggc tcaσtaagac tgaccagσσσ gggaσσaagc 780 agttctgggg tcccaacctg ggtggaaggt ctgaactgat gacccaccca ggctgaccag 840 gσσagcccac σtcaσtgaσσ tcctgaσσσc tgaσσtσatc acσtgtgcag σcatggagaa 900 gatgtcccgt gtgaccacag ccctgggtgg cagcgtgctg acaggccgca ccatgcactg 960 ccacσtggat gσtσccgcca atgccatcag tgtgtgσσgc gacgσagccc aggtggtcgt 1020 ggcaggccgt agcatcttca agatctatgc catcgaggag gaacagttcg tggaaaagct 1080 gaacctgcgt gtggggcgca agccttcgct taacctgagc tgtgctgacg tggtσtggca 1140 ccagatggat gagaacctgc tggccacagc agccaccaat ggcgtggtgg tcacgtggaa 1200 cctgggccgg cσatcccgca aσaagσagga ccagσtgttc acagaacaca agσgcacggt 1260 aaacaaagtc tgcttccacc ccaccgaagc ccaσgtgctg ctcagtggct cccaggatgg 1320 cttcatgaag tgctttgacc tccgcagaaa ggaσtσtgtc agσaccttct σgggccagtσ 1380 ggagagcgtg cgggacgtgc agttcagtat ccgggactac ttcaccttcg cctccacctt 1440 tgagaacggc aatgtgcagc tctgggaσat ccggσgtσcc gacσggtgcg agaggatgtt 1500 cacagcccac aaσggacccg tcttctgctg cgactggcac cccgaggaca ggggctggtt 1560 ggccactgga gggcgcgaca agatggtgaa ggtctgggac atgaccacgc accgtgcσaa 1620 ggagatgcaσ tgtgtgσaga ccatcgcctc ggtggcccgt gtgaagtggc ggccagagtg 1680 cσgσσaσcac ctggσσaσgt gσtccatgat ggtggaccac aacatctatg tttgggaσgt 1740 gcgccggccc ttcgtgccag ctgccatgtt tgaggaacac cgagaσgfcca ccacgggaat 1800 tgcctggcgc σaσccccacg acccctcσtt σctgctgtct ggσtσσaagg aσagctcgct 1860 gtgccagcac ctgttccgcg acgccagcca gcccgtcgag cgcgccaacc ctgagggcct 1920 ctgσtaσggc ctcttσgggg acctggcctt cgσσgσcaag gagagcctcg tggctgcσga 1980 gtcggggcgc aagccctaca ctggcgaccg gcgσcacccc atcttcttta agcgcaagct 2040 ggaccctgσc gagcccttσg caggcctσgσ σtccagtgcc ctcagtgtct ttgagacgga 2100 gccaggtggc ggcggcatgc gctggtttgt ggacacagct gagcgttatg cgctggctgg 2160 ccggccactg gccgagσtct gtgaccacaa cgσaaaggtg gσtσgagagσ ttggσcgcaa 2220 ccaggtggcg caaacgtgga ccatgctgcg gatcatctac tgcagccctg gcctagtgcc 2280 cactgcaaac ctcaacσaca gtgtgggcaa gggtggctσσ tgtggcctσσ cgσtσatgaa 2340 σagtttcaac ctgaaggata tggccccagg gttgggσagt gagacgcggσ tggaccgcag 2400 caaaggagat gcacggagcg acacagttσt gctσgaσtσc tσggccacac tcatcaccaa 2460 tgaggataac gaggaaaccg agggcagcga cgtacσtgσc gactaσctgc tgggtgacgt 2520 ggaaggtgag gaggacgagσ tgtacctgct ggatσσggaa cacgcgcacc ccgaggaccc 2580 tgagtgcgtg ctgccgcagg aggcctttcc gctgcgcσaσ gagatσgtgg aσaσgσσtcc 2640 cgggcccgag cacctgcagg aσaaggσσga σtσcccgcac gtgagcggca gcgaggcgga 2700
- tgtggcσtcc ctggcccccg tggaσtcctc cttctcgσtc σtgtσtgtσt σacacgcgct 2760 ctacgaσagσ cgcctgccgc ccgacttctt cggσgtgctg gtgcgcgaca tgσtgcactt 2820 ctacgctgag cagggcgacg tgcagatggc tgtgtctgtg σtσatcgtcc tgggtgaacg 2880 ggtgcgcaag gacatcgacg agcagaccca ggagcactgg tacacttcct acatcgacct 2940 gctgcagcgσ ttσσgσσtσt ggaaσgtgtσ caaσgaggtg gtσaagσtga gcacσagσσg 3000 σgccgtcagc tgcctcaacc aggcσtccac caccctgcac gtcaactgca gccactgcaa 3060 gcggcccatg agσagσσggg gσtgggtσtg σgaσaggtgσ σaσσgσtgcg ccagcatgtg 3120 tgccgtctgc caccacgtag tcaagggtct cttcgtgtgg tgccagggct gcagccacgg 3180 cggccacctg cagσaσatσa tgaagtggσt ggaaggcagc tcccactgtσ cσgcaggctg 3240 cggccacctc tgcgagtact cctgacgggg catctgctgg gcttgcccgg gcggccgcgt 3300 gcagaaσσcc gcctggtctt gtgcccgaga cgggcggagg ctggaacttg agaσσtσaat 3360 aaaggaagta gagccgctgt cggcactgcg tgtcacccca tagcgcggcg ggctcgccgt 3420 aggcσσgσgg ggcagctggc gggcgggggσ tttcggσggg gttgtσgσca tggtaaccgg 3480 cggcgcccat agtcgctctc ggggσggcct tcccagtgcc tcccgtagag gcctcttccg 3540 gggcgggcca aaggaggtca σttσσggggt σσgtcccggt tcccggcσgt t 3591
<210> 38
<211> 3685
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 1773215CB1
<400> 38 gtcgccgcσt ggcσgggσgt agaσgσggtg gcagagcccg cgcggσgctg gaagσgagtg 60 gcggagcggc gggacctcgg cggaσtcgcc atggaggagg agggtgtgaa ggaagccggt 120 gagaagcctc ggggagcaσa gatggtggaσ aaggctggct ggatσaagaa gagcagtggg 180 ggcctcctgg gtttctggaa agaccgatat ctgctcctct gccaggccca gctgctggtc 240 tatgagaafcg aggatgatca gaagtgtgtg gagactgtgg agctgggσag σtatgagaag 300 tgccaggacc ttcgtgccct cctcaagcga aaacaccgct ttatcctgct gcgatcccca 360 gggaacaagg tcagcgacat caaattccag gcacσσaσσg gggaggagaa ggaatcctgg 420 atcaaagccc tcaatgaagg gattaaccga ggcaaaaaca aggctttcga tgaggtaaag 480 gtggacaaga gctgcgccct ggagcatgtg acacgggacc gggtgcgagg gggccagcga 540 cgccggccac caacgagagt ccacctgaag gaggtggcca gtgcagcttc tgacggtctt 600 ctgσgσσtgg atcttgatgt tccggacagt gggσcacσag tgtttgcccc cagcaatcat 660 gtcagtgaag cccaacctcg ggagacaccc cggcccctca tgcctcctac caagcctttc 720 ctagσaσσtg agaσσaσcag σcctggtgac agggtggaga cσσctgtggg ggagagagcc 780 ccaacccctg tctcagcaag ctctgaggtc tcccctgaga gccaagagga ctcagagacc 840 ccagcagagg aggacagtgg ctσtgagσag cctcccaaca gσgtσctgcc tgacaaaσtg 900 aaggtgagct gggagaaccc cagcccccag gaggcccctg ctgcagagag tgcagaaccg 960 tσσσaggσaσ σσtgttctga gacttctgag gctgccσσσa gggagggtgg gaagccccct 1020 acacccccac ccaagatctt atcagagaaa ctgaaagcct ccatgggtga gatgcaggct 1080 tctgggccac ctgctcσagg cacagtgcag gtctcagtga atggcatgga tgacagtσσt 1140 gagcctgcca agccctctca ggctgagggc accccaggaa ctcctccaaa ggatgcaaca 1200 aσatσσaσag σaσtgccccc σtgggaσσtg σσaσctcagt tσσatccσcg ctgctcctcc 1260 cttggggact tgcttgggga aggcccgcgg catcσcttgc agcccaggga acggctatat 1320 σgggcccagσ tggaggtgaa ggtggσσtσg gaaσagaσgg agaaactgtt gaacaaggtg 1380 ctgggcagtg agccggcccc tgttagtgcc gaaacattgc tcagccaggc tgtggagcag 1440 ctgaggcagg cσaσccaggt cctgσaggaa atgagagatt tgggagagct gagccaggaa 1500 gcacctgggc taagggagaa gcggaaggag ctggtgaccc tctacaggag aagtgcaccc 1560 tagggcσttσ tgggccagag gcaccatccc ttctggσσat σcatcaagtc catσaaggcσ 1620 cagccctgct gagaaatgtg cttctgcttc tacagcaatg gctgcaggag ggccattggg 1680 catgtσaggg tttggσσatg aσσσgaagag aσtσctggcg tccttcctac tctgctctgg 1740 ccagtggtgc caggtgccac ccagggctac tgcctggcta tctggσσtgg σctctgggct 1800 ggggctgggg ctgggagcac acacgctggg acσtatgtgt ttgtgtggtc gttccaaact 1860 gccccagggc tttgggggcg gcacttgggg tttctgggaa tgacatcatc tctgttccσσ 1920 atσσσσagta gtttacattc ctgacttctg aatacagcac agctgagccc cctgcagσtc 1980 ccatctccag ctattcctag gcaaagagcc tcatggctaa ggcagcctca aagσcagccc 2040 σtcσtcccac ctattctgag tagσtgcaga ggσσttgggt cσaggσtcta ggttcatccσ 2100 tσagttgggg ggaacgtagg acccagctgg agcctcttga gggagatgag aggcctcttt 2160 gtgaggagga cattagσtgt gtggσσtσtc tσtσtttggσ σσtgtttσct tttttgcaaa 2220 acaaggacat tttctgcagc cccttcctct cagtgagcta tgattggagg gcttaggtct 2280 ggaggattca agagtggaag aggaatttaa ggggtcσcct agtctagtct ctgcccctgg 2340 atagtgtcca gccttgtata tttctgaaga ggtggatccc agagtggctc tgatgtccac 2400 attagaaaaa σttacttgta atgatcatgt cagσσttcag aagagaatcc cσacσaaσtt 2460 ctgtgcctcc tcagatgggg atttatctgg atctctgtgg ttccttctca gccgaaacag 2520 gtccagtatc ccagtσattt cttcaaatgσ tgataggggt atgttggaat ccgaagccac 2580 ttccccgcct tcaagcccca gatgggctgc tctcctgtaa ctttctagga gaagagacat 2640 tttcttcttt σcctttσctg gtccatcσct gcaσσctggt cctctcσcag cctctσσσσσ 2700 acattgtccc tgactctagg ggcacatcca gtctccatcg tgctgcagca gctggactga 2760 gggcagagcσ tgtaggtgca gaggccσtgg σtσccgaggt cσagσcaσtσ tσσσtggggc 2820 ctctggggtg agagcagctt ccgataggaσ ctgcccagat ttctgcatgt gcacttttgt 2880 ttactgaaag agagaaaggg gggggtcaca gcaacatgcc ctggcctttc tgctctgttc 2940 ccσaaccσca ctgaggcctg ctgcacaggt caatgccttc gttatcgtta ttgtactgtc 3000 actttgttct tgaggtagta gtcaaggatc aggaggggca gatgtσttct ctgggctgcg 3060 tggggccgga gcagaggtga gcagcaatgc actggttcgg gagcccccat cagcctcctt 3120 gtgσaaaσtg ggσcσσσatg σcacagtctg gσtttccctc catctgcccσ aggaσaagag 3180 caagaaggac atcagttgcc cagtcatgtg atcccctgcc atcttgcctt aggaacagcc 3240 ttcσcccacσ agcagccatg gctggctggg gctttagcca agccacctac tgcσaggaat 3300 tggagcctca gttccctcct gtgtcaagta gctaactgca gcagctggac tgagggcaga 3360 gtctgtgggt gcagagacσc tgcatgtagg tcaσaggttg aggcccagcc actctccσtg 3420 gggcctggtg ggtaggcaag tagctctggg gccacctcaa gtgaccaaat gctattaatt 3480 tcσatccttt agcaggσtgg gccctaggσa ggaagctggc ttσtgggaga ggagtgagaa 3540 cgtgcagggc ctgcctagct tgcgtgcttg aggaaggtgg cattccgtgc ttgcctcctt 3600 gaggagggtg gcattσtgtg tσttctgctt atgaagcgcσ tttcttaaag tttggcaata 3660 aatccatttt tatggaaaaa aaaaa 3685
<210> 39
<211> 3143
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No : 303698SCB1
<400> 39 ttcggctcga ggtaagactg cgacggtacc ggggcggcgg ggaaggaccg agaggcggga 60 ggagcagσgg ctcaggcgcc tgcaaaσtgg tggσσtgaac gaggtagaσσ atgaσtgtgg 120 tttcagtggc gtcactcgct gggσtgctct tcctgaggtt ttσctaagcc atcccctggc 180 ggaaccgccσ σcagtttttg tccagaagtg cttatagaaa agatggσtaa tattaaσσta 240 aaagaaataa ccttaatagt aggtgtggtt actgcctgct attggaacag cctcttttgt 300 ggttttgttt ttgatgatgt ttcagcaata ctggataaca aagacttgσa tccatctaca 360 σctttaaaaa ctttatttca aaatgacttc tggggaaccc ctatgtctga ggagagaagc 420 cacaagtctt accgtccctt aaσagtattg acatttσgσt taaattattt gttaagtgaa 480 σtaaaacσaa tgtcatatca tctcctgaafc atgattttfcc atgctgtggt tagtgtgata 540 tttctcaaag tatgcaaact ttttctggac aacaagagta gtgtgattgc ttctttactt 600 tttgcagtgc acccaataca cacagaagca gtaacaggag ttgttggaag agcagaactt 660 ttgtcatcta tcttttttct agσagctttt ttgtcatata ccagatcaaa aggaccagac 720 aattccataa tatggactcσ aattgccttg acagtgtttt tagtggctgt tgcaacatta 780 tgtaaagaac aaggaataac agttgtagga atttgσtgtg tgtatgaagt gtttattgcc 840 caggggtata ctttgccatt actatgtact actgctggaσ agtttctccg tggaaagggt 900 agcattccat tttctatgct gσagaσaσta gtaaaaσtca ttgtcttgat gttcagtaca 960 ttattacttg ttgtgattag agtccaggtt attcaatccc aacttσcagt attσaccagg 1020 tttgataacc cagctgσtgt aagσccaaσt cctacaaggc aactaacttt taactacσtσ 1080 σttcctgtga atgcttggtt gttattaaat ccttcagagc tctgctgtga ttggaccatg 1140 ggaaσaataσ σaσttataga gtcattacta gatattcgaa atctggccac atttactttc 1200 ttttgttttc tggggatgtt gggagtattc agtatcagat actctggtga ttcctσcaag 1260 actgttttaa tggcgctttg tttaatggca ttaccattta ttcctgcatc gaaccttttt 1320 tttcσagtfcg gatttgttgt tgcσgagcga gtattatatg ttcccagcat ggggttctgt 1380 attttggtag cccatggatg gcagaaaata tcaacaaaaa gtgtatttaa aaagctatcc 1440 tggatttgtc tgtctatggt gatactcact cattccttaa aaacattcca cagaaattgg 1500 gattgggagt ctgaatatac attgtttatg tcagccttga aggtaaataa aaataafcgcσ 1560 aaactttgga ataatgtggg tcatgctctg gaaaatgaaa agaactttga gagagctttg 1620 aaatacttct taσaggσtaσ σcatgttcag cσagatgata ttggtgccca tatgaatgta 1680 ggaagaactt ataaaaattt aaatagaacc aaagaagctg aagaatctta catgatggct 1740 aaatcaσtga tgcσtcaaat tattcctggt aaaaaatatg cagccagaat tgcccctaac 1800 cacctaaatg tttatatcaa tctggctaac ctgatccgag caaatgagtc ccgactggaa 1860 gaagcagatc agctgtaccg tcaagcaata agcatgaggc ccgacttcaa gσaggcttac 19-20 attagcagag gagaattgct tttaaaaatg aataaacctc ttaaagcaaa ggaagcatat 1980 cttaaagcac tagagctgga σagaaataat gcagatcttt ggtacaactt ggcaattgta 2040 catattgaac ttaaagaacc aaatgaagcc ctaaaaaact ttaatcgtgc tctggaacta 2100 aatσcaaagc ataaactagc attattcaac tctgctatag taatgσaaga atcaggtgag 2160 gttaaactca gacctgaagc tagaaaacga cttσtaagtt atataaatga agagccacta 2220 gatgctaatg ggtatttcaa tttgggaatg cttgσσatgg atgacaaaaa ggaσaatgaa 2280 gσagagattt ggatgaagaa agccataaag ttacaagccg acttccgaag tgctttgttt 2340 aatσtggσtσ tcctgtattc σσagactgca aaggaattaa aggσtttgcc aattttggag 2400 gagttactca gatactaccc tgatcatatc aagggcctca ttttaaaagg agacattctg 2460 atgaatcaaa agaaagatat actaggagca aaaaaatgtt ttgaaaggat tttggagatg 2520 gatccaagca atgtgcaagg aaaacacaat ctttgtgttg tttattttga agaaaaagac 2580 ttattaaaag ctgaaagatg cσttσttgaa acaσtggcat tagσaccaσa tgaagaatat 2640 attcagcgcc atttgaatat agtcagggat aagatttcct catctagttt tatagagcca 2700 atattcσσaa σσagtaagat ttcaagtgtg gaaggaaaga aaattccaac tgaaagtgta 2760 aaagaaatta gaggtgaatc cagacaaaca caaatagtaa aaacaagtga taataaaagt 2820 cagtctaaat cσaaσaaaca attaggaaaa aatggagacg aagagacacc cσaσaaaaσa 2880 acaaaagaca tcaaagaaat tgagaagaaa agagttgctg ctttaaaaag actagaagag 2940 attgaacgta ttttaaatgg tgaataacat taatatttat cgtgaσaatg gtatcaaaga 3000 acatcaatcc gtatcatgtg attgctttta ctgggagctt tgaaaaaaag ttcaagggtt 3060 cctaatggtc aatcatgagσ tgccttgaag taggatcaaa ataagatttt cattaaagac 3120 ctgtattatc ccaaaaaaaa aaa 3143
<210> 40
<211> 1759
<212> DNA
<213> Homo sapiens
<220>
<221> misc_feature
<223> Incyte ID No: 2041080CB1
<400> 40 ggggctctgc gcgagcgagg gacgacggaa gggacgggca ggtgtgggcg cggggccacg 60 cagccσgaσg gcgggagtcg caggtgctgg gtgcatgggσ σagtgaggac gcacagagat 120 ccctcgccgc gcggaggagg agcagcgcgg gagccaggcg ctgccccaag accctgcctg 180 cgtσσgagσg agσggaaσσt σgcgσttcgc ccggggacaa tσσgaagtσσ gσgctatgga 240 agaggagaaa tatttgcctg agctgatggc agagaaagat agcctggatc catcttttgt 300 gcatgcgtcg cgccttttgg cagaagaaat tgaaaagttt σaaggttσtg atggaaaaaa 360 ggaagacgaa gaaaagaagt atcttgatgt catcagcaac aaaaacataa agctctcaga 420 aagagtactg attσσtgtca agcagtatcc aaagttcaat tttgtgggga aattgcttgg 480 accaagagga aactccttga agaggctaca ggaagaaaca ggtgctaaaa tgtctatcct 540 gggcaaagga tcaatgagag ataaagctaa ggaagaagaa σtaaggaaga gtggggaagσ 600 caaatatgcc cacttgagtg atgagcttca tgtattaatt gaagtgtttg ctccacctgg 660 ggaagcttat- tσaσgtatga gtcatgσatt ggaagagatt aaaaaattcc tggttcctga 720 ctacaatgat gaaattcgtc aggaacaact acgtgaatta tcttacttaa atggctcaga 780 ggaσtctggt σgtggcagag gtattagagg cagagggatσ agaatagσtσ σcacagσtcc 840 ttcaaggggc cgtgggggtg ccattcctcc tcccccacca cctggacgag gtgttctcac 900 cσσtσgggga agσaσtgtaa cccgtggagc gcttσσagtg ccacctgtag caagaggtgt 960 ccctacccct cgagcccggg gggcaccaac agtgccagga tacagggcac ctcctcctcc 1020 agcccatgaa gcttatgaag aatatggtta tgatgatggσ tacgggggtg aatatgatga 1080 ccagacctat gagacttatg ataacagcta tgcgacccaa acacaaagtg tgcctgaata 1140 ctatgactaσ ggtcatggag taagtgagga tgcctatgac agσtacgcac cagaagaatg 1200 ggccaσaacc cgctctagct tgaaggcacc accgcagagg tcagccagag ggggatacag 1260 ggaacaσccc tatggtagat attgaaggtc σttσccaσct gtgacctσaσ σtcaaagaσa 1320 attcatagcc tgtggtctcc acataaacag caacaagaca agtacccaca aacaaagaac 1380 acataattga ataaccgatg tgatttgcta agaaggatgg aggtgaagat gtatgataga 1440 aaaacgaaaa actggaataa acccaagact ggctgtttgg aggtttaatg tttttaacaa 1500 tggaggccaa aaaagaagat aatttggatt tgaaccaata ttttcttσat σtcaactσaσ 1560 ctcaccacta cagatccaag cctgacacag agcctaacag aaatactttc ctccaaatac 1620 aatσctgttc accatggaag acattaaaaσ cttcagtggt tgctgσtggg ggcσttσacc 1680 aaaatgcagc cattcacctt gctgctgggg gccttcacca aaatgcagcc atfccaccttc 1740 tttgcttttg ctctaccta 1759
PCT/US2001/032090 2000-10-13 2001-10-12 Intracellular signaling molecules WO2002031152A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2002216625A AU2002216625A1 (en) 2000-10-13 2001-10-12 Intracellular signaling molecules
US10/399,456 US20040043395A1 (en) 2001-10-12 2001-10-12 Intracellular signaling molecules

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US24087100P 2000-10-13 2000-10-13
US60/240,871 2000-10-13
US24472300P 2000-10-30 2000-10-30
US60/244,723 2000-10-30
US24940200P 2000-11-15 2000-11-15
US60/249,402 2000-11-15
US25262200P 2000-11-22 2000-11-22
US60/252,622 2000-11-22
US25562200P 2000-12-13 2000-12-13
US60/255,622 2000-12-13

Publications (2)

Publication Number Publication Date
WO2002031152A2 true WO2002031152A2 (en) 2002-04-18
WO2002031152A3 WO2002031152A3 (en) 2004-02-26

Family

ID=27540140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/032090 WO2002031152A2 (en) 2000-10-13 2001-10-12 Intracellular signaling molecules

Country Status (2)

Country Link
AU (1) AU2002216625A1 (en)
WO (1) WO2002031152A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002026818A3 (en) * 2000-09-27 2003-08-28 Aeomica Int Human nedd-1
WO2003095483A1 (en) * 2002-05-10 2003-11-20 Universitaetsklinikum Heidelberg Dna encoding for a gtpase activating protein
EP1507004A1 (en) * 2003-08-14 2005-02-16 DKFZ Deutsches Krebsforschungszentrum Method to inhibit the propagation of an undesired cell population
CN102766194A (en) * 2011-05-03 2012-11-07 中国医学科学院医药生物技术研究所 Oligopeptide compound with HIV-1 protease inhibitory activity, and preparation method and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3395900A (en) * 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL [Online] EBI; 12 April 2000 (2000-04-12) NCI-CGAP: "zs52c04.r1 NCI_GAP_GCB1 Homo sapiens cDNA clone IMAGE:701094 5', mRNA sequence." Database accession no. AA287437 XP002224184 *
DATABASE EMBL [Online] EBI; 3 March 2000 (2000-03-03) NIH-MGC: "UI-HF-BN0-akf-c-03-0-UI.r1 NIH_MGC_50 Homo sapiens cDNA clone IMAGE:3076972 5', mRNA sequence." Database accession no. AW499907 XP002224185 *
DATABASE NAGENESEQ [Online] DERWENT; Publication date of WO00/55350: 21-09-200, 8 February 2001 (2001-02-08) HUMAN GENOME SCI INC.: "Human cancer associated gene sequence SEQ ID NO:419" retrieved from EBI Database accession no. AAC78025 XP002224183 & WO 00 55350 A (HUMAN GENOME SCI INC.) 21 September 2000 (2000-09-21) *
DRUEY K M ET AL: "INHIBITION OF G-PROTEIN-MEDIATED MAP KINASE ACTIVATION BY A NEW MAMMALIAN GENE FAMILY" NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 379, February 1996 (1996-02), pages 742-746, XP002929721 ISSN: 0028-0836 cited in the application *
KLOEKER SUSANNE ET AL: "Purification and identification of a novel subunit of protein serine/threonine phosphatase 4." JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 274, no. 9, 26 February 1999 (1999-02-26), pages 5339-5347, XP002224179 ISSN: 0021-9258 cited in the application *
MINAMOTO SEIJIRO ET AL: "Cloning and functional analysis of new members of STAT induced STAT inhibitor (SSI) family: SSI-2 and SSI-3." BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 237, no. 1, 1997, pages 79-83, XP002224181 ISSN: 0006-291X *
OKADA TAKAHARU ET AL: "BCAP: The tyrosine kinase substrate that connects B cell receptor to phosphoinositide 3-kinase activation." IMMUNITY, vol. 13, no. 6, December 2000 (2000-12), pages 817-827, XP002224182 ISSN: 1074-7613 *
VINCENT STEPHANE ET AL: "The Drosophila protein Dof is specifically required for FGF signaling." MOLECULAR CELL, vol. 2, no. 4, October 1998 (1998-10), pages 515-525, XP002224178 ISSN: 1097-2765 *
YU YINHUA ET AL: "NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 96, no. 1, 5 January 1999 (1999-01-05), pages 214-219, XP002224180 Jan. 5, 1999 ISSN: 0027-8424 cited in the application *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002026818A3 (en) * 2000-09-27 2003-08-28 Aeomica Int Human nedd-1
WO2003095483A1 (en) * 2002-05-10 2003-11-20 Universitaetsklinikum Heidelberg Dna encoding for a gtpase activating protein
EP1507004A1 (en) * 2003-08-14 2005-02-16 DKFZ Deutsches Krebsforschungszentrum Method to inhibit the propagation of an undesired cell population
WO2005016366A2 (en) * 2003-08-14 2005-02-24 Deutsches Krebsforschungszentrum Method to inhibit the propagation of an undesired cell population
WO2005016366A3 (en) * 2003-08-14 2005-07-21 Deutsches Krebsforsch Method to inhibit the propagation of an undesired cell population
CN102766194A (en) * 2011-05-03 2012-11-07 中国医学科学院医药生物技术研究所 Oligopeptide compound with HIV-1 protease inhibitory activity, and preparation method and application thereof

Also Published As

Publication number Publication date
AU2002216625A1 (en) 2002-04-22
WO2002031152A3 (en) 2004-02-26

Similar Documents

Publication Publication Date Title
EP1266001A2 (en) Human transcription factors
EP1203081A2 (en) Human intracellular signaling molecules
WO2002024895A2 (en) Transcription factors and zinc finger proteins
CA2449272A1 (en) Intracellular signaling molecules
WO2003052049A2 (en) Molecules for disease detection and treatment
US20030211513A1 (en) Intracellular signaling proteins
US6962799B2 (en) Microtubule-associated proteins and tubulins
WO2002031152A2 (en) Intracellular signaling molecules
EP1278843A2 (en) Rna metabolism proteins
US20040053396A1 (en) Molecules for disease detection and treatment
US20040024182A1 (en) Gtp-binding proteins
US20040126759A1 (en) Molecules for disease detection and treatment
WO2004067712A2 (en) Intracellular signaling molecules
WO2002046413A2 (en) Molecules for disease detection and treatment
WO2002078420A2 (en) Molecules for disease detection and treatment
US20040029144A1 (en) Transcription factors and zinc finger proteins
EP1390410A1 (en) Molecules for disease detection and treatment
WO2002077235A2 (en) Intracellular signaling molecules
US20040038267A1 (en) Intracellular signaling molecules
EP1379654A2 (en) Intracellular signaling molecules
WO2002004510A2 (en) Gtp-binding proteins
US20030186379A1 (en) Secretion and trafficking molecules
US20040043395A1 (en) Intracellular signaling molecules
US20040092715A1 (en) Intracellular signaling molecules
US20040023251A1 (en) Cell cycle proteins and mitosis-associated molecules

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 10399456

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: JP