EP1268506A4 - Secreted proteins and uses thereof - Google Patents

Secreted proteins and uses thereof

Info

Publication number
EP1268506A4
EP1268506A4 EP00950916A EP00950916A EP1268506A4 EP 1268506 A4 EP1268506 A4 EP 1268506A4 EP 00950916 A EP00950916 A EP 00950916A EP 00950916 A EP00950916 A EP 00950916A EP 1268506 A4 EP1268506 A4 EP 1268506A4
Authority
EP
European Patent Office
Prior art keywords
seq
accession number
tango
amino acid
number pta
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP00950916A
Other languages
German (de)
French (fr)
Other versions
EP1268506A1 (en
Inventor
Christopher C Fraser
John D Sharp
Susan J Kirst
Thomas M Barnes
Nick Wrighton
Paul S Myers
Yang Pan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Millennium Pharmaceuticals Inc
Original Assignee
Millennium Pharmaceuticals 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 Millennium Pharmaceuticals Inc filed Critical Millennium Pharmaceuticals Inc
Publication of EP1268506A1 publication Critical patent/EP1268506A1/en
Publication of EP1268506A4 publication Critical patent/EP1268506A4/en
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • secreted proteins for example, cytokines and cytokine receptors
  • cytokines and cytokine receptors play a vital role in the regulation of cell growth, cell differentiation, and a variety of specific cellular responses.
  • a number of medically useful proteins including erythropoietin, granulocyte- macrophage colony stimulating factor, human growth hormone, and various interleukins, are secreted proteins.
  • an important goal in the design and development of new therapies is the identification and characterization of secreted and transmembrane proteins and the genes which encode them.
  • receptors which bind a ligand and transduce an intracellular signal, leading to a variety of cellular responses.
  • the identification and characterization of such a receptor enables one to identify both the ligands which bind to the receptor and the intracellular molecules and signal transduction pathways associated with the receptor, permitting one to identify or design modulators of receptor activity, e.g., receptor agonists or antagonists and modulators of signal transduction.
  • the present invention is based, at least in part, on the discovery of cDNA molecules which encode the TANGO 339, TANGO 353, TANGO 358, TANGO 365,
  • the TANGO 339 proteins are transmembrane 4 domain-containing polypeptides that exhibit homology to human CD9 antigen, a cell surface antigen associated with platelet activation and aggregation.
  • the TANGO 353, TANGO 358, and TANGO 365 proteins are transmembrane proteins.
  • the TANGO 368 proteins are secreted proteins encoded by sequences with homology to genomic sequences of the human T-cell receptor gamma VI gene region.
  • the TANGO 383 proteins are transmembrane polypeptides with homology to retinopathy proteins.
  • the TANGO 393 protein are transmembrane proteins with homology to proteins containing Leucine-rich repeats (LRR) such as the Leucine-Rich -2-Glycoprotein (LRG), SLIT- 1 , and Platelet Glycoprotein V (GPV) precursor.
  • LRR Leucine-rich repeats
  • the TANGO 402 proteins are homologous to the LOX-1 protein, which has been associated with low density lipoprotein metabolism and atherosclerosis.
  • the MANGO 346, MANGO 349, and TANGO 369 proteins are secreted proteins.
  • TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 proteins, fragments, derivatives, and variants thereof are collectively referred to herein as "polypeptides of the invention” or “proteins of the invention.”
  • Nucleic acid molecules encoding the polypeptides or proteins of the invention are collectively referred to as "nucleic acids of the invention.”
  • nucleic acids and polypeptides of the present invention are useful as modulating agents in regulating a variety of cellular processes. Accordingly, in one aspect, this invention provides isolated nucleic acid molecules encoding a polypeptide of the invention or a biologically active portion thereof. The present invention also provides nucleic acid molecules which are suitable for use as primers or hybridization probes for the detection of nucleic acids encoding a polypeptide of the invention.
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:2, the nucleotide sequence of the cDNA insert of an EpT339 clone deposited with the American Type Culture Collection (ATCC®) as Accession Number PTA-292, or a complement thereof.
  • ATCC® American Type Culture Collection
  • the invention features nucleic acid molecules, which are at least 30% ⁇ , 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:27, SEQ ID NO:28, the nucleotide sequence of the cDNA insert of an EpT353 clone deposited with the ATCC® as Accession Number PTA-292, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:36, SEQ ID NO:37, the nucleotide sequence of the cDNA insert of an EpT358 clone deposited with the ATCC® as Accession Number PTA-292, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:44, SEQ ID NO:45, the nucleotide sequence of the cDNA insert of an EpT365 clone deposited with the ATCC® as Accession Number PTA-291, or a complement thereof.
  • the invention features nucleic acid molecules, preferably cDNA molecules, which are at least 98% identical to the nucleotide sequence of SEQ ID NO:52, SEQ ID NO:53, the nucleotide sequence of the cDNA insert of an EpT368 clone deposited with the
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%,
  • SEQ ID NO:58 SEQ ID NO:59, the nucleotide sequence of the cDNA insert of an EpT369 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 40%, 45%, 50%,
  • the invention also features nucleic acid molecules which are at least 65%, 75%,
  • nucleic acid molecules which are at least 30%, 35%, 40%,
  • SEQ ID NO:73 SEQ ID NO:74, the nucleotide sequence of the cDNA insert of a human
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%,
  • SEQ ID NO:93 SEQ ID NO:94, the nucleotide sequence of a mouse EpT393 cDNA, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO: 110,
  • SEQ ID NO:l 11 the nucleotide sequence of the cDNA insert of an EpT402 clone deposited with the ATCC® as Accession Number PTA-294, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%,
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:128, SEQ ID NO:129, the nucleotide sequence of the cDNA insert of an EpM349 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof.
  • the invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, a complement thereof, or the
  • the invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100 contiguous nucleotides of nucleic acids 1 to 2102 of SEQ ID NO:l, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900 or 1000 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:2, or a complement thereof.
  • the invention features nucleic acid molecules of at least 575, 600, 650, 700, 800, 900, 1000, 1100 or 1200 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:27, the nucleotide sequence of an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 630 contiguous nucleotides of nucleic acids 1 to 634 of SEQ ID NO:27, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650 or 690 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:28, or a complement thereof.
  • the invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550 or 560 contiguous nucleotides of nucleic acids 1 to 560 of SEQ ID NO:28, or a complement thereof.
  • the invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
  • nucleic acid molecules which include a fragment of at least
  • the invention features nucleic acid molecules of at least 20, 50, 100, 150, 200, 250, 300, 340, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200 or 1300 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:44, the nucleotide sequence of an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, or 450 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:45, or a complement thereof.
  • the invention features nucleic acid molecules, preferably a cDNA molecule, which are at least 970 nucleotides of the nucleotide sequence the of the nucleotide sequence of SEQ ID NO:44, the nucleotide sequence of an EpT368 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
  • nucleic acid molecules which include a fragment of at least 20, 50, 100, 150 or 174 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:59, or a complement thereof.
  • the invention features nucleic acid molecules of at least 510, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200 or 1300 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:63, the nucleotide sequence of an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 270, 300, 350, or 400 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 64, or a complement thereof.
  • the invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200 or 250 contiguous nucleotides of nucleic acids 1 to 255 of SEQ ID NO:64, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 610 contiguous nucleotides of nucleic acids 775 to 1386 of SEQ ID NO:63, or a complement thereof.
  • the invention features nucleic acid molecules of at least 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600 or 1700 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 73, the nucleotide sequence of a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, or 1400 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:74, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least
  • the invention features nucleic acid molecules of at least 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, or 1900 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 93, the nucleotide sequence of a mouse EpT393 cDNA, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 984 contiguous nucleotides nucleotides of nucleic acids 1 to 984 of SEQ ID NO:93, or a complement thereof.
  • the invention also features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250 or 292 contiguous nucleotides of the nucleic acids 1177 to 1469 of SEQ ID NO:93, or a complement thereof.
  • the invention also features nucleic acid molecules which include a fragment of at least 20,
  • nucleic acids 1666 to 1946 of SEQ ID NO:93 50, 100, 150, 200, 250 or 280 contiguous nucleotides of the nucleic acids 1666 to 1946 of SEQ ID NO:93, or a complement thereof.
  • the invention features nucleic acid molecules which include a fragment of at least
  • nucleic acid molecules of at least 20, 50, 100, 150, 200,
  • nucleic acid molecules which include a fragment of at least
  • the invention features nucleic acid molecules of at least 450, 500, 550, 600, 650,
  • the invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
  • nucleic acid molecules which include a fragment of at least
  • nucleotide sequence of SEQ ID NO:124 50, 75, 100, or 150 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:124, or a complement thereof.
  • the invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
  • the invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450 or 500 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 129, or a complement thereof.
  • the invention features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650 or more contiguous nucleotides identical to the nucleic acid sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212,
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:29, the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:38, the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:46, the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention also features nucleic acid molecules, preferably cDNA molecules, which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 99% identical to the amino acid sequence of SEQ ID NO:54, the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:60, the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:65, the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA- 295, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:75, the amino acid sequence encoded by a human
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:95, the amino acid sequence encoded by a mouse
  • EpT393 cDNA or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 26%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ED NO:l 12, the amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO: 125, the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO: 130, the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25% preferably 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA- 292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof, wherein the protein encoded by the nucleotide sequence also exhibits at least one structural and/or functional feature of a polypeptide of the invention.
  • the nucleic acid molecules have the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA clones of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number P
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:3, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175, 200, 225, 250, 260 or 265 contiguous amino acids of SEQ ID NO:3, or the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:29, or a fragment thereof including at least 45, 50, 75, 100, 125, 150, 175, 200 or 225 contiguous amino acids of SEQ ID NO:29, or the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:38, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, or 80 contiguous amino acids of SEQ ID NO:38, or the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:46, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 90, 100, 125, 150 or 160 contiguous amino acids of SEQ ID NO:46, or the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
  • nucleic acid molecules preferably cDNA molecules, which encode a polypeptide having the amino acid sequence of SEQ ID NO:54, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 60, 70, 75 or 80 contiguous amino acids of SEQ ID NO:54, or the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:60, or a fragment thereof including at least 10, 15, 20, 25, 30, 50 or 55 contiguous amino acids of SEQ ID NO:60, or the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:65, or a fragment thereof including at least 90, 100, 110, 125 or 135 contiguous amino acids of SEQ ID NO:65, or the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:75, or a fragment thereof including at least 60, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400 or 450 contiguous amino acids of SEQ ID NO:75, or the amino acid sequence encoded by a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 95, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400 or 450 contiguous amino acids of SEQ ID NO:95, or the amino acid sequence encoded by a mouse EpT393 cDNA, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:l 12, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175 or 200 contiguous amino acids of SEQ ID NO:l 12, he amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 125, or a fragment thereof including at least 10, 15, 20, 25, 30, 35, 40, 45, 50 or 55 contiguous amino acids of SEQ ID NO:125, or the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
  • nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 130, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150 or 160 contiguous amino acids of SEQ ID NO: 130, or the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention also features nucleic acid molecules which encode a polypeptide fragment of at least 10, 15, 25, 30, 50, 75, 100, 125, 150, 175, 200 or more contiguous amino acids of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA- 292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof, wherein the fragment exhibits at least one structural and/or functional feature of a polypeptide of the invention.
  • the invention includes nucleic acid molecules which encode an allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof under stringent conditions.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, or the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:29, or the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:38, or the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:46 or the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins having an amino acid sequence that is at least 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:54, or the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins having an amino acid sequence that is at least 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:60, or the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 65%, preferably 70%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:65 or the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA-295.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:75 or the amino acid sequence encoded by a human EpT393 cDNA of ATCC® Accession Number PTA-295.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%o, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:95 or the amino acid sequence encoded by a mouse EpT393 cDNA.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 26%, preferably 30%, 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:l 12, or the amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 125 or the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 130 or the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295.
  • the invention also features isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 52, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:3, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 1 or 2, a complement thereof, or the non-coding strand of an EpT339 cDNA of ATCC® Accession Number PTA-292.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:29, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:27 or 28, a complement thereof, or the non-coding strand of an EpT353 cDNA of ATCC® Accession Number PTA-292.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:38, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:36 or 37, a complement thereof, or the non-coding strand of an EpT358 cDNA of ATCC® Accession Number PTA-292.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:46, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:45, a complement thereof, or the non-coding strand of an EpT365 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:54, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:52 or 53, a complement thereof, or the non-coding strand of an EpT368 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:60, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 58 or 59, a complement thereof, or the non-coding strand of an EpT369 cDNA of ATCC® Accession Number PTA-295.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 65%, preferably 70%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:65, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:64, a complement thereof, or the non-coding strand of an EpT383 cDNA of ATCC® Accession Number PTA-295.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 75, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:74, a complement thereof, or the non-coding strand of a human EpT393 cDNA of ATCC® Accession Number PTA 295.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:96, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:95, a complement thereof, or the non-coding strand of a mouse EpT393 cDNA.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about preferably 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:l 12, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l 10 or 111, a complement thereof, or the non-coding strand of an EpT402 cDNA of ATCC® Accession Number PTA-294.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 125, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 124, a complement thereof, or the non-coding strand of an EpM346 cDNA of ATCC® Accession Number PTA-291.
  • isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 130, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 129, a complement thereof, or the non-coding strand of an EpM349 cDNA of ATCC® Accession Number PTA-295.
  • the invention features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216
  • the invention also features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to a nucleic acid sequence encoding SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, isolated polypeptides or proteins which are encode
  • polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
  • polypeptides which are allelic variants of a polypeptide that includes the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC®
  • polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule having the sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129, or a complement thereof under stringent conditions.
  • polypeptides which are allelic variants of a polypeptide that includes the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, respectively, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule having the sequence of SEQ ID NO: 1, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129, or a complement thereof under stringent conditions, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l or 2, or an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the nucleic acid molecules are at least 480, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600 or 2700 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900 or 1000 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2 or nucleic acids 1 to 2100 of SEQ ID NO:l, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:27, or an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the nucleic acid molecules are at least 575, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150 or 1200 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:27 or 28, an EpT353 cDNA of ATCC® Accession Number PTA- 292, or a complement thereof.
  • the nucleic acid molecules are at least 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650 or 690 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:28, an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550 or 560 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising nucleotides 1 to 500 of SEQ ID NO:28, an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:36 or 37, or an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the nucleic acid molecules are at least 50, 100, 150, 200 or 240 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:36 or 37, an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:44, or an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, or 1150 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:44, an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450 or 490 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:45, an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:58 or 59, or an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150 or 174 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:58 or 59, an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 63, or an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, or 600 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides of SEQ ID NO:63, or an EpT 283 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 250 and/or 800 to 1386 of SEQ ID NO:63.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 73, or a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350 or 1386 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides from of SEQ ID NO:73, a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 1250 of SEQ ID NO:73.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:93, or a mouse EpT393 cDNA, or a complement thereof.
  • the nucleic acid molecules are at least 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800 or 1900 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides of SEQ ID NO:93, a mouse EpT393 cDNA or a complement thereof.
  • such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 950 and or 1200 to 1800 of SEQ ID NO:93.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l 10 or 111, or an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 620 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 110 or 111, an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 123, or an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the nucleic acid molecules are at least 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100 or 1150 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 123, an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
  • the nucleic acid molecules are at least 50, 75, 100, 125, 150 or 175 contiguous nucleotides in length and hybridize under stringent conditions to the nucleotide sequence of SEQ ID NO: 124, or a complement thereof.
  • the invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 128, or an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
  • the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, 3000, 3100, 3200, 3300, 3400, 3500, or 3600 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 128, an EpM349 cDNA of ATCC® Accession Number PTA- 295, or a complement thereof.
  • the invention also features nucleic acid molecules at least 15, preferably 50, 75, 100, 150, 200, 250, 300, 350, 400, 500, 600 or more contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220,
  • the invention also features nucleic acid molecules at least 15, preferably 50, 75, 100, 150, 200, 250, 300, 350, 400, 500, 600 or more contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220,
  • the isolated nucleic acid molecules encode a cytoplasmic, transmembrane, or extracellular domain of a polypeptide of the invention.
  • the invention provides an isolated nucleic acid molecule which is antisense to the coding strand of a nucleic acid of the invention.
  • vectors e.g., recombinant expression vectors, comprising a nucleic acid molecule of the invention.
  • the invention provides host cells containing such a vector or engineered to contain and/or express a nucleic acid molecule of the invention.
  • the invention also provides methods for producing a polypeptide of the invention by culturing, in a suitable medium, a host cell of the invention containing a recombinant expression vector encoding a polypeptide of the invention such that the polypeptide of the invention is produced.
  • Another aspect of this invention features isolated or recombinant proteins and polypeptides of the invention.
  • Preferred proteins and polypeptides possess at least one biological activity possessed by the corresponding naturally-occurring human polypeptide.
  • An activity, a biological activity, or a functional activity of a polypeptide or nucleic acid of the invention refers to an activity exerted by a protein, polypeptide or nucleic acid molecule of the invention on a responsive cell as determined in vivo or in vitro, according to standard techniques.
  • activities can be a direct activity, such as an association with or an enzymatic activity on a second protein, or an indirect activity, such as a cellular signaling activity mediated by interaction of the protein with a second protein.
  • biological activities include, e.g., (1) the ability to modulate (this term, as used herein, includes, but is not limited to, stabilize, promote, inhibit or disrupt) the development, differentiation, proliferation and/or activity of immune cells (e.g., B-lymphocyte function); (2) the ability to modulate the development and progression of cancer (e.g.
  • lymphomas and/or melanoma-associated cancer (3) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein- ligand interactions, e.g., in receptor-ligand recognition; (4) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; (5) the ability to modulate hematopoietic processes; (6) the ability to modulate platelet activation and aggregation; (7) the ability to modulate intracellular signaling cascades
  • the ability to modulate intercellular signaling e.g., in the nervous system
  • the ability modulate the development, differentiation, proliferation and/or activity of neuronal cells and glial cells e.g., oligodendrocytes and astrocytes
  • the ability to modulate the development, differentiation and activity of eye structures, such as the retina e.g., the ability to modulate photoreceptor disk morphogenesis
  • (11) the ability to modulate the development of organs, tissues and/or cells in an embryo and/or fetus e.g., signal transduction cascades
  • the ability to modulate intercellular signaling e.g., in the nervous system
  • the ability modulate the development, differentiation, proliferation and/or activity of neuronal cells and glial cells e.g., oligodendrocytes and astrocytes
  • the ability to modulate the development, differentiation and activity of eye structures such as the retina
  • the ability to modulate photoreceptor disk morphogenesis e.g
  • biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of immune cells, such as lymphocytes (e.g., T cells and B cells); (2) ability to modulate cell proliferation, e.g., abnormal cell proliferation;(3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. , in receptor-ligand recognition.
  • immune cells such as lymphocytes (e.g., T cells and B cells);
  • ability to modulate cell proliferation e.g., abnormal cell proliferation
  • intracellular signaling cascades e.g., signal transduction cascades
  • intercellular signaling e.g., in
  • biological activities include, e.g., (1) the ability to modulate development, differentiation, maturation, proliferation and/or activity of immune cells such as thymocytes, e.g., T- lymphocytes; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
  • immune cells such as thymocytes, e.g., T- lymphocytes
  • intracellular signaling cascades e.g., signal transduction cascades
  • intercellular signaling e.g., in the immune system
  • protein-protein interactions e.g., homophil
  • biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of prostate cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor-ligand recognition.
  • biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of cells, such as immune cells, e.g., natural killer cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
  • cells such as immune cells, e.g., natural killer cells
  • intracellular signaling cascades e.g., signal transduction cascades
  • intercellular signaling e.g., in the immune system
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions
  • biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of cells, such as immune cells, e.g., natural killer cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
  • cells such as immune cells, e.g., natural killer cells
  • intracellular signaling cascades e.g., signal transduction cascades
  • intercellular signaling e.g., in the immune system
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions
  • biological activities include, e.g., (1) the ability to modulate, e.g. , stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of prostate cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; and (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions.
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor- ligand recognition
  • the ability to modulate the proliferation, differentiation and/or activity of prostate cells e.g., in receptor- ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • intercellular signaling e.g.
  • biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions e g-, homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of hypothalamus cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; and (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions.
  • biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions e g-, homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of hypothalamus cells; (3) the ability to modulate intra
  • biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of immune cells (e.g., leukocytes and macrophages), endothelial cells and smooth muscle cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate the development of organs, tissues and/or cells of the embryo and/or fetus; (5) the ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; (6) the ability to modulate atherosclerosis, e.g., the initiation and progression of atherosclerosis; (7) the ability to modulate low-density lipoproteins e.g., the ability to modulate levels, metabolism and/or cellular uptake of oxidized low-density lipoprotein (Ox-LDL), the ability to bind to Ox-LDL, and the ability to modul
  • biological activities include, e.g., (1) the ability to modulate, e.g. , stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (5) the ability to modulate neural signaling; (6) the ability to modulate intercellular signaling (e.g., in the neural system); and (7) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions.
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor- ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); and (5) the ability to modulate intercellular signaling (e.g., in the immune system).
  • a polypeptide of the invention has an amino acid sequence sufficiently identical to an identified domain of a polypeptide of the invention.
  • the term "sufficiently identical" refers to a first amino acid or nucleotide sequence which contains a sufficient or minimum number of identical or equivalent (e.g., with a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have or encode a common structural domain and/or common functional activity.
  • amino acid or nucleotide sequences which contain or encode a common structural domain having about 60% identity, preferably 65% identity, more preferably 75%, 85%, 95%, 98% identity.
  • a TANGO 339 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain, transmembrane 4 domain, a transmembrane 4-like domain, a peripherin/rom-1 domain, a peripherin/rom-1-like domain, and an intracellular or cytoplasmic domain.
  • a TANGO 353 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain and an intracellular or cytoplasmic domain.
  • an TANGO 358 includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain, and an intracellular or cytoplasmic domain.
  • a TANGO 365 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, at least one transmembrane domain and an intracellular or cytoplasmic domain.
  • a TANGO 368 protein includes at least a signal peptide.
  • a TANGO 369 protein includes at least a signal peptide.
  • a TANGO 383 protein includes at least one or more of the following domains: a signal sequence, at least one transmembrane domain, an intracellular or cytoplasmic domain, and an extracellular domain.
  • a TANGO 393 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, and a transmembrane domain, a leucine-rich repeat domain and an intracellular or cytoplasmic domain.
  • a TANGO 402 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a C-type lectin domain, a C-type lectin-like domain, a transmembrane domain, and an intracellular or cytoplasmic domain.
  • a MANGO 346 protein includes at least a signal sequence.
  • a MANGO 349 protein includes at least a signal sequence.
  • the isolated polypeptide of the invention lacks both a transmembrane and a cytoplasmic domain. In another embodiment, the polypeptide lacks both a transmembrane domain and a cytoplasmic domain and is soluble under physiological conditions.
  • polypeptides of the present invention can be operably linked to a heterologous amino acid sequence to form fusion proteins.
  • the invention further features antibodies that specifically bind a polypeptide of the invention such as monoclonal or polyclonal antibodies.
  • polypeptides of the invention or biologically active portions thereof, or antibodies of the invention can be incorporated into pharmaceutical compositions, which optionally include pharmaceutically acceptable carriers.
  • the present invention provides methods for detecting the presence, activity or expression of a polypeptide of the invention in a biological sample by contacting the biological sample with an agent capable of detecting an indicator of the presence, activity or expression such that the presence activity or expression of a polypeptide of the invention is detected in the biological sample.
  • the invention provides methods for modulating activity of a polypeptide of the invention comprising contacting a cell with an agent that modulates (inhibits or stimulates) the activity or expression of a polypeptide of the invention such that activity or expression in the cell is modulated.
  • the agent is an antibody that specifically binds to a polypeptide of the invention.
  • the agent is a fragment of a polypeptide of the invention or a nucleic acid molecule encoding such a polypeptide fragment.
  • the agent modulates expression of a polypeptide of the invention by modulating transcription, splicing, or translation of an mRNA encoding a polypeptide of the invention.
  • the agent is a nucleic acid molecule having a nucleotide sequence that is antisense to the coding strand of an mRNA encoding a polypeptide of the invention.
  • the present invention also provides methods to treat a subject having a disorder characterized by aberrant activity of a polypeptide of the invention or abe ⁇ ant expression of a nucleic acid of the invention by administering an agent which is a modulator of the activity of a polypeptide of the invention or a modulator of the expression of a nucleic acid of the invention to the subject.
  • the modulator is a protein of the invention.
  • the modulator is a polypeptide (e.g., an antibody or a fragment of a polypeptide of the invention), a peptidomimetic, or other small molecule (e.g., a small organic molecule).
  • the present invention also provides diagnostic assays for identifying the presence or absence of a genetic lesion or mutation characterized by at least one of: (i) abe ⁇ ant modification or mutation of a gene encoding a polypeptide of the invention, (ii) mis- regulation of a gene encoding a polypeptide of the invention, and (iii) abe ⁇ ant post- translational modification of the invention wherein a wild-type form of the gene encodes a protein having the activity of the polypeptide of the invention.
  • the invention provides a method for identifying a compound that binds to or modulates the activity of a polypeptide of the invention.
  • such methods entail, measuring a biological activity of the polypeptide in the presence and absence of a test compound and identifying those compounds which alter the activity of the polypeptide.
  • the invention also features methods for identifying a compound which modulates the expression of a polypeptide or nucleic acid of the invention by measuring the expression of the polypeptide or nucleic acid in the presence and absence of the compound.
  • the invention provides human or non-human antibodies or fragments thereof which specifically bind to a protein of the invention.
  • an antibody or a fragment thereof i.e., human and non- human antibodies or fragments thereof and/or monoclonal antibodies or fragments thereof of the invention, specifically bind to an extracellular domain having the amino acid sequence of SEQ ID NO:20, 21, 32, 41, 51, 89, 109, 112, 115, 136 or 233.
  • Any of the antibodies of the invention can be conjugated to a therapeutic moiety or to a detectable substance.
  • detectable substances that can be conjugated to the antibodies of the invention are an enzyme, a prosthetic group, a fluorescent material, a luminescent material, a bioluminescent material, and a radioactive material.
  • kits containing an antibody of the invention and instructions for use.
  • kits can also comprise an antibody of the invention conjugated to a detectable substance and instructions for use.
  • Still another aspect of the invention is a pharmaceutical composition comprising an antibody of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition contains an antibody of the invention, a therapeutic moiety, and a pharmaceutically acceptable carrier.
  • FIGURE 1 depicts the cDNA sequence of human TANGO 339 (SEQ ID NO: 1) and the predicted amino acid sequence of human TANGO 339 (SEQ ID NO:3).
  • the open reading frame of SEQ ID NO:l extends from nucleotide 210 to nucleotide 1019 of SEQ ID NO:l (SEQ ID NO:2).
  • FIGURE 2 depicts a hydropathy plot of human TANGO 339. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line.
  • the cysteine residues (cys) and N-glycosylation site (Ngly) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 42 of SEQ ID NO:3; SEQ ID NO:5) on the left from the mature protein (amino acids 43 to 270 of SEQ ID NO:3; SEQ ID NO:4) on the right.
  • FIGURE 3 depicts an alignment of the amino acid sequence of human CD9 antigen (SEQ ID NO:24; Accession Number NM 001769) and the amino acid sequence of human TANGO 339 (SEQ ID NO:3).
  • the amino acid sequences of human CD9 antigen and human TANGO 339 are 24.1% identical. This alignment was performed using the ALIGN alignment program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4.
  • FIGURE 4 depicts an alignment of the nucleotide sequence of the coding region of human CD9 antigen (SEQ ID NO:25; Accession Number NM__001769) and the nucleotide sequence of the coding region of human TANGO 339 (SEQ ID NO:l).
  • the nucleotide sequences of the coding regions of human CD9 antigen and human TANGO 339 are 45.9% identical.
  • This alignment was performed using the ALIGN alignment program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4.
  • FIGURE 5 depicts the cDNA sequence of human TANGO 353 (SEQ ID NO:27) and the predicted amino acid sequence of human TANGO 353 (SEQ ID NO:29).
  • the open reading frame of human TANGO 353 extends from nucleotide 76 to nucleotide 765 of SEQ ID NO:27 (SEQ ID NO:28).
  • FIGURE 6 depicts a hydropathy plot of human TANGO 353. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and potential N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 14 of SEQ ID NO:29; SEQ ID NO:31) on the left from the mature protein (amino acids 15 to 230 of SEQ ID NO:29; SEQ ID NO:30) on the right.
  • FIGURE 7 depicts a cDNA sequence of human TANGO 358 (SEQ ID NO:36) and the predicted amino acid sequence of human TANGO 358 (SEQ ID NO:38).
  • the open reading frame of human TANGO 358 extends from nucleotide 184 to 429 of SEQ ID NO:36 (SEQ ID NO:37).
  • FIGURE 8 depicts a hydropathy plot of human TANGO 358. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 42 of SEQ ID NO:36; SEQ ID NO:40) on the left from the mature protein (amino acids 43 to 82 of SEQ ID NO:36; SEQ ID NO:39) on the right.
  • FIGURE 9 depicts the cDNA sequence of human TANGO 365 (SEQ ID NO:44) and the predicted amino acid sequence of human TANGO 365 (SEQ ID NO:46).
  • the open reading frame of SEQ ID NO:44 extends from nucleotide 56 to nucleotide 550 (SEQ ID NO:45).
  • FIGURE 10 depicts a hydropathy plot of human TANGO 365. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (Cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 36 of SEQ ID NO:46; SEQ ID NO:47) preceding the mature protein (amino acids 37 to 165 of SEQ ID NO:46; SEQ ID NO:48) on the right.
  • FIGURE 11 depicts the cDNA sequence of human TANGO 368 (SEQ ID NO: 52) and the predicted amino acid sequence of TANGO 368 (SEQ ID NO: 54).
  • FIGURE 12 depicts a hydropathy plot of human TANGO 368. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (Cys) and potential N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace.
  • FIGURE 13 depicts a local alignment of the nucleotide sequence of full-length human TANGO 368 (SEQ ID NO:52) and a fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57).
  • the nucleotide sequence of human TANGO 368 and the human T-cell receptor gamma VI gene region are 99.3 % identical for a 973 bp overlap. This alignment was performed using the LALIGN program with a PAM120 scoring matrix, a gap length penalty of 12 and a gap penalty of 4.
  • FIGURE 14 depicts a cDNA sequence of human TANGO 369 (SEQ ID NO: 58) and the predicted amino acid sequence of human TANGO 369 (SEQ ID NO:60).
  • the open reading frame of human TANGO 369 extends from nucleotide 162 to 335 of SEQ ID NO:58 (SEQ ED NO:61).
  • FIGURE 15 depicts a hydropathy plot of human TANGO 369. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:60; SEQ ID NO:62) on the left from the mature protein (amino acids 27 to 58 of SEQ ID NO:60; SEQ ID NO:61) on the right.
  • FIGURE 16 depicts the cDNA sequence of human TANGO 383 (SEQ ID NO:63) and the predicted amino acid sequence of human TANGO 383 (SEQ ID NO:65).
  • the open reading frame of SEQ ED NO:63 extends from nucleotide 104 to nucleotide 523 (SEQ ID NO:64).
  • FIGURE 17 depicts a hydropathy plot of human TANGO 383. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 20 of SEQ ID NO: 65; SEQ ID NO:66) on the left from the mature protein (amino acids 21 to 140 of SEQ ID NO:65; SEQ ID NO:67) on the right.
  • FIGURE 18 depicts an alignment of the amino acid sequence of TANGO 383
  • FIGURE 19 depicts the cDNA sequence of human TANGO 393 (SEQ ID NO:73) and the predicted amino acid sequence of human TANGO 393 (SEQ ID NO:75).
  • the open reading frame of SEQ ID NO: 73 extends from nucleotide 40 to nucleotide 1458 (SEQ ID NO:74).
  • FIGURE 20 depicts a hydropathy plot of human TANGO 393. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line.
  • the cysteine residues (cys) and N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:75; SEQ ID NO:76) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:75; SEQ ID NO:77) on the right.
  • FIGURE 21 depicts the cDNA sequence of mouse TANGO 393 (SEQ ID NO:93) and the predicted amino acid sequence of mouse TANGO 393 (SEQ ID NO:95).
  • the open reading frame of SEQ ID NO:93 extends from nucleotide 226 to nucleotide 1644 (SEQ ID NO:94).
  • FIGURE 22 depicts a hydropathy plot of mouse TANGO 393. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line.
  • the cysteine residues (cys) and N-glycosylation sites (N-Gly) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:95; SEQ ID NO:96) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:95; SEQ ID NO:97) on the right.
  • FIGURE 23 depicts an alignment of the open reading frames of human TANGO 393 (SEQ ID NO:74) and mouse TANGO 393 (SEQ ID NO:94) demonstrating an identity of 82.8%.
  • the algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989)
  • FIGURE 24 depicts an alignment of the immature proteins of human TANGO 393 (SEQ ID NO:75) and mouse TANGO 393 (SEQ ID NO:95) demonstrating an identity of 89.2%.
  • the algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences.
  • FIGURE 25 depicts the cDNA sequence of human TANGO 402 (SEQ ED NO : 110) and the predicted amino acid sequence of human TANGO 402 (SEQ ID NO: 112).
  • the open reading frame of human TANGO 402 extends from nucleotide 87 to nucleotide 707 of SEQ ID NO: 110 (SEQ ID NO: 111).
  • FIGURE 26 depicts a hydropathy plot of human TANGO 402. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and N-glycosylation (Ngly) sites are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 50 of SEQ ID NO: 112; SEQ ID NO: 114) on the left from the mature protein (amino acids 51 to 207 of SEQ ED NO:112; SEQ ID NO:113) on the right.
  • FIGURE 27 depicts an alignment of the amino acid sequence of human TANGO 402 (SEQ ID NO:l 12) and the amino acid sequence of human LOX-1 (SEQ ID NO: 122; Accession Number AB010710).
  • the alignment demonstrates that the amino acid sequences of human TANGO 402 and human LOX-1 are 25.1% identical. This alignment was performed using the ALIGN program with a PAM120 scoring matrix, a gap length penalty of 12 and a gap penalty of 4.
  • FIGURE 28 depicts an alignment of the nucleotide sequences of the open reading frames of human TANGO 402 (SEQ ID NO: 111) and human LOX-1 (SEQ ID NO: 121 ; Accession Number AB010710).
  • the alignment of the open reading frame of human TANGO 402 and that of human LOX-1 demonstrates that those two coding regions are 42.0 % identical.
  • An alignment demonstrates that the nucleotide sequences of the cDNA of human TANGO 402 and human LOX-1 are 44.0 % identical.
  • the alignments were performed using the ALIGN program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4.
  • FIGURE 29 depicts the cDNA sequence of human MANGO 346 (SEQ ED
  • SEQ ID NO: 123 and the predicted amino acid sequence of human MANGO 346 (SEQ ID NO: 125).
  • the open reading frame of SEQ ID NO: 123 extends from nucleotide 319 to nucleotide 498 (SEQ ID NO: 124).
  • FIGURE 30 depicts a hydropathy plot of human MANGO 346. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 19 of SEQ ID NO: 125; SEQ ID NO: 126) on the left from the mature protein (amino acids 20 to 60 of SEQ ID NO: 125; SEQ ID NO: 127) on the right.
  • FIGURE 31 depicts the cDNA sequence of human MANGO 349 (SEQ ID NO: 129).
  • SEQ ID NO: 1278 and the predicted amino acid sequence of human MANGO 349 (SEQ ID NO: 130).
  • the open reading frame of SEQ ED NO:128 extends from nucleotide 221 to nucleotide 721 (SEQ ED NO: 129).
  • FIGURE 32 depicts a hydropathy plot of human MANGO 349. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:130; SEQ ID NO:131) on the left from the mature protein (amino acids 27 to 167 of SEQ ID NO: 130; SEQ ID NO: 132) on the right.
  • proteins and nucleic acid molecules comprise families of molecules having certain conserved structural and functional features.
  • family or “families” are intended to mean two or more proteins or nucleic acid molecules having a common structural domain and having sufficient amino acid or nucleotide sequence identity as defined herein.
  • Family members can be from either the same or different species.
  • a family can comprises two or more proteins of human origin, or can comprise one or more proteins of human origin and one or more of non-human origin. Members of the same family may also have common structural domains.
  • TANGO 339 proteins, TANGO 353 proteins, TANGO 358 proteins, TANGO 365 proteins, TANGO 368 proteins, TANGO 369 proteins, TANGO 383 proteins, TANGO 393 proteins, TANGO 402 proteins, MANGO 346 proteins and MANGO 349 proteins of the invention can have signal sequences.
  • a "signal sequence" includes a peptide of at least about 15 or 20 amino acid residues in length which occurs at the N-terminus of secretory and membrane-bound proteins and which contains at least about 70% hydrophobic amino acid residues such as alanine, leucine, isoleucine, phenylalanine, proline, tyrosine, tryptophan, or valine.
  • a signal sequence contains at least about 10 to 40 amino acid residues, preferably about 19-34 amino acid residues, and has at least about 60-80%, more preferably 65-75%, and more preferably at least about 70% hydrophobic residues.
  • a signal sequence serves to direct a protein containing such a sequence to a lipid bilayer.
  • a TANGO 339 protein contains a signal sequence of about amino acids 1 to 42 of SEQ ID NO:3 (SEQ ID NO:5).
  • a TANGO 353 protein contains a signal sequence of about amino acids 1 to 14 of SEQ ID NO:37 (SEQ ID NO:31).
  • a TANGO 358 protein contains a signal sequence at about amino acids 1 to 42 of SEQ ID NO:38 (SEQ ID NO:40).
  • a TANGO 365 protein contains a signal sequence of about amino acids 1 to 36 of SEQ ID NO:46 (SEQ ID NO:47).
  • a TANGO 368 protein contains a signal sequence of about amino acids 1 to 27 of SEQ ID NO:54 (SEQ ID NO:56).
  • a TANGO 369 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ED NO:60 (SEQ ED NO:62).
  • a TANGO 383 protein contains a signal sequence of about amino acids 1 to 20 of SEQ ID NO:65 (SEQ ED NO:66).
  • human TANGO 393 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ID NO:75 (SEQ ID NO:76).
  • mouse TANGO 393 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ED NO:95 (SEQ ED NO:96).
  • a TANGO 402 protein contains a signal sequence of about amino acids 1 to 50 of SEQ ID NO: 112 (SEQ ED NO: 114).
  • a MANGO 346 protein contains a signal sequence of about amino acids 1 to 19 of SEQ ED NO:125 (SEQ ID NO:126).
  • a MANGO 349 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ID NO: 130 (SEQ ID NO: 131).
  • the signal sequence is usually cleaved during processing of the mature protein. In the case of, e.g. , transmembrane 4-type proteins, the signal peptide is generally not cleaved, but becomes a transmembrane-anchoring domain of the polypeptide.
  • a TANGO 339 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • a TANGO 339 protein contains extracellular domains at about amino acid residues 43 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO:20 and SEQ ID NO:
  • transmembrane domains at about amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO:15, SEQ ID NO:16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at about amino acid residues 85 to 92 and 255 to 270 of SEQ ID NO:3 (SEQ ID NO:22 and SEQ ID NO:23, respectively).
  • the mature TANGO 339 protein co ⁇ esponds to amino acids 43 to 270 of SEQ ED NO:3 (SEQ ED NO:4).
  • a TANGO 339 protein contains extracellular domains at about amino acid residues 1 to 16, 85 to 92, and 255 to 270 of SEQ ID NO:3 (SEQ ED NO:l 1, SEQ ED NO: 12, and SEQ ED NO: 13, respectively), transmembrane domains at about amino acid residues 17 to 41, 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ED NO:14, SEQ ID NO:15, SEQ ID NO:16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at about amino acid residues 42 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO: 18 and SEQ ID NO: 19, respectively).
  • the mature TANGO 339 protein co ⁇ esponds to amino acids 1 to 270 of SEQ ID NO:3.
  • a TANGO 339 family member can include a signal sequence.
  • a TANGO 339 family member has the amino acid sequence of SEQ ID NO: 1
  • the signal sequence is located at amino acids 1 to 40, 1 to 41, 1 to 42, 1 to 43 or 1 to 44.
  • the domains and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 40 results in an extracellular domain consisting of amino acids 41 to 61 of SEQ ID NO: 3 and the mature TANGO 339 protein co ⁇ esponding to amino 41 to 270.
  • a TANGO 339 family member can include one or more transmembrane 4 or transmembrane 4-like domains.
  • a transmembrane 4 domain typically has the following consensus sequence: G-xxx-[LIVMF]-xx-[GSA]-[LIVMF][LIVMF]-G-C-x-[GA]-[STA]- xx-[EG]-xx-[CWN]-[LIVM][LIVM], wherein G is a glycine residue, "x" is any amino acid, [L1VMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue.
  • a transmembrane 4 domain is a characteristic of transmembrane 4 superfamily members which include, for example, CD9 antigen, CD37, CD53, CD63, CD81, and CD82.
  • Transmembrane 4 proteins have the following characteristics: they are type III membrane proteins, which contain an N-terminal membrane-anchoring domain that is not cleaved during biosynthesis and that functions both as a translocation signal and as a membrane anchor; they contain a total of four transmembrane domains and at least seven conserved cysteine residues; and they are approximately 218 to 284 amino acid residues.
  • a transmembrane 4-like domain as described herein can have the following consensus sequence: G-xxx-[LIVMF]-xx-[GSA]-[LIVMF]-x-G-C-x-[GA]-[STA]-xx- [EG]-xx-[CWN]-[LIVM][LEVM], wherein G is a glycine residue, "x" is any amino acid, [LIVMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue.
  • a TANGO 339 family member has the amino acid sequence of SEQ ID NO:3 and, preferably, a transmembrane 4 domain-like consensus sequence is located at about amino acid positions 69 to 91 of SEQ ED NO:3 (SEQ ED NO:7).
  • a TANGO 339 family member has the amino acid sequence of SEQ ID NO: 3 and, preferably, a transmembrane 4-like domain is located at about amino acid positions 68 to 260 of SEQ ED NO:3 (SEQ ID NO:6).
  • a TANGO 339 family member includes one or more transmembrane 4-like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 69 to 91 of SEQ ID NO:3 (SEQ ID NO:7).
  • a TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ID NO:6).
  • a TANGO 339 family member includes one or more transmembrane 4-like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 69 to 91 of SEQ ID NO:3 (SEQ ID NO:7), and has at least one TANGO 339 biological activity as described herein.
  • a TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ED NO:6), and has at least one TANGO 339 biological activity as described herein.
  • the transmembrane 4-like domain of TANGO 339 is a transmembrane 4 domain, which has the following consensus sequence: G-xxx-[LEVMF]- xx-[GSA]-[LIVMF][LIVMF]-G-C-x-[GA]-[STA]-xx-[EG]-xx-[CWN]-[LIVM][LIVM], wherein G is a glycine residue, "x" is any amino acid, [LIVMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue.
  • G is a glycine residue
  • "x" is any amino acid
  • TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ID NO:6).
  • a TANGO 339 family member includes one or more peripherin/rom-1 or peripherin/rom-1 -like domains.
  • a peripherin/rom-1 domain typically has the following consensus sequence: D-G-V-P-F-S-C-C-N-P-x-S-P-R-P-C, wherein D is an aspartic acid residue, G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, S is a serine residue, C is a cysteine residue, N is an asparagine residue, x is any amino acid, and R is an arginine residue.
  • Peripherin/rom-1 domains are characteristic of retinal-specific integral membrane proteins that are located at the rims of the photoreceptor disks and that function in disk morphogenesis.
  • Peripherin (or RDS) and rom-1 are examples of proteins that contain the peripherin/rom-1 domain. Defects in the peripherin gene have been shown to cause various diseases, including autosomal dominant retinitis pigmentosa, autosomal dominant punctata albescens, and butterfly-shaped pigment dystrophy.
  • a peripherin/rom-1 -like domain as described herein has the following consensus sequence: G-V-P-F-S-C-C-x-P, wherein G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, and C is a cysteine residue.
  • a TANGO 339 family member has the amino acid sequence of SEQ ID NO:3 and, preferably, a peripherin/rom-1 -like domain consensus sequence is located at about amino acid positions 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9).
  • a TANGO 339 family member has the amino acid sequence of SEQ ID NO:31 and, preferably, a peripherin/rom-1 -like domain is located at about amino acid positions 18 to 270 of SEQ ED NO:3 (SEQ ID NO: 8).
  • a TANGO 339 family member includes one or more peripherin/rom-1 -like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9).
  • a TANGO 339 family member includes one or more peripherin/rom-1 -like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 15%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 18 to 270 of SEQ ID NO:3 (SEQ ID NO:8).
  • a TANGO 339 family member includes one or more peripherin/rom-1 -like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%), more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9), and has at least one TANGO 339 biological activity as described herein.
  • a TANGO 339 family member includes one or more peripherin/rom-1 -like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 18 to 270 of SEQ ID NO:3 (SEQ ED NO:8), and has at least one TANGO 339 biological activity as described herein.
  • the peripherin/rom-1 -like domain of TANGO 339 is a peripherin/rom-1 domain, which has the following consensus sequence: D-G-V-P-F-S-C- C-N-P-x-S-P-R-P-C, wherein D is an aspartic acid residue, G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, S is a serine residue, C is a cysteine residue, N is an asparagine residue, x is any amino acid, and R is an arginine residue.
  • a TANGO 339 family member includes one or more peripherin/rom-1 -like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 18 to 270 of SEQ ID NO:3 (SEQ ID NO:8).
  • a TANGO 353 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • an TANGO 353 protein contains an extracellular domain of about amino acids 1 to 116 of SEQ ID NO:29, or a mature extracellular domain of about amino acids 15 to 116 of SEQ ID NO:29 (SEQ ID NO:32).
  • a TANGO 353 protein contains a transmembrane domain of about amino acids 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33).
  • a TANGO 353 protein contains a cytoplasmic domain of about amino acids 142 to 230 of SEQ ID NO:29 (SEQ ED NO:34).
  • a TANGO 353 protein is a mature protein containing an extracellular, transmembrane and cytoplasmic domain of about amino acids 15 to 230 of SEQ ID NO:29 (SEQ ID NO:30).
  • a TANGO 353 family member can include a signal sequence.
  • a TANGO 353 family member has the amino acid sequence of SEQ ID NO:29, and the signal sequence is located at amino acids 1 to 12, 1 to 13, 1 to 14, 1 to 15 or 1 to 16.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 12 results in an extracellular domain consisting of amino acids 13 to 116 of SEQ ID NO:29 and the mature TANGO 353 protein co ⁇ esponding to amino 13 to 230.
  • a TANGO 358 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • a TANGO 358 protein contains an extracellular domain at amino acids 1 to about 49 of SEQ ID NO:38 or a mature extracellular domain at about amino acid residues 43 to 49 of SEQ ID NO:38 (SEQ ED NO:41), a transmembrane domain at about amino acid residues 50 to 66 of SEQ ID NO:38 (SEQ ID NO:42), and a cytoplasmic domain at about amino acid residues 67 to 82 of SEQ ID NO:38 (SEQ ID NO:43).
  • a TANGO 358 family member can include a signal sequence.
  • a TANGO 358 family member has the amino acid sequence of SEQ ID NO:38, and the signal sequence is located at amino acids 1 to 40, 1 to 41, 1 to 42, 1 to 43 or 1 to 44.
  • the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 40 results in an extracellular domain consisting of amino acids 41 to 50 of SEQ ID NO:38 and the mature TANGO 368 protein co ⁇ esponding to amino 41 to 82.
  • a TANGO 365 family member can include a signal sequence.
  • a TANGO 365 family member has the amino acid sequence of SEQ ED NO:46, and the signal sequence is located at amino acids 1 to 34, 1 to 35, 1 to 36, 1 to 37 or 1 to 38.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 36 results in a mature TANGO 365 protein co ⁇ esponding to amino 37 to 165 of SEQ ID NO:46 (SEQ ID NO:47).
  • a TANGO 365 family member can include one or more of the following domains: (1) an extracellular domain; (2) two transmembrane domains; and (3) a cytoplasmic domain.
  • a TANGO 365 protein contains an extracellular domain of about amino acids 95 to 165 of SEQ ED NO:46 (SEQ ED NO:51), or a mature extracellular domain of about amino acids 30 to 246 of SEQ ID NO:46.
  • a TANGO 365 protein contains a first transmembrane domain of about amino acids 52 to 70 of SEQ ED NO:46 (SEQ ED NO:49).
  • an protein contains a cytoplasmic domain of about amino acids 71 to 77 of SEQ ID NO:46 (SEQ ID NO: 133).
  • a TANGO 365 protein contains a second transmembrane domain of about amino acids 78 to 94 of SEQ ID NO:46 (SEQ ED NO:50).
  • a TANGO 365 protein is a mature protein containing an extracellular domain, two transmembrane domains and a cytoplasmic domain of about amino acids 37 to 165 of SEQ ID NO:46 (SEQ ID NO:48).
  • a TANGO 368 family member can include a signal sequence.
  • a TANGO 368 family member has the amino acid sequence of SEQ ID NO:54, and the signal sequence is located at amino acids 1 to 25, 1 to 26, 1 to 27, 1 to 28 or 1 to 29.
  • the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 27 results in a mature TANGO 368 protein co ⁇ esponding to amino 28 to 59 of SEQ ID NO:54 (SEQ ED NO:55).
  • a TANGO 369 family member can include a signal sequence.
  • a TANGO 369 family member has the amino acid sequence of SEQ ED NO:60, and the signal sequence is located at amino acids 1 to 24, 1 to 25, 1 to 26, 1 to 27 or 1 to 28.
  • the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 26 results in a mature TANGO 368 protein co ⁇ esponding to amino 27 to 58 of SEQ ID NO:60 (SEQ ED NO:61).
  • a TANGO 383 family member can include a signal sequence.
  • a TANGO 383 family member has the amino acid sequence of SEQ ED NO:65, and the signal sequence is located at amino acids 1 to 18, 1 to 19, 1 to 20, or 1 to 21.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 20 results in a mature TANGO 383 protein co ⁇ esponding to amino 21 to 140 of SEQ ID NO:65.
  • a TANGO 383 family member can include one or more of the following domains: (1) an extracellular domain; (2) two transmembrane domains; and (3) a cytoplasmic domain.
  • a TANGO 383 protein contains a cytoplasmic domain of about amino acids 21 to 49 of SEQ ID NO:65.
  • a TANGO 383 protein contains a first transmembrane domain of about amino acids 50 to 70 of SEQ ID NO:65 (SEQ ID NO:68).
  • a TANGO 383 protein contains an extracellular domain of about amino acids 71 to 115 of SEQ ID NO:65 (SEQ ED NO:70).
  • a TANGO 383 protein contains a second transmembrane domain of about amino acids 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69).
  • a TANGO 383 protein is a mature protein containing an extracellular domain, two transmembrane domains and a cytoplasmic domain of about amino acids 21 to 140 of SEQ ED NO:65 (SEQ ED NO:67).
  • a TANGO 393 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain; and (4) a leucine-rich domain.
  • a TANGO 393 protein contains an extracellular domain at amino acids 1 to about 447 of SEQ ID NO: 75 or a mature extracellular domain at about amino acid residues 27 to 447 of SEQ ED NO: 75 (SEQ ED NO:89), a transmembrane domain at about amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a cytoplasmic domain at about amino acid residues 468 to 473 of SEQ ID NO:75 (SEQ ID NO: 134).
  • a TANGO 393 family member contains an extracellular domain at amino acids 1 to about 26 of SEQ ID NO:95 or a mature extracellular domain at about amino acid residues 27 to 449 of SEQ ID NO:95 (SEQ ID NO: 109), a transmembrane domain at about amino acid residues 450 to 467 of SEQ ID NO:95 (SEQ ID NO:98), and a cytoplasmic domain at about amino acid residues 468 to 473 of SEQ ID NO:95 (SEQ ID NO:35).
  • a TANGO 393 family member can include one or more leucine-rich-repeat (LRR) domains.
  • LRR leucine-rich-repeat
  • a leucine-rich-repeat domain typically has the following degenerate consensus sequence: x-L-x-x-L-x-L-x-x-[NCT]-x-L-x-x-x-L-x-x-x-x-L-x-x-L, wherein L is a leucine residue and can be replaced by any aliphatic residue,"x" is any amino acid, and [NCT] is either an asparagine, cysteine or threonine, respectively.
  • Leucine-rich-repeat domains most frequently appear in tandem repeats.
  • leucine-rich-repeat domains are characteristic of a diverse set of signaling proteins that are involved in cell signaling, cell growth and cell differentiation. Defects in leucine-rich-repeat genes have been shown to cause various diseases which include but are not limited to Bernard-Soulier disease, a bleeding disorder. Furthermore, leucine-rich-repeat genes are involved in the pathogenesis of diseases, for example, the leucine-rich-repeat of type- 1 human immunodeficiency virus Rev protein is the trans-activating region of the virus (Kobe and Deisenhofer, 1994, TIBS, 19:415-421).
  • a TANGO 393 family member has the amino acid sequence of SEQ ED NO:76 and, preferably, a leucine-rich-repeat domain consensus sequence is located at about amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 (SEQ ID NO:76), SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively.
  • a TANGO 393 family member has the amino acid sequence of SEQ ID NO:95 and, preferably, a leucine-rich-repeat domain is located at about amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively.
  • a TANGO 393 family member includes one or more leucine-rich-repeat domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 of SEQ ID NO:76 (SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively).
  • a TANGO 393 family member includes one or more leucine-rich-repeat domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively.
  • a TANGO 393 family member includes one or more leucine-rich-repeat domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 of SEQ ID NO:76 (SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively), and has at least one TANGO 393 biological activity as described herein.
  • a TANGO 393 family member includes one or more leucine-rich-repeat domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively, and has at least one TANGO 393 biological activity as described herein.
  • a TANGO 402 family member can include of one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • a TANGO 402 protein contains an extracellular domain at amino acids 1 to about 133 of SEQ ID NO:l 12 or a mature extracellular domain at about amino acid residues 51 to 133 of SEQ ED NO: 112 (SEQ ID NO:l 15), a transmembrane domain at about amino acid residues 134 to 151 of SEQ ED NO:112 (SEQ ED NO: 116), and a cytoplasmic domain at about amino acid residues 152 to 207 of SEQ ED NO:l 12 (SEQ ID NO: 117).
  • a TANGO 402 family member can include a signal sequence.
  • a TANGO 402 family member has the amino acid sequence of SEQ ID NO:l 12, and the signal sequence is located at amino acids 1 to 48, 1 to 49, 1 to 50, 1 to 51 or 1 to 52.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 48 results in an extracellular domain consisting of amino acids 49 to 133 of SEQ ID NO:l 12 and the mature TANGO 402 protein co ⁇ esponding to amino 49 to 207 of SEQ ID NO:l 12.
  • a TANGO 402 family member can include a C-type lectin domain or a C-type lectin-like domain.
  • a C-type lectin domain typically has the following consensus sequence: C- [LIVMFATG]-x(5,12)-[WL]-x-[DNSR]-x(2)-C-x(5,6)-[FYWLIVSTA]-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, x is any amino acid and the number in parentheses indicates the number of amino acids, [WL] is either a tryptophan or leucine residue, [DNSR] is a aspartic acid, asparagine, serine or arginine residue, [FYWLEVSTA] is a phenylalanine, tyrosine, tryptophan, leucine, isoleucine, valine, serine, threonine or alanine residue, and [LIVSTA] is
  • C-type lectin domains contain four cysteines, which are involved in two disulfide bonds, and are about 110 to 130 amino acid residues.
  • C-type lectin domains typically function as calcium-dependent carbohydrate-recognition domains and have been found in various proteins including, but not limited to, asialoglycoprotein receptors (ASGPR), pulmonary surfactant-associated protein A (SP-A), mannan-binding proteins, L-selectin, neurocan, and tetranectin. These proteins have various functions including, for example, cell adhesion (i.e., L-selectin).
  • ASGPR mediates the endocytosis of plasma glycoprotein to which the terminal salic acid-residue in their carbohydrated moieties has been removed.
  • SP-A binds to surfactant phospholipids and contributes to lower the surface tension at the air-liquid interface in the alveoli of the lung.
  • a C-type lectin-like domain as described herein has the following consensus sequence: C-[LIVMFATG]-x-(5,12)-[DNSR]-x(2)-C-x(5,6)-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, "x" is any amino acid and the number in parentheses indicates the number of amino acids [DNSR] is an aspartic acid, asparagine, serine or arginine residue, and [LIVSTA] is a leucine, isoleucine, valine, serine, threonine or alanine residue.
  • a TANGO 402 family member has the amino acid sequence of SEQ ID NO: 112 and, preferably, a C-type lectin-like domain is located at about amino acid positions 104 to 193 of SEQ ID NO:l 12 (SEQ ID NO:l 18), wherein the consensus sequence is at about amino acid positions 172 to 193 of SEQ ID NO: 112 (SEQ ID NO: 119).
  • a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118).
  • a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118), and has at least one TANGO 402 biological activity as described herein.
  • a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118) and includes a cysteine residue N-terminal to the consensus sequence.
  • a TANGO 402 family member includes one or more C-type lectin-like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO:l 12 (SEQ ID NO:l 18), includes a cysteine residue N-terminal to the consensus sequence, and has at least one TANGO 402 biological activity as described herein.
  • the C-type lectin-like domain of TANGO 402 is a C-type lectin domain, which has the following consensus sequence: C-[LIVMFATG]-x(5,12)- [WL]-x-[DNSR]-x(2)-C-x(5,6)-[FYWLEVSTA]-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, x is any amino acid, [WL] is either a tryptophan or leucine residue, [DNSR] is a aspartic acid, asparagine, serine or arginine residue, [FYWLIVSTA] is a phenylalanine, tyrosine, tryptophan, leucine, isoleucine, valine, serine, threonine or alanine
  • a TANGO 402 family member includes one or more C-type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118).
  • a MANGO 346 family member can include a signal sequence.
  • a MANGO 346 family member has the amino acid sequence of SEQ ID NO:125, and the signal sequence is located at amino acids 1 to 17, 1 to 18, 1 to 19, 1 to 20 or 1 to 21.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 19 results in the mature MANGO 346 protein co ⁇ esponding to amino 20 to 60 (SEQ ID NO: 127).
  • a MANGO 349 family member can include a signal sequence.
  • a MANGO 349 family member has the amino acid sequence of SEQ ID NO: 130, and the signal sequence is located at amino acids 1 to 24, 1 to 25, 1 to 26, 1 to 27 or 1 to 28.
  • the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein.
  • the cleavage of a signal sequence consisting of amino acids 1 to 26 results in the mature MANGO 349 protein co ⁇ esponding to amino 27 to 167 of SEQ ID NO: 130 (SEQ ID NO: 132).
  • TANGO 339 Various features of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 are summarized below.
  • a cDNA encoding human TANGO 339 was identified by analyzing the sequences of clones present in a human fetal library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthgalOOgOl, encoding full-length human TANGO 339.
  • the human TANGO 339 cDNA of this clone is 2715 nucleotides long ( Figure 1; SEQ ID NO:l).
  • Figure 2 depicts a hydropathy plot of human TANGO 339. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line.
  • the cysteine residues are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 339 includes a 42 amino acid signal peptide (amino acid 1 to amino acid 42 of SEQ ID NO:3; SEQ ID NO: 5) preceding the mature human TANGO 339 protein (co ⁇ esponding to amino acid 43 to amino acid 270 of SEQ ID NO:3; SEQ ID NO:4).
  • the molecular weight of human TANGO 339 protein without post-translational modifications is 30.7 kDa prior to the cleavage of the signal peptide, and 25.6 kDa after cleavage of the signal peptide.
  • Human TANGO 339 protein is a transmembrane protein that contains extracellular domains at amino acid residues 43 to 61 and 116 to 232 of SEQ ED NO:3 (SEQ ED NO:20 and SEQ ID NO:21, respectively), transmembrane domains at amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 15, SEQ ED NO: 16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at amino acid residues 85 to 92 and 255 to 270 of SEQ ID NO:3 (SEQ ID NO:22 and SEQ ID NO:23, respectively).
  • human TANGO 339 has extracellular domains at amino acid residues 1 to 16, 85 to 92, and 255 to 270 of SEQ ID NO:3 (SEQ ID NO: 11, SEQ ED NO: 12, and SEQ ED NO: 13, respectively), transmembrane domains at amino acid residues 17 to 41, 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively), and cytoplasmic domains of amino acid residues 42 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO: 18 and SEQ ID NO: 19, respectively).
  • a human TANGO 339 protein contains cytoplasmic domains at amino acid residues 43 to 61 and 116 to 232 of SEQ ID NO: 3, transmembrane domains at amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 15, SEQ ID NO: 16 and SEQ ID NO: 17, respectively), and extracellular domains at amino acid residues 85 to 92 and 255 to 270 of SEQ ED NO:3.
  • nucleotide sequence of human TANGO 339 the nucleotide at position 29 is adenine (A)(SEQ ED NO:2).
  • amino acid at position 10 is lysine (K)(SEQ ED NO:3).
  • a species variant of human TANGO 339 has a nucleotide at position 29 which is guanine (G)(SEQ ID NO: 136).
  • amino acid at position 10 is arginine (R)(SEQ ID NO:137), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 339 is thymine (T)(SEQ ED NO:2).
  • the amino acid at position 20 is phenylalanine (F)(SEQ ID NO:3).
  • a species variant of human TANGO 339 has a nucleotide at position 59 which is adenine (A)(SEQ ID NO: 138).
  • the amino acid at position 20 is tyrosine (Y)(SEQ ID NO:139), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 339 is cytosine (C)(SEQ ID NO:2).
  • amino acid at position 40 is alanine (A)(SEQ ID NO:3).
  • a species variant of human TANGO 339 has a nucleotide at position 119 which is thymine (T)(SEQ ID NO: 140).
  • the amino acid at position 40 is valine (V)(SEQ ID NO: 141), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 339 the nucleotide at position 180 is cytosine (C)(SEQ ID NO:2).
  • amino acid at position 60 is aspartate (D)(SEQ ID NO:3).
  • a species variant of human TANGO 339 has a nucleotide at position 180 which is guanine (G)(SEQ ID NO: 142).
  • the amino acid at position 60 is glutamate (E)(SEQ ID NO: 143), i.e., a conservative substitution.
  • Human TANGO 339 includes a transmembrane 4-like domain (at amino acids 68 to 260 of SEQ ID NO:3; SEQ ID NO:6) and a peripherin /rom-1-like domain (at amino acids 18 to 270 of SEQ ID NO:3; SEQ ID NO:8).
  • Human TANGO 339 has an N-glycosylation site with the sequence NCSG (at amino acid residues 169 to 172 of SEQ ED NO:3). Two protein kinase C phosphorylation sites are present in human TANGO 339. The first has the sequence SEK (at amino acid residues 42 to 44 of SEQ ED NO:3) and the second has the sequence SYR (at amino acid residues 133 to 135 of SEQ ED NO:3). Human TANGO 339 has three casein kinase EE phosphorylation sites.
  • the first has the sequence SYRD (at amino acid residues 133 to 136 of SEQ ID NO:3), the second has the sequence SKWD (at amino acid residues 210 to 213 of SEQ ID NO:3), and the third has the sequence SDIE (at amino acid residues 259 to 262 of SEQ ED NO:3).
  • SYRD amino acid residues 133 to 136 of SEQ ID NO:3
  • SKWD amino acid residues 210 to 213 of SEQ ID NO:3
  • SDIE at amino acid residues 259 to 262 of SEQ ED NO:
  • the first has the sequence GCVGAL (at amino acid residues 79 to 84 of SEQ ID NO:3)
  • the second has the sequence GASYSR (at amino acid residues 172 to 177 of SEQ ID NO:3)
  • the third has the sequence GVPFSC (at amino acid residues 181 to 186 of SEQ ED NO:3)
  • the fourth has the sequence GCEQAL (at amino acid residues 220 to 225 of SEQ ED NO:3)
  • the fifth has the sequence GVFIAI (at amino acid residues 238 to 243 of SEQ ID NO:3)
  • the sixth has the sequence GIFLAR (at amino acid residues 250 to 255 of SEQ ID NO:3).
  • Human TANGO 339 has a prokaryotic membrane lipoprotein lipid attachment site with the sequence VVMFTLGFAGC (at amino acid residues 70 to 80 of SEQ ID NO:3; SEQ ID NO:10).
  • the human TANGO 339 gene maps to human chromosome 10 between markers D10S201 and D10S551.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be used to diagnose disorders associated with G protein coupled receptors and/or modulate G protein coupled receptor-related processes, e.g., retinal processes and/or pulmonary-related processes.
  • Figure 3 shows an alignment of the human TANGO 339 amino acid sequence (SEQ ID NO:3) with the human CD9 antigen amino acid sequence (SEQ ID NO:24; Accession Number NM_001769).
  • the alignment shows that there is a 24.1% overall amino acid sequence identity between human TANGO 339 and human CD9 antigen.
  • the CD9 antigen is a widely expressed cell surface glycoprotein that has been shown to be involved in such processes as cell activation, proliferation, and adhesion.
  • CD9 antigen expression on platelets mediates platelet activation and aggregation.
  • CD9 antigen has also been shown to be expressed by neural cells and can play a role in intercellular signaling in the nervous system, in particular, controlling cellular attraction or repulsion in guiding neural growth to target points.
  • CD9 antigen has been shown to associate with beta 1 integrins and other transmembrane 4 superfamily members, including CD81 and CD82.
  • TANGO 339 proteins nucleic acids and modulators thereof could be useful in modulating cellular interaction such as between immune cells, and also can be involved in modulating intercellular signaling, such as neural cell intercellular signaling.
  • Figure 4 shows an alignment of the nucleotide sequence of human CD9 antigen coding region (SEQ ED NO:25; Accession Number NM_001769) and the nucleotide sequence of human TANGO 339 coding region (SEQ ID NO:2). The alignment shows a 45.9 % overall sequence identity between the two nucleotide sequences.
  • the full-length human CD9 antigen nucleic acid sequence (SEQ ID NO:26; Accession Number NP_001760) and human TANGO 339 cDNA (SEQ ID NO:l) have an overall sequence identity of 30.3%.
  • Clone EpT339 which encodes human TANGO 339, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-292. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. ⁇ 112.
  • TANGO 339 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of cells, tissues and/or organs, e.g., the proliferation of tissues and cells of lymphoid origin and neural origin.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be used to treat immune related disorders, e.g., immunodeficiency disorders (e.g., HIV), viral disorders, cancers, autoimmune disorders, (e.g., arthritis and graft rejection) and inflammatory disorders (e.g. , bacterial or viral infection, psoriasis, septicemia, arthritis, allergic reactions).
  • TANGO 339 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the development of cells, tissues and/or organs in the embryo and/or fetus.
  • TANGO 339 has characteristics of transmembrane 4 proteins
  • nucleic acids, proteins and modulators thereof can be utilized to modulate (e.g., stabilize, promote, inhibit or disrupt) cellular activation, cellular proliferation, motility, and differentiation.
  • TANGO 339 compositions and modulators thereof can be used to modulate binding to extracellular matrix (ECM)- associated factors such as integrins and can function to modulate ligand binding to cell surface receptors.
  • ECM extracellular matrix
  • TANGO 339 nucleic acids, proteins and modulators thereof can be used to modulate disorders associated with abe ⁇ ant signal transduction in response to ECM- associated proteins and cell surface receptors such as other transmembrane 4 proteins.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development and progression of proliferative disorders, e.g., neoplasms or tumors
  • carcinomas, sarcomas, adenomas or myeloid lymphomas associated with cancer, (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leimyosarcoma, rhabdothehosarcoma, colon sarcoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hematoma,
  • acute lymphocytic leukemia and acute myelocytic leukemia myelolastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia
  • chronic leukemia chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia
  • polycythemia vera lymphoma
  • TANGO 339 proteins exhibit similarity to human CD9 antigen, a member of the transmembrane 4 superfamily.
  • nucleic acids, proteins and modulators thereof can be utilized to modulate platelet activation and aggregation.
  • antagonists to TANGO 339 action such as peptides, antibodies or small molecules that decrease or block TANGO 339 binding to extracellular matrix components (e.g., integrins) or that prevent TANGO 339 signaling, can be used as platelet activation and aggregation blockers.
  • agonists that mimic TANGO 339 activity such as peptides, antibodies or small molecules, can be used to induce platelet activation and aggregation.
  • Antibodies may activate or inhibit the cell adhesion, proliferation and activation, and may help in treating inflammation, cancer, cardiovascular disease or stroke by affecting these cellular processes.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate platelet-related processes and disorders, e.g., Glanzmann's thromboasthemia, which is a bleeding disorder characterized by failure of platelet aggregation in response to cell stimuli.
  • TANGO 339 proteins exhibit similarity to human CD9 antigen
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling in the nervous system.
  • the CD9 antigen which is expressed at the surface of central nervous system (CNS) mature myelin, may modulate intercellular signal transduction and enhance myelin membrane adhesion to extracellular matrices at very late stages of development, thereby playing a role in the maintenance of the entire myelin sheath.
  • CNS central nervous system
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development and function of the eye, such as retinal development and function, (e.g., photoreceptor disk morphogenesis).
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to treat eye diseases and/or disorders, e.g., autosomal dominant retinitis pigmentosa, autosomal dominant punctata albescens, butterfly-shaped pigment dystrophy, cataracts, macular degeneration, myopia, stigmatism and retinoblastoma.
  • TANGO 339 maps to a region of chromosome 10 which encodes polypeptides expressed in the lung
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development, differentiation and activity of pulmonary structures, e.g., lung.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate or treat pulmonary disorders, such as atelectasis, pulmonary congestion or edema, cystic fibrosis, chronic obstructive airway disease (e.g., emphysema, chronic bronchitis, bronchial asthma, and bronchiectasis), diffuse interstitial diseases (e.g., sarcoidosis, pneumoconiosis, hypersensitivity pneumonitis, Goodpasture's syndrome, idiopathic pulmonary hemosiderosis, pulmonary alveolar proteinosis, desquamative interstitial pneumonitis, chronic interstitial pneumonia, fibrosing alveolitis, hamman-rich syndrome, pulmonary eosinophilia, diffuse interstitial fibrosis, Wegener's granulomatosis, lymphomatoid granulomatosis, and lipid pneumonia), lung cancer or tumors (e.g.,
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in the development, differentiation and activity of the brain, including, but not limited to development, differentiation and activation of neuronal cells and glial cells (e.g., oligodendrocytes astrocytes), and amelioration of one or more symptoms associated with abnormal function of such cell types.
  • TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to treat neural diseases and/or disorders, e.g.
  • neurodepsy e.g., Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis and progressive supra-nuclear palsy.
  • TANGO 339 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the brain) and/or cells (e.g., neurons) in which TANGO 339 is expressed.
  • TANGO 339 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human TANGO 353 was identified by analyzing the sequences of clones present in a mixed lymphocyte reaction library for sequences that encode a wholly secreted or transmembrane protein. This analysis led to the identification of a clone, jthLa03 lgl2 encoding full-length human TANGO 353.
  • the human TANGO 353 cDNA of this clone is 1239 nucleotides long ( Figure 5; SEQ ID NO:27).
  • Figure 6 depicts a hydropathy plot of human TANGO 353. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and N-glycosylation sites (NGly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
  • SIGNALP Signal peptide prediction program
  • human TANGO 353 includes a 14 amino acid signal peptide (amino acid 1 to amino acid 14 of SEQ ID NO:29; SEQ ID NO:31) preceding the mature human TANGO 353 protein (co ⁇ esponding to amino acid 15 to amino acid 230 of SEQ ID NO:29; SEQ ID NO:30).
  • the molecular weight of human TANGO 353 protein without post-translational modifications is 24.8 kDa prior to the cleavage of the signal peptide and 23.3 kDa after cleavage of the signal peptide.
  • Human TANGO 353 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • the human TANGO 353 protein contains an extracellular domain at amino acid residues 15 to 116 of SEQ ID NO:29 (SEQ ID NO:32), a transmembrane domain at amino acid residues 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33), and a cytoplasmic domain at amino acid residues 142 to 230 of SEQ ID NO:29 (SEQ ID NO:34).
  • a human TANGO 353 protein contains a cytoplasmic domain at amino acid residues 15 to 116 of SEQ ID NO:29, a transmembrane domain at amino acid residues 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33), and an extracellular domain at amino acid residues 142 to 230 of SEQ ID NO:29.
  • the nucleotide at position 68 is thymine (T)(SEQ ID NO:28).
  • the amino acid at position 23 is valine (V)(SEQ ID NO:29).
  • a species variant of human TANGO 353 has a nucleotide at position 68 which is cytosine (C)(SEQ ID NO: 144).
  • the amino acid at position 23 is alanine (A)(SEQ ID NO:145), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 353 the nucleotide at position 77 is adenine (A)(SEQ ID NO:28).
  • amino acid at position 26 is tyrosine (Y)(SEQ ID NO:29).
  • a species variant of human TANGO 353 has a nucleotide at position 77 which is thymine (T)(SEQ ID NO: 146).
  • amino acid at position 26 is phenylalanine (F)(SEQ ID NO: 147), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 353 is guanine (G)(SEQ ID NO:28).
  • amino acid at position 68 is arginine (R)(SEQ ID NO:29).
  • a species variant of human TANGO 353 has a nucleotide at position 203 which is adenine (A)(SEQ ID NO: 148).
  • amino acid at position 68 is histidine (H)(SEQ ID NO: 149), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 353 the nucleotide at position 309 is cytosine (C)(SEQ ID NO:28).
  • amino acid at position 103 is aspartate (D)(SEQ ID NO:29).
  • a species variant of human TANGO 353 has a nucleotide at position 309 which is guanine (G)(SEQ ID NO: 150).
  • the amino acid at position 103 is glutamate (E)(SEQ ID NO: 151), i.e., a conservative substitution.
  • E glutamate
  • the first has the sequence NFTL (at amino acid residues 48 to 51 of SEQ ID NO:29), the second has the sequence NLSG (at amino acid residues 73 to 76 of SEQ ID NO:29), the third has the sequence NQSQ (at amino acid residues 97 to 100 of SEQ ID NO:29), and the fourth has the sequence NVSF (at amino acid residues 109 to 112 of SEQ ED NO:29).
  • Human TANGO 353 has one cAMP- and cGMP-dependent protein kinase phosphorylation site with the sequence KRPT (at amino acid residues 209 to 212 of SEQ ED NO:29). Five protein kinase C phosphorylation sites are present in human TANGO 353.
  • the first has the sequence SIR (at amino acid residues 19 to 21 of SEQ ID NO:29), the second has the sequence SSK (at amino acid residues 78 to 80 of SEQ ID NO:29), the third has the sequence SAK (at amino acids 180 to 182 of SEQ ID NO:29), the fourth has the sequence TRK (at amino acid residues 207 to 209 of SEQ ED NO:29), and the fifth has the sequence TFR (at amino acid residues 225 to 227 of SEQ ID NO:29).
  • Human TANGO 353 has four casein kinase II phosphorylation sites.
  • the first has the sequence SSQE (at amino acid residues 28 to 31 of SEQ ID NO:29), the second has the sequence TMPE (at amino acid residues 183 to 186 of SEQ ID NO:29), the third has the sequence TLDD (at amino acid residues 191 to 194 of SEQ ID NO:29), and the fourth has the sequence SSPE (at amino acid residues 216 to 219 of SEQ ID NO:29).
  • Human TANGO 353 has two N- myristylation sites.
  • the first has the sequence GNFPGA (at amino acid residues 42 to 47 of SEQ ID NO:29) and the second has the sequence GVTFNL (at amino acid residues 69 to 74 of SEQ ID NO:29).
  • Clone EpT353 which encodes human TANGO 353, was deposited with the
  • TANGO 353 was originally found in a mixed lymphocyte library, TANGO 353 nucleic acids, proteins, and modulators thereof can be utilized to diagnose disorders and/or modulate processes involved in lymphocyte development, differentiation and activity, including, but not limited to development, differentiation and activation of T cells, including T helper, T cytotoxic and non-specific T killer cell types and subtypes, and B cells, immune functions associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types.
  • T cells including T helper, T cytotoxic and non-specific T killer cell types and subtypes, and B cells, immune functions associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types.
  • Such disorders can include, but are not limited to, autoimmune disorders (e.g., autoimmune thyroiditis, Type I diabetes mellitus, insulin-resistant diabetes, autoimmune anemia, multiple sclerosis, rheumatoid arthritis, lupus or sclerodoma, allergy, including allergic rhinitis and food allergies, asthma, psoriasis, graft rejection, transplantation rejection, graft versus host disease, pathogenic susceptibilities), inflammatory disorders (e.g., bacterial or viral infections, wound healing and inflammatory bowel disease and arthritis), apoptotic disorders, and cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF).
  • autoimmune disorders e.g., autoimmune thyroiditis, Type I diabetes mellitus, insulin-resistant diabetes, autoimmune anemia, multiple sclerosis, rheumatoid arthritis, lupus or sclerodoma
  • allergy including
  • TANGO 353 associated disorders can include TNF related disorders (e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), immunological differentiative and apoptotic disorders (e.g., hyper- proliferative syndromes such as systemic lupus erythematosus (lupus)), and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer).
  • TNF related disorders e.g., acute myocarditis, myocardial infarction, congestive heart failure
  • T cell disorders e.g., dermatitis, fibrosis
  • immunological differentiative and apoptotic disorders e.g., hyper- proliferative syndromes such as systemic lupus erythematosus (lupus)
  • disorders related to angiogenesis e.g., tumor formation and/or metastas
  • TANGO 353 is expressed in mixed lymphocyte cultures, and hence likely expressed in bone marrow
  • nucleic acids, proteins, and modulators thereof can be used to diagnose disorders associated with cells in the bone ma ⁇ ow and/or modulate the proliferation, differentiation, and/or function of cells that appear in the bone ma ⁇ ow, e.g., stem cells (e.g., hematopoietic stem cells), and blood cells, e.g., erythrocytes, platelets, and leukocytes.
  • stem cells e.g., hematopoietic stem cells
  • blood cells e.g., erythrocytes, platelets, and leukocytes.
  • TANGO 353 nucleic acids, proteins, and modulators thereof can be used to treat bone ma ⁇ ow, blood, and hematopoietic associated diseases and disorders, e.g., acute myeloid leukemia, hemophilia, leukemia, anemia (e.g., sickle cell anemia), and thalassemia.
  • diseases and disorders e.g., acute myeloid leukemia, hemophilia, leukemia, anemia (e.g., sickle cell anemia), and thalassemia.
  • TANGO 353 is a transmembrane protein
  • nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling cascades, or alternatively.
  • TANGO 353 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., spleen) and/or cells (e.g., lymphocytes) in which TANGO 353 is expressed.
  • TANGO 353 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g. , in radiation hybrid mapping.
  • a cDNA encoding human TANGO 358 was identified by analyzing the sequences of clones present in a fetal thymus library for sequences that encode a wholly secreted or transmembrane protein. This analysis led to the identification of a clone, jthTbl28c07 encoding full-length human TANGO 358.
  • the human TANGO 358 cDNA of this clone is 1608 nucleotides long ( Figure 7; SEQ ED NO:36).
  • Figure 8 depicts a hydropathy plot of human TANGO 358. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10: 1-6) predicted that human TANGO 358 includes a 42 amino acid signal peptide (amino acid 1 to amino acid 42 of SEQ ID NO:36; SEQ ID NO:40) preceding the mature human TANGO 358 protein (co ⁇ esponding to amino acid 43 to amino acid 82 of SEQ ID NO:36; SEQ ID NO:39).
  • the molecular weight of human TANGO 358 protein without post-translational modifications is 9.5 kDa prior to the cleavage of the signal peptide and 4.5 kDa after cleavage of the signal peptide.
  • Human TANGO 358 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • the human TANGO 358 protein contains an extracellular domain at amino acid residues 43 to 49 of SEQ ID NO:38 (SEQ ED NO:41), a transmembrane domain at amino acid residues 50 to 66 of SEQ ED NO:38 (SEQ ED NO:42), and a cytoplasmic domain at amino acid residues 67 to 82 of SEQ ED NO:38 (SEQ ED NO:43).
  • a human TANGO 358 protein contains a cytoplasmic domain at amino acid residues 43 to 49 of SEQ ID NO:38, a transmembrane domain at amino acid residues 50 to 66 of SEQ ED NO:38 (SEQ ED NO:42), and an extracellular domain at amino acid residues 67 to 82 of SEQ ID NO:38.
  • human TANGO 358 has a protein kinase C phosphorylation site with the sequence SIK (at amino acid residues 45 to 47 of SEQ ID NO:38).
  • the nucleotide at position 20 is adenine (A)(SEQ ID NO:37).
  • the amino acid at position 7 is histidine (H)(SEQ ID NO:38).
  • a species variant of human TANGO 358 has a nucleotide at position 20 which is guanine (G)(SEQ ID NO: 152).
  • the amino acid at position 7 is arginine (R)(SEQ ID NO: 153), i. e. , a conservative substitution.
  • nucleotide sequence of human TANGO 358 the nucleotide at position 35 is thymine (T)(SEQ ID NO:37).
  • amino acid at position 12 is valine (V)(SEQ ID NO:38).
  • a species variant of human TANGO 358 has a nucleotide at position 35 which is cytosine (C)(SEQ ID NO:154).
  • the amino acid at position 12 is alanine (A)(SEQ ID NO:155), i.e., a conservative substitution.
  • the nucleotide at position 85 is thymine (T)(SEQ ID NO:37).
  • the amino acid at position 29 is serine (S)(SEQ ID NO:38).
  • a species variant of human TANGO 358 has a nucleotide at position 85 which is adenine (A)(SEQ ID NO: 156).
  • the amino acid at position 29 is threonine (T)(SEQ ED NO:157), i.e., a conservative substitution.
  • the nucleotide at position 91 is cytosine (C)(SEQ ID NO:37).
  • the amino acid at position 31 is glutamine (Q)(SEQ ID NO:38).
  • a species variant of human TANGO 358 has a nucleotide at position 91 which is guanine (G)(SEQ ED NO: 158).
  • the amino acid at position 31 is glutamate (E)(SEQ ID NO: 159), i.e., a conservative substitution.
  • TANGO 358 nucleic acids, proteins, and modulators thereof can be used to diagnose thymus associated disorders.
  • TANGO 358 nucleic acids, proteins, and modulators thereof can also be used modulate the proliferation, development, differentiation, maturation and/or function of thymocytes, e.g., modulate development and maturation of T-lymphocytes.
  • TANGO 358 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes such as the ability to modulate host immune response by, e.g., modulating the formation of and/or binding to immune complexes, and modulating the positive and negative selection of thymocytes.
  • Such TANGO 358 compositions and modulators thereof can be utilized, e.g., to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to infection and autoimmune disorders (e.g., insulin-dependent mellitus, multiple sclerosis, systemic lupus, erythematosus, sjogren's syndrome, autoimmune thyroiditis, idiotpathic Addison's disease, vitiligo, Grave's disease, idiopathic thrombocytopenia purpura, rheumatoid arthritis, and scleroderma).
  • autoimmune disorders e.g., insulin-dependent mellitus, multiple sclerosis, systemic lupus, erythematosus, sjogren's syndrome, autoimmune thyroiditis, idiotpathic Addison's disease, vitiligo, Grave's disease, idiopathic thrombocytopenia purpura, rheumatoid arthritis,
  • TANGO 358 nucleic acids, proteins and modulators thereof can also be utilized to treat viral infections, inflammatory immune disorders and immune-related cancers including but not limited to, leukemia (e.g., acute leukemia, chronic leukemia, Hodgkin's disease non-Hodgkin's lymphoma ,and multiple myeloma).
  • leukemia e.g., acute leukemia, chronic leukemia, Hodgkin's disease non-Hodgkin's lymphoma ,and multiple myeloma.
  • TANGO 358 disorders including those which TANGO 358 proteins, nucleic acids and modulators thereof may be an antagonist
  • immune disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)).
  • autoimmune disorders e.g., arthritis, graft rejection (e.g., allograft rejection)
  • T cell disorders e.g., AIDS
  • inflammatory disorders e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis,
  • Disorders associated with modulated TANGO 358 activity can also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF).
  • apoptotic disorders e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus
  • cytotoxic disorders e.g., septic shock, cachexia
  • proliferative disorders e.g., B cell cancers stimulated by TNF.
  • TANGO 358 proteins, nucleic acids and modulators thereof can be used to treat disorders that include TNF-related disorders (e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer).
  • TNF-related disorders e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer).
  • Modulators of TANGO 358 expression and/or activity can be used to treat such disorders.
  • TANGO 358 is a transmembrane protein
  • nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extra-cellular matrix.
  • TANGO 358 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., T-lymphocytes) in which TANGO 358 is expressed.
  • TANGO 358 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding TANGO 365 was identified by analyzing the sequences of clones present in a human prostate fibroblast library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthqc001g06, encoding full-length Human TANGO 365.
  • the TANGO 365 cDNA of this clone is 1338 nucleotides long ( Figure 9; SEQ ID NO:44).
  • Figure 10 depicts a hydropathy plot of human TANGO 365. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 36 of SEQ ID NO:46; SEQ ID NO:47) on the left from the mature protein (amino acids 37 to 165 of SEQ ID NO:46; SEQ ID NO:48) on the right.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 365 includes a 36 amino acid signal peptide (amino acid 1 to amino acid 36 of SEQ ID NO:46; SEQ ID NO:47) preceding the mature protein (co ⁇ esponding to amino acid 37 to amino acid 165 of SEQ ED NO:46; SEQ ED NO:48).
  • the molecular weight of TANGO 365 protein without post-translational modifications is 17.4 kDa prior to the cleavage of the signal peptide, 13.6 kDa after cleavage of the signal peptide.
  • Human TANGO 365 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • the human TANGO 365 protein contains two extracellular domains; one at amino acid residues 37 to 51 of SEQ ID NO:46 (SEQ ED NO:232); and a second at amino acid residues 95 to 165 of SEQ ID NO:46 (SEQ ID NO:51), two hydrophobic transmembrane domains; one at amino acids 52 to 70 of SEQ ED NO:46 (SEQ ID NO:49); and a second at amino acids 78 to 94 of SEQ ID NO:46 (SEQ ID NO:50), and a cytoplasmic domain at amino acid residues 71 to 77 of SEQ ID NO:46 (SEQ ID NO:234).
  • a human TANGO 365 protein contains two cytoplasmic domains; one at amino acid residues 37 to 51 of SEQ ID NO:46; and a second at amino acid residues 95 to 165 of SEQ ID NO:46, two hydrophobic transmembrane domains; one at amino acids 52 to 70 of SEQ ID NO:46 (SEQ ID NO:49); and a second at amino acids 78 to 94 of SEQ ED NO:46 (SEQ ED NO:50), and an extracellular domain at amino acid residues 71 to 77 of SEQ ED NO:46.
  • nucleotide sequence of human TANGO 365 the nucleotide at position 14 is cytosine (C)(SEQ ID NO:45).
  • amino acid at position 5 is alanine (A)(SEQ ID NO:46).
  • a species variant of human TANGO 365 has a nucleotide at position 14 which is thymidine (T)(SEQ ID NO: 160).
  • the amino acid at position 5 is valine (V)(SEQ ID NO: 161), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 365 the nucleotide at position 41 is guanine (G)(SEQ ID NO:45).
  • amino acid at position 14 is arginine (R)(SEQ ID NO:46).
  • a species variant of human TANGO 365 has a nucleotide at position 41 which is adenine (A)(SEQ ID NO: 162).
  • the amino acid at position 14 is histidine (H)(SEQ ID NO: 163), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 365 is cytosine (C)(SEQ ID NO:45).
  • amino acid at position 20 is threonine (T)(SEQ ID NO:46).
  • a species variant of human TANGO 365 has a nucleotide at position 59 which is guanine (G)(SEQ ED NO: 164).
  • the amino acid at position 20 is serine (S)(SEQ ID NO: 165), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 365 the nucleotide at position 115 is adenine (A)(SEQ ID NO:45).
  • amino acid at position 39 is asparagine (N)(SEQ ID NO:46).
  • a species variant of human TANGO 365 has a nucleotide at position 115 which is guanine (G)(SEQ ID NO: 166).
  • amino acid at position 39 is aspartate (D)(SEQ ID NO: 167), i.e., a conservative substitution.
  • One protein kinase C phosphorylation site is present in human TANGO 365.
  • the site has the sequence SLR and is found (at amino acids 139 to 141 of SEQ ID NO:46).
  • the TANGO 365 protein has four N-myristoylation sites. The first has the sequence GGTRCR and is found (at amino acids 18 to 23 of SEQ ID NO:46), the second has the sequence GTSMAC and is found (at amino acids 32 to 37 of SEQ ID NO:46), the third has the sequence GAACSL and is found (at amino acids 87 to 92 of SEQ ED NO:46), and the fourth has the sequence GSSDSS and is found (at amino acids 144 to 149 of SEQ ID NO:46).
  • Human TANGO 365 also has an amidation site which has the sequence of LGRR (at amino acids 69 to 72 of SEQ ID NO:46).
  • TANGO 365 was identified as being expressed in a prostate fibroblast library.
  • nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate processes involved in prostate development, differentiation and activity, including, but not limited to development, and differentiation and activation of prostate tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types.
  • disorders can include, but are not limited to, malignant or benign prostate cell growth.
  • disorders can include, but are not limited to, malignant or benign prostate cell growth.
  • the TANGO 365 proteins can be used to treat subjects with or without prostate cancer e.g., prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato-rectal fistulas, atypical prostatic stromal lesions.
  • prostate cancer e.g., prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato-rectal fistulas, atypical prostatic stromal lesions.
  • TANGO 365 nucleic acids, proteins, and modulators thereof can also be used to treat disorders of the cells and tissues in which it is expressed.
  • TANGO 365 is a transmembrane protein
  • proteins, nucleic acids and modulators thereof can be used to diagnose disorders and/or modulate intercellular signaling processes by disrupting or enhancing ligand-receptor or cell interaction with the extracellular matrix.
  • TANGO 365 could be used in detection and diagnostic assays to assay for normal or inappropriate expression of TANGO 365 proteins in abe ⁇ antly growing cells.
  • TANGO 365 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the prostate) and/or cells (e.g., fibroblasts) in which TANGO 365 is expressed.
  • TANGO 365 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human TANGO 368 was identified by analyzing the sequences of clones present in a natural killer cell library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthta080f06, encoding full-length human TANGO 368.
  • the human TANGO 368 cDNA of this clone is 983 nucleotides long ( Figure 11; SEQ ID NO:52).
  • Figure 12 depicts a hydropathy plot of human TANGO 368. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and N-glycosylation sites (NGly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 368 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 27 of SEQ ED NO:54; SEQ ID NO:56) preceding the mature human TANGO 368 protein (co ⁇ esponding to amino acid 28 to amino acid 59 of SEQ ED NO:54; SEQ ED NO:55).
  • the molecular weight of TANGO 368 protein without post-translational modifications is 6.5 kDa prior to the cleavage of the signal peptide and 3.5 kDa after cleavage of the signal peptide.
  • nucleotide sequence of human TANGO 368 the nucleotide at position 8 is cytosine (C)(SEQ ID NO:53).
  • amino acid at position 3 is threonine (T)(SEQ ID NO:54).
  • a species variant of human TANGO 368 has a nucleotide at position 8 which is guanine (G)(SEQ ED NO: 168).
  • the amino acid at position 3 is serine (S)(SEQ ID NO: 169), i.e., a conservative substitution.
  • the nucleotide at position 10 is cytosine (C)(SEQ ID NO:53).
  • the amino acid at position 4 is glutamine (Q)(SEQ ID NO:54).
  • a species variant of human TANGO 368 has a nucleotide at position 10 which is guanine (G)(SEQ ID NO: 170).
  • the amino acid at position 4 is glutamate (E)(SEQ ID NO: 171), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 368 the nucleotide at position 16 is cytosine (C)(SEQ ID NO:53).
  • amino acid at position 6 is leucine (L)(SEQ ID NO:54).
  • a species variant of human TANGO 368 has a nucleotide at position 16 which is guanine (G)(SEQ ID NO: 172).
  • the amino acid at position 6 is valine (V)(SEQ ID NO:173), i.e., a conservative substitution.
  • the nucleotide at position 110 is adenine (A)(SEQ ID NO:53).
  • the amino acid at position 37 is histidine (H)(SEQ ID NO:54).
  • a species variant of human TANGO 368 has a nucleotide at position 110 which is guanine (G)(SEQ ID NO: 174).
  • the amino acid at position 37 is arginine (R)(SEQ ID NO:175), i.e., a conservative substitution.
  • Human TANGO 368 has an N-glycosylation site with the sequence NFTC (at amino acid residues 40 to 43 of SEQ ID NO: 54), a protein kinase C phosphorylation site with the sequence SLK (at amino acid residues 24 to 26 of SEQ ID NO:54), and a casein kinase II phosphorylation site with the sequence TQPE (at amino acid residues 27 to 30 of SEQ ID NO:54).
  • Figure 13 depicts a local alignment of the nucleotide sequence of full length human TANGO 368 (SEQ ID NO:52) and a fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57), which maps to a region of human chromosome 7.
  • the full-length nucleic acid sequence of human TANGO 368 (SEQ ID NO:52) has 99.3% identity to a 973 bp fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57).
  • Northern blots were performed to analyze the expression of human TANGO 368 mRNA in human tissues. A weak signal was observed in the spleen and lymph node, however, no expression was detected in the thymus, peripheral blood leukocytes or fetal liver.
  • Clone EpT368, which encodes human TANGO 368 was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-291. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. ⁇ 112.
  • TANGO 368 was originally found in a natural killer cell library, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of immune cells, such as lymphocytes, e.g., natural killer cells, T-cells and B-cells.
  • TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes e.g., the host immune response by, for example, modulating the formation of and/or binding to immune complexes, detection and defense against surface antigens and bacteria, and immune surveillance for rapid removal or pathogens.
  • Such TANGO 368 nucleic acids, proteins and modulators thereof can be utilized, e.g., to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to viral or bacterial infection, autoimmune disorders (e.g., Grave's disease, Hashimoto's disease, and arthritis), immunodeficiency disorders (e.g., HIV, and inflammatory disorders (e.g., asthma, arthritis, psoriasis, septicemia, inflammatory bowel disease and allergies).
  • autoimmune disorders e.g., Grave's disease, Hashimoto's disease, and arthritis
  • immunodeficiency disorders e.g., HIV
  • inflammatory disorders e.g., asthma, arthritis, psoriasis, septicemia, inflammatory bowel disease and allergies.
  • TANGO 368 exhibits expression in the spleen
  • nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the spleen, e.g., cells of the splenic connective tissue, e.g., splenic smooth muscle cells and/or endothelial cells of the splenic blood vessels.
  • TANGO 368 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., regenerated or phagocytized within the spleen, e.g., erythrocytes and/or B and T lymphocytes and macrophages.
  • TANGO 368 nucleic acids, proteins, and modulators thereof can be used to treat spleen, e.g., the fetal spleen, associated diseases and disorders.
  • splenic diseases and disorders include e.g., splenic lymphoma and/or splenomegaly, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
  • TANGO 368 exhibits expression in the lymph nodes
  • nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the lymph node, e.g., cells of the lymph node connective tissue, e.g., lymph node smooth muscle cells and/or endothelial cells of the lymph node blood vessels.
  • TANGO 368 nucleic acids, proteins, and modulators thereof can also be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., phagocytized within the lymph node, e.g., erythrocytes and/or B and T lymphocytes and macrophages.
  • TANGO 368 nucleic acids, proteins, and modulators thereof can be used to treat lymph node associated diseases and disorders.
  • lymph node diseases and disorders include e.g., lymphadenopathy, lymphoma, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
  • lymph node diseases and disorders include e.g., lymphadenopathy, lymphoma, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
  • TANGO 368 is
  • TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate the recognition of antigens in association with the major histocompatibility complex.
  • TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate diseases and/or disorders associated with abe ⁇ ant TCR-MHC interactions. Further, TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate cell-cell receptor interactions.
  • TANGO 368 exhibits homology to human T-cell receptor gamma VI gene region (Accession Numbers AF057177), which maps to a region of chromosome 7, TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate diseases associated with that region of chromosome 7, e.g., Stiff-Mann syndrome.
  • TANGO 368 is a secreted protein and thus likely a signaling molecule
  • nucleic acids, proteins or modulators thereof can be used to modulate TANGO 368 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. , in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor-ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • TANGO 368 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., natural killer cells) in which TANGO 368 is expressed.
  • TANGO 368 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • Human TANGO 369 A cDNA encoding human TANGO 369 was identified by analyzing the sequences of clones present in a natural killer cell library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthta088h08, encoding full-length human TANGO 369.
  • the human TANGO 369 cDNA of this clone is 1119 nucleotides long ( Figure 14; SEQ ID NO:58).
  • Figure 15 depicts a hydropathy plot of human TANGO 369. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 369 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ED NO:60; SEQ ID NO:62) preceding the mature human TANGO 369 protein (co ⁇ esponding to amino acid 27 to amino acid 58 of SEQ ED NO:60; SEQ ED NO:61).
  • the molecular weight of TANGO 369 protein without post-translational modifications is 6.8 kDa prior to the cleavage of the signal peptide and 3.7 kDa after cleavage of the signal peptide.
  • a methionine residue at positions 17 and 25 of SEQ ID NO:60 indicates that there can be alternative forms of human TANGO 369 of 42 amino acids of SEQ ED NO:60, and 34 amino acids of SEQ ED NO:60, respectively.
  • the nucleotide at position 58 is cytosine (C)(SEQ ID NO:59).
  • the amino acid at position 20 is leucine (L)(SEQ ID NO:54).
  • a species variant of human TANGO 369 has a nucleotide at position 58 which is guanine (G)(SEQ ID NO: 176).
  • the amino acid at position 20 is valine (V)(SEQ ID NO: 177), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 369 the nucleotide at position 68 is guanine (G)(SEQ ID NO:59).
  • amino acid at position 23 is arginine (R)(SEQ ID NO:60).
  • a species variant of human TANGO 369 has a nucleotide at position 68 which is adenine (A)(SEQ ID NO: 178).
  • amino acid at position 23 is lysine (K)(SEQ ID NO: 179), i.e., a conservative substitution.
  • the nucleotide at position 70 is thymine (T)(SEQ ID NO:59).
  • the amino acid at position 24 is leucine (L)(SEQ ID NO:60).
  • a species variant of human TANGO 369 has a nucleotide at position 70 which is adenine (A)(SEQ ID NO: 180).
  • the amino acid at position 24 is threonine (T)(SEQ ID NO: 181), i.e., a conservative substitution.
  • the nucleotide at position 120 is guanine (G)(SEQ ID NO:59).
  • the amino acid at position 40 is glutamate (E)(SEQ ID NO:60).
  • a species variant of human TANGO 369 has a nucleotide at position 120 which is cytosine (C)(SEQ ID NO: 182).
  • the amino acid at position 40 is aspartate (D)(SEQ ID NO: 183), i.e., a conservative substitution.
  • Northern blots were performed to analyze the expression of human TANGO 369 mRNA in human tissues. A very weak signal was observed in the spleen and lymph node, however, no expression was detected in the thymus, peripheral blood leukocytes or fetal liver.
  • TANGO 369 was originally found in a natural killer cell library, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of lymphocytes, e.g., natural killer cells.
  • TANGO 369 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes, e.g., the host immune response by, for example, modulating the formation of and/or binding to immune complexes, detection and defense against surface antigens and bacteria, and immune surveillance for rapid removal or pathogens.
  • Such TANGO 369 compositions and modulators thereof can be utilized, eg-, to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to viral or bacterial infection, autoimmune disorders (e.g., Grave's disease, Hashimoto's disease, arthritis, graft rejection), and inflammatory disorders (e.g., bacterial or viral infection, psoriasis, allergies and inflammatory bowel diseases).
  • autoimmune disorders e.g., Grave's disease, Hashimoto's disease, arthritis, graft rejection
  • inflammatory disorders e.g., bacterial or viral infection, psoriasis, allergies and inflammatory bowel diseases.
  • TANGO 369 exhibits expression in the spleen
  • nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the spleen, e.g., cells of the splenic connective tissue, e.g., splenic smooth muscle cells and/or endothelial cells of the splenic blood vessels.
  • TANGO 369 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., regenerated or phagocytized within the spleen, e.g., erythrocytes and/or B and T lymphocytes and macrophages.
  • TANGO 369 nucleic acids, proteins, and modulators thereof can be used to treat spleen, e.g., the fetal spleen, associated diseases and disorders.
  • splenic diseases and disorders include e.g., splenic lymphoma and/or splenomegaly, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
  • TANGO 369 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the lymph node, e.g., cells of the lymph node connective tissue, e.g., lymph node smooth muscle cells and/or endothelial cells of the lymph node blood vessels.
  • TANGO 369 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., phagocytized within the lymph node, e.g., erythrocytes and/or B and T lymphocytes and macrophages.
  • TANGO 369 nucleic acids, proteins, and modulators thereof can be used to treat lymph node associated diseases and disorders.
  • lymph node diseases and disorders include e.g., lymphadenopathy, lymphoma, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
  • TANGO 369 is associated with immune cells.
  • immune disorders associated TANGO 369 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate or treat immune disorders that include, but are not limited to, immune proliferative disorders (e.g., carcinoma, lymphoma, e.g., follicular lymphoma), and disorders associated with fighting pathogenic infections, e.g. , bacterial (e.g., chlamydia) infection, parasitic infection, and viral infection (e.g., HSV infection), and pathogenic disorders associated with immune disorders (e.g., immunodeficiency disorders, such as HIV).
  • immune proliferative disorders e.g., carcinoma, lymphoma, e.g., follicular lymphoma
  • pathogenic infections e.g. bacterial (e.g., chlamydia) infection, parasitic infection, and viral infection (e.g., HSV infection
  • pathogenic disorders associated with immune disorders
  • TANGO 369 nucleic acids, proteins and modulators thereof can be used to modulate, identify, diagnose or treat, include, e.g., autoimmune disorders, such as arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders, such as bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis), apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF).
  • autoimmune disorders such as arthritis, graft rejection (e.g., allograft rejection)
  • TANGO 369 nucleic acids, proteins and modulators thereof can be used to treat such disorders.
  • TANGO 369 is a secreted protein
  • TANGO 369 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extracellular matrix.
  • TANGO 369 is a secreted protein and thus likely a signaling molecule
  • nucleic acids, proteins or modulators thereof can be used TANGO 369 biological activities, which can also include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor-ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • TANGO 369 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., natural killer cells) in which TANGO 369 is expressed.
  • TANGO 369 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human TANGO 383 was identified by analyzing the sequences of clones present in a human prostate epithelium cDNA library. This analysis led to the identification of a clone, jthqb083bl0, encoding full-length TANGO 383.
  • the human cDNA of this clone is 1386 nucleotides long ( Figure 16; SEQ ID NO:63).
  • Figure 17 depicts a hydropathy plot of human TANGO 383. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 20 of SEQ ID NO:65; SEQ ID NO:66) on the left from the mature protein (amino acids 21 to 140 of SEQ ID NO:65; SEQ ID NO:67) on the right.
  • the signal peptide prediction program SIGNALP (Nielsen, et al. (1997) Protein Engineering 10:1-6) predicted that TANGO 383 includes a 20 amino acid signal peptide (amino acid 1 to amino acid 20 of SEQ ID NO:65; SEQ ED NO:66) preceding the mature protein (co ⁇ esponding to amino acid 21 to amino acid 140 of SEQ ID NO:65; SEQ ED NO:67).
  • the molecular weight of TANGO 383 without post-translational modifications is 14.9 kDa prior to the cleavage of the signal peptide, 12.7 kDa after cleavage of the signal peptide.
  • TANGO 383 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain.
  • the TANGO 383 protein contains an extracellular domain at amino acids 71 to 115 of SEQ ID NO:65 (SEQ ID NO:70), a first transmembrane domain at amino acid residues 50 to 70 of SEQ ID NO:65 (SEQ ED NO:68), a second transmembrane domain at amino acid residues 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69), a first cytoplasmic domain at amino acid residues 21 to 49 of SEQ ID NO:65 (SEQ ED NO:235) and a second cytoplasmic domain at amino acid residues 134 to 140 of SEQ ID NO:65 (SEQ ID NO: 136).
  • a TANGO 383 protein contains a cytoplasmic domain at amino acids 71 to 115 of SEQ ID NO: 65, a first transmembrane domain at amino acid residues 50 to 70 of SEQ ID NO:65 (SEQ ID NO:68), a second transmembrane domain at amino acid residues 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69), a first extracellular domain at amino acid residues 21 to 49 of SEQ ID NO:65 and a second extracellular domain at amino acid residues 134 to 140 of SEQ ID NO:65.
  • nucleotide sequence of human TANGO 383 the nucleotide at position 4 is cytosine (C)(SEQ ID NO:64).
  • amino acid at position 2 is leucine (L)(SEQ ID NO:65).
  • a species variant of human TANGO 383 has a nucleotide at position 4 which is adenine (A)(SEQ ID NO: 184).
  • the amino acid at position 2 is isoleucine (I)(SEQ ED NO: 185), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 383 the nucleotide at position 8 is guanine (G)(SEQ ID NO:64).
  • amino acid at position 3 is serine (S)(SEQ ID NO:65).
  • a species variant of human TANGO 383 has a nucleotide at position 8 which is cytosine (C)(SEQ ID NO: 186).
  • the amino acid at position 3 is threonine (T)(SEQ ID NO: 187), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 383 the nucleotide at position 17 is adenine (A)(SEQ ID NO:64).
  • amino acid at position 6 is lysine (K)(SEQ ID NO:65).
  • a species variant of human TANGO 383 has a nucleotide at position 17 which is guanine (G)(SEQ ID NO: 188).
  • amino acid at position 6 is arginine (R)(SEQ ID NO: 189), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 383 the nucleotide at position 57 is cytosine (C)(SEQ ID NO:64).
  • amino acid at position 19 is aspartate (D)(SEQ ID NO:65).
  • a species variant of human TANGO 383 has a nucleotide at position 57 which is guanine (G)(SEQ ID NO: 190).
  • the amino acid at position 19 is glutamate (E)(SEQ ID NO: 191), i.e., a conservative substitution.
  • TANGO 383 One protein kinase C phosphorylation site is present in TANGO 383, and has the sequence SPR (at amino acids 21 to 24 of SEQ ID NO:65).
  • TANGO 383 has one casein kinase II phosphorylation site which has the sequence SKAE (at amino acids 42 to 45 of SEQ ID NO:65).
  • TANGO 383 has three N-myristylation sites. The first has the sequence GVELAS (at amino acids 24 to 29 of SEQ ID NO:65), the second has the sequence GAVLAH (at amino acids 84 to 89 of SEQ ED NO:65), and the third has the sequence GSSDSH (at amino acids 96 to 101 of SEQ ID NO:65).
  • TANGO 383 has a consensus tyrosine phosphorylation site which has the amino acid sequence RGKREAGLY and (at amino acids 33 to 41 of SEQ ID NO:65). TANGO 383 also has an amidation site with the sequence RGKR (at amino acids 33-36 of SEQ ID NO:65).
  • Figure 18 depicts an alignment of the amino acid sequence of TANGO 383 (SEQ ID NO:65).
  • TANGO 383 exhibits homology to neural thread proteins which are phospho-proteins expressed in the central nervous system which are phosphorylated during neuritic sprouting. Therefore, TANGO 383 nucleic acids, proteins and modulators thereof may be used to diagnose disorders and/or inhibit or modulate neurodegenerative sprouting and synaptic disassociation associated with, e.g., Alzheimer's disease, and other diseases in neural tissue as discussed below.
  • TANGO 383 was originally found in a prostate epithelium library, TANGO 383 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of prostate cells.
  • TANGO 383 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in prostate development, differentiation and activity, including, but not limited to development, and differentiation and activation of prostate tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types or disorders associated with such cell types.
  • disorders can include, but are not limited to, malignant or benign prostate cell growth or inflammatory disorders (e.g.
  • prostatitis benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato-rectal fistulas, atypical prostatic stromal lesions).
  • BPH benign prostatic hyperplasia
  • prostatic paraganglioma prostate adenocarcinoma
  • prostatic intraepithelial neoplasia prostatic intraepithelial neoplasia
  • prostato-rectal fistulas atypical prostatic stromal lesions
  • TANGO 383 exhibits homology to neural thread proteins which are phospho- proteins expressed in the central nervous system which are phosphorylated during neuritic sprouting. Therefore, TANGO 383 nucleic acids, proteins and modulators thereof may be used to diagnose disorders and/or inhibit or modulate neurodegenerative sprouting and synaptic disassociation associated with, e.g., Alzheimer's disease.
  • TANGO 383 nucleic acids, proteins and modulators thereof may also be utilized to diminish the effects of stroke and other neural damage, e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra- nuclear palsy, and other dementias.
  • stroke and other neural damage e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra- nuclear palsy, and other dementias.
  • TANGO 383 is a transmembrane protein
  • nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extracellular matrix.
  • TANGO 383 is a transmembrane protein and thus likely a signaling molecule
  • nucleic acids, proteins or modulators thereof, activities can include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor-ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • TANGO 383 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the prostate) and/or cells (e.g., epithelial cells) in which TANGO 383 is expressed.
  • TANGO 383 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human TANGO 393 was identified by analyzing the sequences of clones present in a human fetal hypothalamus cDNA library for sequences containing signal peptides. This analysis led to the identification of a clone, jthhb039f09, encoding full-length human TANGO 393.
  • the human cDNA of this clone is 1778 nucleotides long ( Figure 19; SEQ ID NO:73).
  • Figure 20 depicts a hydropathy plot of human TANGO 393. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line.
  • the cysteine residues are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line.
  • human TANGO 393 includes an 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:75; SEQ ID NO:76) preceding the mature protein (co ⁇ esponding to amino acid 27 to amino acid 473 of SEQ ED NO:75; SEQ ID NO:77).
  • the molecular weight of human TANGO 393 without post-translational modifications is 50.7 kDa prior to the cleavage of the signal peptide, 47.8 kDa after cleavage of the signal peptide.
  • the presence of a methionine residue at position 229 of SEQ ID NO: 75 indicates that there can be alternative forms of human TANGO 393 of 245 amino acids of SEQ ID NO:75.
  • Human TANGO 393 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain; and (4) a leucine-rich domain.
  • the human TANGO 393 protein contains an extracellular domain at amino acids 27 to 447 of SEQ ID NO:75 (SEQ ID NO:89), a transmembrane domain at amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a cytoplasmic domain at amino acid residues 468 to 473 of SEQ ED NO:75 (SEQ ED NO: 134).
  • a human TANGO 393 protein contains a cytoplasmic domain at amino acids 27 to 447 of SEQ ID NO:75, a transmembrane domain at amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a extracellular domain at amino acid residues 468 to 473 of SEQ ID NO:75.
  • the nucleotide at position 5 is adenine (A)(SEQ ID NO:74).
  • the amino acid at position 2 is lysine (K)(SEQ ID NO:75).
  • a species variant of human TANGO 393 has a nucleotide at position 5 which is guanine (G)(SEQ ID NO: 192).
  • the amino acid at position 2 is arginine (R)(SEQ ID NO: 193), i. e. , a conservative substitution.
  • the nucleotide at position 17 is cytosine (C)(SEQ ID NO:74).
  • the amino acid at position 6 is alanine (A)(SEQ ID NO:75).
  • a species variant of human TANGO 393 has a nucleotide at position 17 which is thymidine (T)(SEQ ED NO: 194).
  • the amino acid at position 6 is valine (V)(SEQ ED NO: 195), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 393 the nucleotide at position 55 is cytosine (C)(SEQ ID NO:74).
  • amino acid at position 19 is glutamine (Q)(SEQ ID NO:75).
  • a species variant of human TANGO 393 has a nucleotide at position 55 which is guanine (G)(SEQ ID NO: 196).
  • the amino acid at position 19 is glutamate (E)(SEQ ID NO: 197), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 393 the nucleotide at position 118 is adenine (A)(SEQ ID NO:74).
  • amino acid at position 40 is threonine (T)(SEQ ID NO:75).
  • a species variant of human TANGO 393 has a nucleotide at position 118 which is thymine (T)(SEQ ID NO: 198).
  • the amino acid at position 40 is serine (S)(SEQ ID NO: 199), i.e., a conservative substitution.
  • Human TANGO 393 has LRR from amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and 260 to 310 of SEQ ID NO:75 (SEQ ID NO:79, 80, 81, 82, 83, 84, 85, 86, 87 and 88, respectively). These repeats are spaced in beta-alpha folds in the structure of the protein, so as to create a hydrophobic face that induces particular folding of the protein.
  • Human TANGO 393 has five N-glycosylation sites. The first has a sequence of NLTI (at amino acids 82-85 of SEQ ID NO:75), the second has a sequence of NLTH (at amino acids 179 to 182 of SEQ ID NO:75), the third has a sequence of NLSA (at amino acids 237 to 240 of SEQ ID NO:75), the fourth has a sequence of NGSG (at amino acids 372 to 375 of SEQ ID NO:75), and the fifth has a sequence of NRTR (at amino acids 423 to 426 of SEQ ID NO:75). Human TANGO 393 has one Glycosaminoglycan attachment site, the sequence of which is SGGG (at amino acids 436 to 439 of SEQ ID NO:75).
  • Human TANGO 393 has one cAMP and cGMP-dependent protein kinase phosphorylation site, the sequence of which is KRAS (at amino acids 2 to 5 of SEQ ID NO:75). Human TANGO 393 has five protein kinase C phosphorylation sites, where the first has a sequence SQR of (at amino acids 59 to 61 of SEQ ID NO:75), the second has a sequence SFR of (at amino acids 76 to 78 of SEQ ID NO:75), the third has a sequence TFR of (at amino acids 173 to 175 of SEQ ID NO:75), the fourth has a sequence TGR of (at amino acids 321 to 323 of SEQ ID NO:75), and the fifth has a sequence SRK of (at amino acids 420 to 422 of SEQ ID NO:75).
  • Human TANGO 393 has five casein kinase II phosphorylation sites, where the first has a sequence of TFRD (at amino acids 173 to 176 of SEQ ID NO:75), the second has a sequence of SVPE (at amino acids 192 to 195 of SEQ ID NO:75), the third has a sequence of SSSE (at amino acids 281 to 284 of SEQ ID NO:75), the fourth has a sequence of TDEE (at amino acids 325 to 328 of SEQ ID NO: 75), and the fifth has a sequence of SVLE (at amino acids 345 to 348 of SEQ ED NO: 75).
  • TFRD at amino acids 173 to 176 of SEQ ID NO:75
  • the second has a sequence of SVPE (at amino acids 192 to 195 of SEQ ID NO:75)
  • the third has a sequence of SSSE (at amino acids 281 to 284 of SEQ ID NO:75)
  • the fourth has a sequence of TDEE (at amino acids
  • Human TANGO 393 has eleven N-myristylation sites, where the first has the sequence GACVCY (at amino acids 29 to 34 of SEQ ID NO:75), the second has the sequence GFPAAS (at amino acids 54 to 59 of SEQ ED NO:75), and the third has the sequence GNRESH (at amino acids 66 to 71 of SEQ ED NO:75), the fourth has the sequence GLFRGL (at amino acids 148 to 153 of SEQ ED NO:75), and the fifth has the sequence GNRISS (at amino acids 187 to 192 of SEQ ED NO:75), the sixth has the sequence GCAVAT (at amino acids 308 to 313 of SEQ ED NO:75), and the seventh has the sequence GLPKCC (at amino acids 331 to 336 of SEQ ED NO:75), the eighth has the sequence GTLPGS (at amino acids 385 to 390 of SEQ ED NO:75), and the ninth has the sequence GQAGSG (at amino acids 432 to 437 of SEQ ED
  • LHLDRCGLQELGPGLFRGLAAL (at amino acids 135 to 156 of SEQ ID NO:75).
  • Human TANGO 393 maps by homology to ESTs to Chromosome 22 between D22S420 and D22S446.
  • a cDNA encoding mouse TANGO 393 was identified in an analysis of a fetal hypothalamus library for screening encoding signal peptides. This analysis led to the identification of a clone, jtmoa038d08, encoding full-length mouse TANGO 393.
  • the mouse cDNA of this clone is 1946 nucleotides long ( Figure 21; SEQ ID NO:93).
  • the open reading frame is from nucleotides 226 to 1644 of SEQ ED NO:93 (SEQ ED NO:94), encodes a 473 amino acid mouse TANGO 393 transmembrane protein ( Figure 21; SEQ ED NO:95).
  • Figure 22 depicts a hydropathy plot of mouse TANGO 393. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and potential N-glycosylation sites are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:95; SEQ ID NO:96) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:95; SEQ ID NO:97) on the right.
  • mouse TANGO 393 includes an 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:95; SEQ ID NO:96) preceding the mature protein (co ⁇ esponding to amino acid 27 to amino acid 473 of SEQ ID NO:95; SEQ ID NO:97).
  • the molecular weight of mouse TANGO 393 without post-translational modifications is 51.0 kDa prior to the cleavage of the signal peptide, 48.1 kDa after cleavage of the signal peptide.
  • Mouse TANGO 393 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; (3) a cytoplasmic domain; and (4) leucine-rich repeat domain.
  • the mouse TANGO 393 protein contains an extracellular domain at amino acids 27 to 449 of SEQ ID NO:95 (SEQ ID NO: 109), a transmembrane domain at amino acid residues 450 to 467 of SEQ ID NO:75 (SEQ ID NO:98), and a cytoplasmic domain at amino acid residues 468 to 473 of SEQ ED NO:95 (SEQ ID NO:135).
  • a mouse TANGO 393 protein contains a cytoplasmic domain at amino acids 27 to 449 of SEQ ID NO:95, a transmembrane domain at amino acid residues 450 to 467 of SEQ ID NO:75 (SEQ ED NO:98), and an extracellular domain at amino acid residues 468 to 473 of SEQ ED NO:95.
  • nucleotide sequence of mouse TANGO 393 the nucleotide at position 5 is adenine (A)(SEQ ID NO:94).
  • amino acid at position 2 is lysine (K)(SEQ ID NO:95).
  • a species variant of mouse TANGO 393 has a nucleotide at position 5 which is guanine (G)(SEQ ED NO:200).
  • the amino acid at position 2 is arginine (R)(SEQ ID NO:201), i.e., a conservative substitution.
  • nucleotide sequence of mouse TANGO 393 the nucleotide at position 59 is cytosine (C)(SEQ ID NO:94).
  • amino acid at position 20 is alanine (A)(SEQ ID NO:95).
  • a species variant of mouse TANGO 393 has a nucleotide at position 59 which is thymidine (T)(SEQ ID NO:202).
  • the amino acid at position 20 is valine (V)(SEQ ID NO:203), i.e., a conservative substitution.
  • nucleotide sequence of mouse TANGO 393 the nucleotide at position 118 is adenine (A)(SEQ ID NO:94).
  • amino acid at position 40 is threonine (T)(SEQ ID NO:95).
  • a species variant of mouse TANGO 393 has a nucleotide at position 118 which is thymidine (T)(SEQ ID NO:204).
  • the amino acid at position 40 is serine (S)(SEQ ID NO:205), i.e., a conservative substitution.
  • nucleotide sequence of mouse TANGO 393 the nucleotide at position 178 is cytosine (C)(SEQ ID NO:94).
  • amino acid at position 60 is glutamine (Q)(SEQ ID NO:95).
  • a species variant of mouse TANGO 393 has a nucleotide at position 178 which is guanine (G)(SEQ ID NO:206).
  • the amino acid at position 60 is glutamate (E)(SEQ ED NO:207), i.e., a conservative substitution.
  • Mouse TANGO 393 has five N-glycosylation sites. The first has a sequence of NLTI (at amino acids 82-85 of SEQ ID NO:95), the second has a sequence of NLTH (at amino acids 179 to 182 of SEQ ID NO:95), the third has a sequence of NLSM (at amino acids 237 to 240 of SEQ ID NO:95), the fourth has a sequence of NGSG (at amino acids 372 to 375), and the fifth has a sequence of NRTR (at amino acids 423 to 426 of SEQ ID NO:95). Mouse TANGO 393 has one Glycosaminoglycan attachment site, the sequence of which is SGTG (at amino acids 439 to 442 of SEQ ID NO:95).
  • Mouse TANGO 393 has one cAMP- and cGMP-dependent protein kinase phosphorylation site, the sequence of which is KRAS (at amino acids 2 to 5 of SEQ ID NO:95).
  • Mouse TANGO 393 has four protein kinase C phosphorylation sites, where the first has a sequence SQR of (at amino acids 59 to 61 of SEQ ID NO:95), the second has a sequence SCR of (at amino acids 79 to 81 of SEQ ID NO:95), the third has a sequence TFR of (at amino acids 173 to 175 of SEQ ID NO:95), and the fourth has a sequence SRK of (at amino acids 420 to 422 of SEQ ID NO:95).
  • Mouse TANGO 393 has eight casein kinase II phosphorylation sites, where the first has a sequence of TLLE (at amino acids 105 to 108 of SEQ ID NO:95), the second has a sequence of TFRD (at amino acids 173 to 176 of SEQ ID NO:95), the third has a sequence of SVPE (at amino acids 192 to 195 of SEQ ID NO:95), the fourth has a sequence of SSSE (at amino acids 281 to 284 of SEQ ID NO:95), the fifth has a sequence of SDLE (at amino acids 304 to 307 of SEQ ID NO:95), the sixth has a sequence of TDEE (at amino acids 325 to 328 of SEQ ID NO:95), the seventh has a sequence of SVLE (at amino acids 345 to 348 of SEQ ID NO:95), and the eighth has a sequence of SS AE (at amino acids 389 to 392 of SEQ ID NO:95).
  • TLLE at amino acids 105 to 108 of
  • Mouse TANGO 393 has ten N-myristylation sites, where the first has the sequence GACVCY (at amino acids 29 to 34 of SEQ ID NO:95), the second has the sequence GIPAAS (at amino acids 54 to 59 of SEQ ID NO:95), and the third has the sequence GN ISH (at amino acids 66 to 71 of SEQ ED NO:95), the fourth has the sequence GLFRGL (at amino acids 148 to 153 of SEQ ID NO:95), and the fifth has the sequence GCAVAS (at amino acids 308 to 313 of SEQ ED NO:95), the sixth has the sequence GTLPSS (at amino acids 385 to 390 of SEQ ED NO:95), and the seventh has the sequence GLPTTG (at amino acids 407 to 412 of SEQ ID NO:95), the eighth has the sequence GQAGSG (at amino acids 432 to 437 of SEQ ED NO:95), and the ninth has the sequence GTGDAE (at amino acids 440 to 445 of SEQ ED NO:95
  • Mouse TANGO 393 has a prokaryotic membrane lipoprotein lipid attachment site with the sequence of SHVPAASFQSC (at amino acids 70 to 80 of SEQ ID NO:95).
  • Mouse TANGO 393 has a Leucine zipper pattern which has the amino acid sequence LHLDRCGLRELGPGLFRGLAAL (at amino acids 135 to 156 of SEQ ID NO:95).
  • Mouse TANGO 393 has LRR from amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and 260 to 310 of SEQ ID NO:95 (SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively). These repeats are spaced in beta-alpha folds in the structure of the protein, so as to create a hydrophobic face that induces particular folding of the protein.
  • Figure 23 depicts an alignment of the open reading frames of human TANGO 393 (SEQ ID NO:74) and mouse TANGO 393 (SEQ ID NO:94) demonstrating an identity of 82.8%.
  • the algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989)
  • Figure 24 depicts an alignment of the immature proteins of human TANGO 393
  • TANGO 393 nucleic acids Polypeptides. and Modulators Thereof
  • TANGO 393 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of endocrine cells, in particular hypothalamus, cells.
  • TANGO 393 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in hypothalamus development, differentiation and activity, including, but not limited to development, and differentiation and activation of hypothalamus tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types.
  • disorders can include, but are not limited to, malignant or benign hypothalamus cell growth.
  • TANGO 393 nucleic acids, proteins and modulators thereof can be used as a therapeutic agent to treat mammals with abnormal hypothalamic function wherein the mammal exhibits abnormal whole animal homeostasis, appetite-related disorders, obesity, cachexia, food intake disorders, stress responsiveness disorders, adrenal function disorders, pituitary disorders and adrenal disorders.
  • TANGO 393 proteins, nucleic acids, or modulators thereof can be used to treat disorders of the adrenal cortex, such as hypoadrenalism (e.g.
  • neoplasia e.g., adrenal adenoma and cortical carcinoma
  • TANGO 393 polypeptides, nucleic acids, or modulators thereof can be used to treat disorders of the thyroid gland, which is partially regulated by the hypothalamus, such as hyperthyroidism (e.g., diffuse toxic hype ⁇ lasia, toxic multinodular goiter, toxic adenoma, and acute or subacute thyroiditis), hypothyroidism (e.g., cretinism and myxedema), thyroiditis (e.g., Hashimoto's thyroiditis, subacute granulomatous thyroiditis, subacute lymphocytic thyroiditis, Riedel's thryroiditis), Graves' disease, goiter (e.g., simple diffuse goiter and multinodular goiter), or tumors (e.g., adenoma, papillary carcinoma, follicular carcinoma, medullary carcinoma, undifferentiated malignant carcinoma, Hodgkin's disease, and non-Hodgkin'
  • LRR Leucine Rich Repeats
  • TANGO 393 nucleic acids, proteins and modulators thereof can be used to treat subjects with defects in leucine-rich-repeat genes shown to cause various diseases, including but not limited to Bernard-Soulier disease, a bleeding disorder. Further, as TANGO 393 has homology to Platelet Glycoprotein V
  • GMV precursor TANGO 393 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate platelet activity, thrombin activity, von Willebrand Factor assembly and activation, or ADP/epinephrine-, cathepsin G-, and TRAP-induced decrease in platelet surface GPV expression.
  • TANGO 393 proteins, nucleic acids and modulators thereof can be used to modulate the pathogenesis of infectious diseases, for example, diseases that are affected by the expression of leucine-rich-repeat proteins such as the type-1 human immunodeficiency virus (HIV-1) Rev protein, which is the trans-activating region of the virus (Kobe and Deisenhofer, 1994, TEBS, 19:415-421).
  • HIV-1 human immunodeficiency virus
  • Rev protein the trans-activating region of the virus
  • LRR containing proteins are tissue organizers, wherein they orient and order collagen fibrils during ontogeny and are involved in pathological processes such as wound healing, tissue repair, and tumor stroma formation. These properties are rooted in their bifunctional character: the protein moiety binding collagen fibrils at strategic loci, the microscopic gaps between staggered fibrils, and the highly charged glycosaminoglycans extending out to regulate interfibrillar distances and thereby establishing the exact topology of fibrillar collagens in tissues. Therefore, TANGO 393 nucleic acids, proteins and modulators thereof can be used to disrupt intercellular and intracellular protein interactions or cellular signaling in tissues or cells, for example in the hypothalamus.
  • TANGO 393 nucleic acids, proteins and modulators thereof can be used to modulate wound healing (e.g., platelet activation), tissue repair and tumor stroma formation as well.
  • TANGO 393 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate the function of the hypothalamus as it relates to control of endocrine function, regulation of whole animal homeostasis and modulation of diurnal requirements, appetite as related to obesity or cachexia, and generally weight control in mammals. Proteins with LRR also interact with soluble growth factors, modulate their functional activity, and bind to cell surface receptors.
  • TANGO 393 nucleic acids, proteins and modulators thereof can be used to modulate disorders associated with abe ⁇ ant expression of TANGO 393 in cancerous (e.g., tumor) cells that do not normally express TANGO 393.
  • disorders can include, for example, ones associated with tumor cell migration and progression to metastasis.
  • TANGO 393 exhibits homology to the SLIT-1 proteins, TANGO 393 proteins, nucleic acids and modulators thereof may participate in the formation and maintenance of the nervous and endocrine systems by e.g., protein-protein interactions.
  • Northern blot analysis has revealed that the human SLIT-1, -2, and -3 mRNAs are exclusively expressed in the brain, spinal cord, and thyroid, respectively.
  • In situ hybridization studies indicated that the rat SLIT-1 mRNA is specifically expressed in the neurons of fetal and adult forebrains (Itoh et al., Brain Res Mol Brain Res 1998 Nov 20;62(2): 175-86.) This suggests a role for TANGO 393 nucleic acids, proteins and modulators thereof in brain development and neural function.
  • TANGO 393 nucleic acids, proteins and modulators thereof may be useful to disrupt protein interaction or cellular signaling in brain tissues or cells.
  • TANGO 393 protein, nucleic acids and modulators thereof could be useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., multiple sclerosis, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementia.
  • neural related disorders or neural damage such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., multiple sclerosis, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but
  • TANGO 393 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the hypothalamus) and/or cells (e.g., hypothalamic cells) in which TANGO 393 is expressed.
  • TANGO 393 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human TANGO 402 was identified by analyzing the sequences of clones present in a human 9 week fetus library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthga055h07, encoding full-length human TANGO 402.
  • the human TANGO 402 cDNA of this clone is 1348 nucleotides long ( Figure 25; SEQ ID NO:l 10).
  • Figure 26 depicts a hydropathy plot of human TANGO 402. Relatively hydrophobic regions of the protein are above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line.
  • the cysteine residues (cys) and N- glycosylation sites are (Ngly) are indicated by short vertical lines just below the hydropathy trace.
  • the dashed vertical line separates the signal sequence from the mature protein described below.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 402 includes a 50 amino acid signal peptide (amino acid 1 to amino acid 50 of SEQ ID NO:112; SEQ ID NO:114) preceding the mature human TANGO 402 protein (co ⁇ esponding to amino acid 51 to amino acid 207 of SEQ ID NO:112; SEQ ED NO:113).
  • the molecular weight of human TANGO 402 protein without post-translational modifications is 24.0 kDa prior to the cleavage of the signal peptide, 18.1 kDa after cleavage of the signal peptide.
  • Human TANGO 402 protein is a transmembrane protein that contains an extracellular domain at amino acids 1 to 133 of SEQ ID NO:l 12 or a mature extracellular domain at amino acid residues 51 to 133 of SEQ ID NO:l 12 (SEQ ID NO:l 15), a transmembrane domain at amino acid residues 134 to 151 of SEQ ED NO: 112 (SEQ ID NO:l 16), and a cytoplasmic domain at amino acid residues 152 to 207 of SEQ ID NO:l 12 (SEQ ID NO: 117).
  • a human TANGO 402 protein contains a cytoplasmic domain at amino acids 1 to 133 of SEQ ID NO:l 12 or a mature cytoplasmic domain at amino acid residues 51 to 133 of SEQ ID NO:l 12, a transmembrane domain at amino acid residues 134 to 151 of SEQ ID NO:112 (SEQ ID NO: 116), and an extracellular domain at amino acid residues 152 to 207 of SEQ ED NO: 112.
  • nucleotide sequence of human TANGO 402 the nucleotide at position 28 is cytosine (C)(SEQ ID NO: 111).
  • amino acid at position 10 is leucine (L)(SEQ ID NO: 112).
  • a species variant of human TANGO 402 has a nucleotide at position 28 which is guanine (G)(SEQ ID NO:208).
  • the amino acid at position 10 is valine (V)(SEQ ID NO:209), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 402 the nucleotide at position 58 is cytosine (C)(SEQ ID NO:l 11).
  • amino acid at position 20 is glutamine (A)(SEQ ID NO:l 12).
  • a species variant of human TANGO 402 has a nucleotide at position 58 which is guanine (G)(SEQ ID NO:210).
  • the amino acid at position 20 is glutamate (E)(SEQ ID NO:211), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 402 the nucleotide at position 61 is adenine (A)(SEQ ID NO:l 11).
  • amino acid at position 21 is lysine (K)(SEQ ID NO:l 12).
  • a species variant of human TANGO 402 has a nucleotide at position 61 which is guanine (G)(SEQ ED NO:212).
  • amino acid at position 21 is arginine (R)(SEQ ID NO:213), i.e., a conservative substitution.
  • nucleotide sequence of human TANGO 402 the nucleotide at position 64 is thymine (T)(SEQ ID NO: 111).
  • amino acid at position 22 is serine (S)(SEQ ID NO:l 12).
  • a species variant of human TANGO 402 has a nucleotide at position 64 which is adenine (A)(SEQ ID NO:214).
  • the amino acid at position 22 is threonine (T)(SEQ ID NO:215), i.e., a conservative substitution.
  • N-glycosylation sites Two N-glycosylation sites are present in human TANGO 402. The first has the sequence NISS (at amino acid residues 67 to 70 of SEQ ID NO: 112) and the second has the sequence NGTS (at amino acid residues 202 to 205 of SEQ ED NO: 112). Six protein kinase C phosphorylation sites are present in human TANGO 402.
  • the first has the sequence SFK (at amino acid residues 11 to 13 of SEQ ID NO: 112)
  • the second has the sequence SFK (at amino acid residues 95 to 97 of SEQ ED NO: 112)
  • the third has the sequence TWK (at amino acid residues 98 to 100 of SEQ ED NO: 112)
  • the fourth has the sequence SQR (at amino acid residues 102 to 104 of SEQ ID NO: 112)
  • the fifth has the sequence SLK (at amino acid residues 128 to 130 of SEQ ID NO:l 12)
  • the sixth has the sequence TFK (at amino acid residues 188 to 190 of SEQ ID NO:l 12).
  • Three casein kinase II phosphorylation sites are present in human TANGO 402.
  • the first has the sequence TGID (at amino acid residues 49 to 52 of SEQ ID NO: 112)
  • the second has the sequence TWKE (at amino acid residues 98 to 101 of SEQ ID NO:l 12)
  • the third has the sequence SQRD (at amino acid residues 102 to 105 of SEQ ID NO: 112).
  • Human TANGO 402 has a tyrosine kinase phosphorylation site having the sequence KSKDFSLY at amino acid residues 21 to 28 of SEQ ID NO:l 12).
  • Human TANGO 402 has an N- myristylation site having the sequence GLYVTF at amino acid residues 138 to 143 of SEQ ID NO: 112.
  • Human TANGO 402 includes a C-type lectin (CTL)-like domain at amino acid residues 104 to 193 of SEQ ID NO:112 (SEQ ID NO: 118). CTL domains have been shown to function as a calcium-dependent carbohydrate-recognition domain. Human TANGO 402 is homologous to human lectin-like oxidized LDL receptor 1
  • LOX-1 oxidized low-density lipoprotein
  • Ox-LDL oxidized low-density lipoprotein
  • Figure 27 shows an alignment of the human TANGO 402 amino acid sequence (SEQ ID NO:l 12) with the human LOX-1 amino acid sequence (SEQ ID NO:67; Accession Number ABO 10710). As shown in the figure, the amino acid sequence of LOX-1 is 25.1% identical to the amino acid sequence of human TANGO 402 (SEQ ID NO:l 12). As shown in Figure 28.
  • the coding regions of the human TANGO 402 nucleic acid sequence (SEQ ID NO:56) and LOX-1 nucleic acid sequence (SEQ ID NO:66) are 42.0 % identical.
  • the overall nucleic acid sequence identity between full-length human TANGO 402 (SEQ ID NO: 110) and full-length LOX-1 (SEQ ID NO:65) is 44.0 %.
  • Clone EpT402 which encodes human TANGO 402 was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-294. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Pu ⁇ oses of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. ⁇ 112.
  • TANGO 402 Nucleic acids. Polypeptides. and Modulators Thereof
  • TANGO 402 was originally found in a human fetal library, TANGO 402 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders associated with cells, tissues, and/or organs in the embryo or fetus, or modulate the proliferation, development, differentiation, and/or function of cells, tissues, and/or organs in the embryo or fetus.
  • TANGO 402 is homologous to LOX-1
  • TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders, modulate development, differentiation, proliferation and/or activity of immune cells, such as macrophages and endothelial cells, e.g., the phagocytosis of aged/apoptotic cells by endothelial cells.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat, inhibit and/or prevent disorders and diseases associated with the abe ⁇ ant activity of the cells, tissues or organs in which TANGO 402 is expressed, e.g. endothelial activity.
  • TANGO 402 nucleic acids, proteins and modulations thereof can also be used to diagnose disorders and/or modulate symptoms associated with atherosclerosis (e.g., atherosclerotic cardiovascular disease) and Alzheimer's disease.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to diagnose disorders associated with host immune defenses and/or modulate host immune defenses, e.g., modulating the activation of macrophages.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat and/or prevent obesity, diabetes, and inflammatory disorders (e.g. , asthma, arthritis, multiple sclerosis, allergies, hepatitis and infections).
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate TANGO 402 biological activities, which include, e.g., (1) the ability to bind proteins, e.g., lipoproteins, e.g., low- density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (2) the ability to modulate internalization of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (3) the ability to modulate degradation, e.g., proteolytic degradation, of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (4) the ability to modulate, e.g., increase, uptake
  • proteins e.g., lipoproteins,
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to modulate, e.g., prevent, lipid deposition, e.g., in arteries, and thus modulate, e.g., prevent, intimal thickening; (2) the ability to modulate, e.g., induce or prevent, changes in cells, e.g., transformation of cells (e.g., macrophages and smooth muscle cells) into foam cells and functional alteration of cells (e.g., endothelial cells, e.g., intimal neovascular endothelial cells); (3) the ability to bind and phagocytose cells, e.g., aged and apoptotic cells; and (4) the ability to remove debris, e.g., apoptotic cells, from blood vessel walls.
  • modulate e.g., prevent, lipid deposition, e.g., in arteries
  • modulate e
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to modulate homeostasis, e.g., vascular homeostasis, e.g., by modulating, e.g., preventing the impairment of, nitric oxide production; (2) the ability to modulate, e.g., inhibit, the expression of molecules, e.g., adhesion molecules (e.g., leukocyte adhesion molecules) and growth factors (e.g., smooth- muscle growth factors); (3) the ability to alter, e.g., increase, expression in response to stimuli, e.g. , TNF, shear stress, and pathophysiological stimuli relevant to disorders (e.g. , atherosclerosis and inflammation).
  • homeostasis e.g., vascular homeostasis
  • modulators thereof can be used to modulate e.g., (1) the ability to modulate homeostasis, e.g.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to form, e.g., stabilize, promote, facilitate, inhibit, or disrupt, cell-extracellular matrix interactions, e.g., adhesion between cells and extracellular matrix; (2) the ability to form, e.g., stabilize, promote, facilitate, inhibit, or disrupt, cell to cell and cell to blood product interaction, e.g. , between leukocytes and platelets or leukocytes and vascular endothelial cells; and (3) the ability to recognize large molecules, e.g., carbohydrates.
  • TANGO 402 is homologous to LOX-1
  • nucleic acids, proteins and modulators thereof have biological activities that can also include the ability to perform one or more of the functions of LOX-1 described, for example, in the following: Sawamura et al. (1997) Nature. 386:73-77; Kataoka et al. (1999) Circulation. 99:3110-3117; and Kita (1999) Circulation Research. 84:1113-1115, the contents of each of which is inco ⁇ orated herein by reference in its entirety.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate or treat disorders in which LOX-1 plays a role, some of which are described in the following references: Sawamura et al. (1997) Nature. 386:73-77; Kataoka et al. (1999) Circulation. 99:3110-3117; and Kita (1999) Circulation Research. 84:1113-1115, the contents of each of which is inco ⁇ orated herein by reference in its entirety.
  • TANGO 402 nucleic acids, proteins and modulators thereof can modulate or treat atherosclerosis, e.g., by binding to oxidatively modified low density lipoprotein (Ox-LDL) and its lipid constituents, thus preventing lipid deposition and intimal thickening in the arteries, and thus preventing the induction of endothelial expression of leukocyte adhesion molecules and smooth-muscle growth factors (both which are implicated in atherogenesis).
  • Ox-LDL oxidatively modified low density lipoprotein
  • TANGO 402 nucleic acids, proteins and modulators thereof modulate or treat immune related diseases and disorders.
  • LOX-1 is implicated in inflammation, and as LOX-1 has highest homology with the NKR-Pl family of proteins, which are involved in target-cell recognition and natural killer cell activation
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate or treat inflammatory disorders such as bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, multiple sclerosis, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis), and processes.
  • inflammatory disorders such as bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, multiple sclerosis, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to identify, diagnose and/or modulate or treat immune disorders including, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), and T cell autoimmune disorders (e.g., AIDS)) and inflammatory disorders.
  • immune disorders including, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), and T cell autoimmune disorders (e.g., AIDS)
  • autoimmune disorders e.g., arthritis, graft rejection (e.g., allograft rejection)
  • T cell autoimmune disorders e.g., AIDS
  • TANGO 402 nucleic acids, proteins and modulators thereof be used to identify, diagnose and/or modulate or treat TNF-related disorders, as LOX-1 expression is induced by tumor necrosis factors.
  • TNF-related disorders include, e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer).
  • TANGO 402 can also play a role in treating hypertension and congestive heart failure.
  • TANGO 402 has C-type lectin domains or C-type lectin-like domains, and is similar in that respect to the selectins, which are implicated in cell-cell recognition (including endothelial-leukocyte adhesion), TANGO 402 nucleic acids, proteins and modulators thereof can be used to identify, diagnose and/or modulate or treat cell adhesion or cell migration/motility related disorders.
  • disorders associated with adhesion and migration of cells include, e.g., disorders associated with adhesion and migration of cells, e.g., platelet aggregation disorders (e.g., Glanzmarm's thromboasthemia, which is a bleeding disorders characterized by failure of platelet aggregation in response to cell stimuli), inflammatory disorders (e.g., leukocyte adhesion deficiency, which is a disorder associated with impaired migration of neutrophils to sites of extravascular inflammation), disorders associated with abnormal tissue migration during embryo development, and tumor metastasis.
  • platelet aggregation disorders e.g., Glanzmarm's thromboasthemia, which is a bleeding disorders characterized by failure of platelet aggregation in response to cell stimuli
  • inflammatory disorders e.g., leukocyte adhesion deficiency, which is a disorder associated with impaired migration of neutrophils to sites of extravascular inflammation
  • disorders associated with abnormal tissue migration during embryo development e.g.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be used to diagnose C-type lectin disorders and/or modulate calcium-dependent carbohydrate recognition.
  • TANGO 402 proteins exhibit homology to lectins.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to modulate cell-cell, cell-extracellular matrix (ECM) interactions, cell adhesion, cell migration and cell signaling.
  • TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat and/or prevent disorders and diseases associated with abe ⁇ ant cell-cell, cell-ECM interactions, cell migration, cell adhesion and cell-signaling, as well as treating and preventing tumor cell metastasis.
  • TANGO 402 nucleic acids, proteins and modulators thereof can also be utilized to treat and/or prevent the migration of cancerous and precancerous cells (e.g., tumor migration).
  • TANGO 402 nucleic acids, proteins and modulators thereof can also be used to modulate cell proliferation, e.g., abnormal cell proliferation. Such modulation may, for example, be via modulation of one or more elements involved in signal transduction cascades.
  • TANGO 402 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., fetal tissues) and/or cells (e.g., fetal cells) in which TANGO 402 is expressed.
  • TANGO 402 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • Human MANGO 346 A MANGO 346 cDNA was identified from clones present in a human brain library among sequences that encode signal peptides. This analysis led to the identification of a clone, jlhbab575g04, encoding full-length human MANGO 346.
  • the human MANGO 346 cDNA of this clone is 1196 nucleotides long ( Figure 29; SEQ ID NO: 123).
  • Figure 30 depicts a hydropathy plot of human MANGO 346. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 19 of SEQ ID NO:125; SEQ ID NO: 126) on the left from the mature protein (amino acids 20 to 60 of SEQ ID NO: 125; SEQ ID NO: 127) on the right.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human MANGO 346 includes a 19 amino acid signal peptide (amino acid 1 to amino acid 19 of SEQ ID NO:125; SEQ ID NO:126) preceding the mature human protein (co ⁇ esponding to amino acid 20 to amino acid 60 of SEQ ED NO: 125; SEQ ID NO: 127).
  • the molecular weight of protein without post-translational modifications is 7.1 kDa prior to the cleavage of the signal peptide, 5.0 kDa after cleavage of the signal peptide.
  • the nucleotide at position 13 is cytosine (C)(SEQ ID NO:124).
  • the amino acid at position 5 is leucine (L)(SEQ ID NO: 125).
  • a species variant of human MANGO 346 has a nucleotide at position 13 which is adenine (A)(SEQ ID NO:216).
  • the amino acid at position 5 is isoleucine (I)(SEQ ID NO:217), i.e., a conservative substitution.
  • the nucleotide at position 59 is adenine (A)(SEQ ID NO: 124).
  • the amino acid at position 20 is tyrosine (Y)(SEQ ID NO: 125).
  • a species variant of human MANGO 346 has a nucleotide at position 59 which is thymidine (T)(SEQ ID NO:218).
  • the amino acid at position 20 is phenylalanine (F)(SEQ ID NO:219), i.e., a conservative substitution.
  • the nucleotide at position 61 is thymidine (T)(SEQ ID NO: 124).
  • the amino acid at position 21 is serine (S)(SEQ ID NO: 125).
  • a species variant of human MANGO 346 has a nucleotide at position 61 which is adenine (A)(SEQ ID NO:220).
  • the amino acid at position 21 is threonine (T)(SEQ ID NO:221), i.e., a conservative substitution.
  • the nucleotide at position 80 is guanine (G)(SEQ ID NO: 124).
  • the amino acid at position 27 is arginine (R)(SEQ ED NO: 125).
  • a species variant of human MANGO 346 has a nucleotide at position 80 which is adenine (A)(SEQ ID NO:222).
  • the amino acid at position 27 is lysine (K)(SEQ ID NO:223), i.e., a conservative substitution.
  • MANGO 346 has three Casein Kinase II phosphorylation sites. The first has the sequence SFLE (at amino acids 21 to 24 of SEQ ED NO: 125), the second has the sequence TIKE (at amino acids 44 to 47 of SEQ ID NO: 125) and the third has the sequence TYYD (at amino acids 51 to 54 of SEQ ID NO: 125). Human MANGO 346 has one prokaryotic membrane lipoprotein lipid attachment site. The sequence is CILPLLLLASC (at amino acids 6 to 16 of SEQ ID NO:125).
  • nucleic acids, proteins, and modulators thereof can be used to diagnose or identify disorders and/or modulate the proliferation, development, differentiation, and/or function of neural organs, e.g., neural tissues and cells, e.g., cells of the central nervous system, e.g., cells of the peripheral nervous system.
  • neural organs e.g., neural tissues and cells, e.g., cells of the central nervous system, e.g., cells of the peripheral nervous system.
  • MANGO 346 nucleic acids, proteins, and modulators thereof can also be used to diagnose or identify disorders and/or modulate symptoms associated with abnormal neural signaling and function, e.g., epilepsy, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
  • abnormal neural signaling and function e.g., epilepsy, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
  • MANGO 346 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to diagnose disorders, regulate or modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs.
  • MANGO 346 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to regulate or diagnose disorders, modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs.
  • the TANGO 346 proteins can be used to disrupt protein interaction or cellular signaling in brain tissues or cells.
  • TANGO 346 proteins are useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g.
  • Alzheimer's disease Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
  • MANGO 346 is a secreted protein and thus likely a signaling molecule
  • nucleic acids, proteins or modulators thereof can be used to modulate MANGO 346 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g. , homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
  • protein-protein interactions e.g. , homophilic and/or heterophilic
  • protein-ligand interactions e.g., in receptor-ligand recognition
  • intracellular signaling cascades e.g., signal transduction cascades
  • MANGO 346 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the brain) and/or cells (e.g., neurons) in which MANGO 346 is expressed.
  • MANGO 346 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
  • a cDNA encoding human MANGO 349 was identified by analyzing the sequences of clones present in a human brain library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jlhbae318gd08, encoding full-length human MANGO 349.
  • the human cDNA of this clone is 3649 nucleotides long ( Figure 31; SEQ ID NO:128).
  • Figure 32 depicts a hydropathy plot of human MANGO 349. Relatively hydrophobic regions of the protein are above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence from the mature protein described below.
  • the signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10: 1-6) predicted that human MANGO 349 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:130; SEQ ID NO:131) preceding the mature human protein (co ⁇ esponding to amino acid 27 to amino acid 167 of SEQ ID NO:130; SEQ ED NO:132).
  • the molecular weight of human protein without post- translational modifications is 17.6 kDa prior to the cleavage of the signal peptide, 15.1 kDa after cleavage of the signal peptide.
  • the nucleotide at position 4 is adenine (A)(SEQ ID NO: 129).
  • the amino acid at position 2 is threonine (T)(SEQ ID NO: 130).
  • a species variant of human MANGO 349 has a nucleotide at position 4 which is thymine (T)(SEQ ID NO:224).
  • the amino acid at position 2 is serine (S)(SEQ ID NO:225), i.e., a conservative substitution.
  • the nucleotide at position 61 is adenine (A)(SEQ ED NO: 129).
  • the amino acid at position 21 is isoleucine (I)(SEQ ED NO: 130).
  • a species variant of human MANGO 349 has a nucleotide at position 61 which is cytosine (C)(SEQ ID NO:226).
  • the amino acid at position 21 is leucine (L)(SEQ ID NO:227), i.e., a conservative substitution.
  • the nucleotide at position 86 is guanine (G)(SEQ ID NO: 129).
  • the amino acid at position 29 is arginine (R)(SEQ ID NO: 130).
  • a species variant of human MANGO 349 has a nucleotide at position 86 which is adenine (A)(SEQ ID NO:228).
  • the amino acid at position 29 is lysine (K)(SEQ ID NO:229), i.e., a conservative substitution.
  • the nucleotide at position 123 is guanine (G)(SEQ ID NO: 129).
  • the amino acid at position 41 is glutamate (E)(SEQ ID NO: 130).
  • a species variant of human MANGO 349 has a nucleotide at position 123 which is cytosine (C)(SEQ ID NO:230).
  • the amino acid at position 41 is aspartate (D)(SEQ ID NO:231), i.e., a conservative substitution.
  • Two Protein C Kinase phosphorylation sites are present in human MANGO 349.
  • the first has the sequence SLK (at amino acids 136 to 139 of SEQ ID NO: 130) and the second has the sequence SGR (at amino acids 152 to 154 of SEQ ID NO: 130).
  • Two casein kinase II phosphorylation sites are present in human MANGO 349.
  • the first has the sequence SGTE (at amino acids 38 to 41 of SEQ ID NO: 130), and the second has the sequence SGRE (at amino acids 152 to 155 of SEQ ID NO: 130).
  • Human MANGO 349 has four N-myristylation sites.
  • the first has the sequence GGILAT (at amino acids 10 to 15 of SEQ ID NO: 130), the second has the sequence GTEVAD (at amino acids 39 to 44 of SEQ ID NO: 130), the third has the sequence GVAASH (at amino acids 89 to 94 of SEQ ID NO: 17), and the fourth has the sequence GGPPSL (at amino acids 132 to 137 of SEQ ID NO: 130).
  • Clone EpM349 which encodes human MANGO 349, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-295. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Pu ⁇ oses of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. ⁇ 112.
  • nucleic acids, proteins, and modulators thereof can be used to diagnose or identify disorders and/or modulate the proliferation, development, differentiation, and/or function of neural organs, e.g., neural tissues and cells, e.g., cells of the central nervous system, e.g., cells of the peripheral nervous system.
  • MANGO 349 nucleic acids, proteins, and modulators thereof can also be used to diagnose or identify disorders and/or modulate symptoms associated with abnormal neural signaling and function, e.g., epilepsy, stroke, traumatic injury, etc.
  • MANGO 349 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to diagnose disorders, regulate or modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs.
  • the TANGO 349 proteins can be used to disrupt protein interaction or cellular signaling in brain tissues or cells.
  • TANGO 349 proteins could be useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
  • neural related disorders or neural damage such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
  • MANGO 349 is a secreted protein and thus likely a signaling molecular
  • nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate MANGO 349 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g. signal transduction cascades).
  • protein-protein interactions e.g., homophilic and/or heterophilic
  • protein-ligand interactions e.g. in receptor- ligand recognition
  • intracellular signaling cascades e.g. signal transduction cascades
  • MANGO 349 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g. , the brain) and/or cells (e.g. , neurons) in which MANGO 349 is expressed.
  • MANGO 349 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g. , in radiation hybrid mapping.
  • Tables 1 provides a summary of the sequence information for TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, human TANGO 393, mouse TANGO 393, TANGO 402, MANGO 346, and MANGO 349.
  • Table 2 provides a summary of the domains of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, human TANGO 393, mouse TANGO 393, TANGO 402, MANGO 346, and MANGO 349. It is noted that human and mouse TANGO 393 leucine-rich repeats are not included in Table 2, but are described supra.
  • nucleic acid molecules that encode a polypeptide of the invention or a biologically active portion thereof, as well as nucleic acid molecules sufficient for use as hybridization probes to identify nucleic acid molecules encoding a polypeptide of the invention and fragments of such nucleic acid molecules suitable for use as PCR primers for the amplification or mutation of nucleic acid molecules.
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • the nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • an “isolated” nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule.
  • an “isolated” nucleic acid molecule is free of sequences (preferably protein encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated nucleic acid molecule can contain less than about 5 kB, 4 kB, 3 kB, 2 kB, 1 kB, 0.5 kB or 0.1 kB of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the term "isolated"when referring to a nucleic acid molecule does not include an isolated chromosome.
  • nucleic acid molecule is a cDNA or RNA, e.g., mRNA, molecule
  • such molecules can include a poly A "tail", or, alternatively, can lack such a 3' tail.
  • tail sequences may be depicted herein with such tail sequences, it is to be understood that cDNA nucleic acid molecules of the invention are also intended to include such sequences lacking the depicted poly A tails.
  • a nucleic acid molecule of the present invention e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230 or a complement thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein.
  • nucleic acid molecules of the invention can be isolated using standard hybridization and cloning techniques (e.g. , as described in Sambrook et al.
  • a nucleic acid molecule of the invention can be amplified using cDNA, mRNA or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques.
  • the nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of the nucleotide sequence of SEQ ED NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone
  • a nucleic acid molecule which is complementary to a given nucleotide sequence is one which is sufficiently complementary to the given nucleotide sequence that it can hybridize to the given nucleotide sequence under the conditions set forth herein, thereby forming a stable duplex.
  • a nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence encoding a full length polypeptide of the invention for example, a fragment which can be used as a probe or primer or a fragment encoding a biologically active portion of a polypeptide of the invention.
  • the nucleotide sequence determined from the cloning one gene allows for the generation of probes and primers designed for use in identifying and/or cloning homologs in other cell types, e.g. , from other tissues, as well as homologs from other mammals.
  • the probe/primer typically comprises substantially purified oligonucleotide.
  • the oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, preferably about 25, more preferably about 50, 75, 100, 125, 150, 175, 200, 250, 300, 350 or 400 consecutive nucleotides of the sense or anti-sense sequence of SEQ ID NO: 1 , 2, 27, 28,
  • the oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least 400, preferably 450, 500, 530, 550, 600, 700,800, 900, 1000 or 1150 consecutive oligonucleotides of the sense or antisense sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180
  • Probes based on the sequence of a nucleic acid molecule of the invention can be used to detect transcripts or genomic sequences encoding the same protein molecule encoded by a selected nucleic acid molecule.
  • the probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • Such probes can be used as part of a diagnostic test kit for identifying cells or tissues which mis-express the protein, such as by measuring levels of a nucleic acid molecule encoding the protein in a sample of cells from a subject, e.g., detecting mRNA levels or determining whether a gene encoding the protein has been mutated or deleted.
  • a nucleic acid fragment encoding a biologically active portion of a polypeptide of the invention can be prepared by isolating a portion of any of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, expressing the encoded portion of
  • the invention further encompasses nucleic acid molecules that differ from the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA
  • TANGO 393 has been mapped to chromosome 22, and therefore TANGO 393 family members can include nucleotide sequence polymorphisms (e.g., nucleotide sequences that vary from SEQ ID NO:73 and SEQ ID NO:74) that map to this chromosome 22 region, and such sequences represent TANGO 393 allelic variants.
  • TANGO 339 has been mapped to chromosome 10, and therefore TANGO 339, family members can include nucleotide sequence polymorphisms (e.g., nucleotide sequences that vary from SEQ ID NO:l and SEQ ID NO:2) that map to this chromosome 10 region, and such sequences represent allelic variants.
  • allelic variant refers to a nucleotide sequence which occurs at a given locus or to a polypeptide encoded by the nucleotide sequence.
  • the terms “gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding a polypeptide of the invention.
  • allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene.
  • Alternative alleles can be identified by sequencing the gene of interest in a number of different individuals. This can be readily carried out by using hybridization probes to identify the same genetic locus in a variety of individuals. Any and all such nucleotide variations and resulting amino acid polymorphisms or variations that are the result of natural allelic variation and that do not alter the functional activity are intended to be within the scope of the invention.
  • polymorphisms that are associated with a particular disease and/or disorder are used as markers to diagnose said disease or disorder.
  • polymorphisms are used as a marker to diagnose abnormal coronary function such as atherosclerosis.
  • nucleic acid molecules, encoding proteins of the invention from other species which have a nucleotide sequence which differs from that of the human or mouse protein described herein are intended to be within the scope of the invention.
  • Nucleic acid molecules corresponding to natural allelic variants and homologs of a cDNA of the invention can be isolated based on their identity to the human nucleic acid molecule disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
  • a cDNA encoding a soluble form of a membrane-bound protein of the invention isolated based on its hybridization to a nucleic acid molecule encoding all or part of the membrane-bound form.
  • a cDNA encoding a membrane-bound form can be isolated based on its hybridization to a nucleic acid molecule encoding all or part of the soluble form.
  • an isolated nucleic acid molecule of the invention is at least 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence, preferably the coding sequence, of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123, or 128 or a complement thereof.
  • an isolated nucleic acid molecule of the invention is at least 20, 50, 100, 200, 300, 400, 500, 600, 700, 800 or 900 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence, preferably the coding sequence, of SEQ ID NO:2, 28, 37, 45, 53, 59, 64, 74, 94, 111, 124, or, 129, or a complement thereof.
  • hybridizes under stringent conditions is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% (65%, 70%, preferably 75%) identical to each other typically remain hybridized to each other.
  • stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • a preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45° C, followed by one or more washes in 0.2 X SSC, 0.1% SDS at 50-65° C.
  • an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or a complement thereof, corresponds to a naturally-occurring nucleic acid molecule.
  • a "naturallyceridel" a
  • allelic variants of a nucleic acid molecule of the invention sequence that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein. For example, one can make nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues.
  • a "non-essential” amino acid residue is a residue that can be altered from the wild-type sequence without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity.
  • amino acid residues that are not conserved or only semi-conserved among homologs of various species may be non-essential for activity and thus would be likely targets for alteration.
  • Specific examples of conservative amino acid alterations from the original sequence are shown in SEQ ID NOs:137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231.
  • amino acid residues that are conserved among the orthologs of various species may be essential for activity and thus would not be likely targets for alteration.
  • another aspect of the invention pertains to nucleic acid molecules encoding a polypeptide of the invention that contain changes in amino acid residues that are not essential for activity.
  • Such polypeptides differ in amino acid sequence from SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, yet retain biological activity.
  • the isolated nucleic acid molecule includes a nucleotide sequence encoding a protein that includes an amino acid sequence that is at least about 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%), 85%o, 95%), or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231.
  • An isolated nucleic acid molecule encoding a variant protein can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ED NO: 1, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or complement thereof, such that one or more amino acid substitutions
  • Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid, asparagine, glutamine
  • uncharged polar side chains e.g., glycine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity.
  • the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
  • a mutant polypeptide that is a variant of a polypeptide of the invention can be assayed for: (1) the ability to form protein-protein interactions with proteins in a signaling pathway of the polypeptide of the invention such as in central nervous system cells, lymphoid cells, hypothalamus cells, or prostate cells with the proteins encoded by the genes of the present invention (e.g., leucine-rich repeat interactions and transmembrane 4); (2) the ability to bind a ligand of the polypeptide of the invention (i.e., in transmembrane proteins of the invention or alternatively, secreted proteins which are the ligand for a cellular receptor); or (3) the ability to bind to an intracellular target protein of the polypeptide of the invention.
  • the mutant polypeptide can be assayed for the ability to modulate cellular proliferation, cellular migration or chemotaxis, or cellular differentiation.
  • the present invention encompasses antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid encoding a polypeptide of the invention, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid.
  • the antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof, e.g., all or part of the protein coding region (or open reading frame).
  • An antisense nucleic acid molecule can be antisense to all or part of a non-coding region of the coding strand of a nucleotide sequence encoding a polypeptide of the invention.
  • the non-coding regions (“5 1 and 3' untranslated regions") are the 5' and 3' sequences which flank the coding region and are not translated into amino acids.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35,
  • An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycar
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a selected polypeptide of the invention to thereby inhibit expression, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site.
  • antisense nucleic acid molecules can be modified to target selected cells and then administered systemically.
  • antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g.
  • antisense nucleic acid molecules by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens.
  • the antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
  • An antisense nucleic acid molecule of the invention can be an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids Res. 15:6625-6641).
  • the antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1981) FEBS Lett. 215:327-330).
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)
  • a ribozyme having specificity for a nucleic acid molecule encoding a polypeptide of the invention can be designed based upon the nucleotide sequence of a cDNA disclosed herein.
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a Cech et al. U.S. Patent No. 4,987,071; and Cech et al. U.S. Patent No. 5,116,742.
  • an mRNA encoding a polypeptide of the invention can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel and Szostak (1993) Science 261 :1411-1418.
  • the invention also encompasses nucleic acid molecules which form triple helical structures.
  • expression of a polypeptide of the invention can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the gene encoding the polypeptide (e.g. , the promoter and/or enhancer) to form triple helical structures that prevent transcription of the gene in target cells.
  • nucleotide sequences complementary to the regulatory region of the gene encoding the polypeptide e.g. , the promoter and/or enhancer
  • the nucleic acid molecules of the invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g. , the stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids (see Hyrup et al. (1996) Bioorganic & Medicinal Chemistry 4(1): 5-23).
  • peptide nucleic acids refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
  • the neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
  • the synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675.
  • PNAs can be used in therapeutic and diagnostic applications.
  • PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication.
  • PNAs can also be used, e.g., in the analysis of single base pair mutations in a gene by, e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., SI nucleases (Hyrup (1996), supra; or as probes or primers for DNA sequence and hybridization (Hyrup (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675).
  • PNAs can be modified, e.g., to enhance their stability or cellular uptake, by attaching Hpophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art.
  • PNA-DNA chimeras can be generated which may combine the advantageous properties of PNA and DNA.
  • Such chimeras allow DNA recognition enzymes, e.g., RNAse H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
  • PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup (1996), supra).
  • the synthesis of PNA-DNA chimeras can be performed as described in Hyrup (1996), supra, and Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63.
  • a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs.
  • the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo ), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Acad. Sci. USA 84:648-652; PCT Publication No. W0 88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. W0 89/10134).
  • peptides e.g., for targeting host cell receptors in vivo
  • agents facilitating transport across the cell membrane see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl
  • oligonucleotides can be modified with hybridization-triggered cleavage agents (see, e.g., Krol et al. (1988) Bio/Techniques 6:958-976) or intercalating agents (see, e.g., Zon (1988) Pharm. Res. 5:539-549).
  • the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
  • isolated proteins and biologically active portions thereof as well as polypeptide fragments suitable for use as immunogens to raise antibodies directed against a polypeptide of the invention.
  • the native polypeptide can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques.
  • polypeptides of the invention are produced by recombinant DNA techniques.
  • a polypeptide of the invention can be synthesized chemically using standard peptide synthesis techniques.
  • an “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the language “substantially free of cellular material” includes preparations of protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%, or 5%> (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein").
  • the protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%>, 10%, or 5% of the volume of the protein preparation.
  • culture medium represents less than about 20%>, 10%, or 5% of the volume of the protein preparation.
  • the protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the protein have less than about 30%, 20%), 10%), 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest.
  • Biologically active portions of a polypeptide of the invention include polypeptides comprising amino acid sequences sufficiently identical to or derived from the amino acid sequence of the protein (e.g., the amino acid sequence shown in any of SEQ ID NO:6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 32, 33, 34, 41, 42, 43, 49, 50, 51, 68, 69, 70, 71, 72, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 115, 116, 117, 118, 119, 133, 134, 135, or 136, which include fewer amino acids than the full length protein, and exhibit at least one activity of the corresponding full-length protein.
  • biologically active portions comprise a domain or motif with at least one activity of the co ⁇ esponding protein.
  • a biologically active portion of a protein of the invention can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length.
  • other biologically active portions, in which other regions of the protein are deleted can be prepared by recombinant techniques and evaluated for one or more of the functional activities of the native form of a polypeptide of the invention.
  • Prefe ⁇ ed polypeptides have the amino acid sequence of SEQ ID NO:6, 7, 8, 9, 10, 19, 20, 21, 22 or 23.
  • Other useful proteins are substantially identical (e.g., at least about 45%, preferably 55%, 65%, 75%, 85%, 95%, or 99%) to any of SEQ ID NO:6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 32, 33, 34, 41, 42, 43, 49, 50, 51, 68, 69, 70, 71, 72, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 115, 116, 117, 118, 119, 133, 134, 135, or 136, and retain the functional activity of the protein of the co ⁇ esponding naturally-occurring protein yet differ in amino acid sequence due
  • the sequences are aligned for optimal comparison purposes (e.g. , gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence).
  • the amino acid residues or nucleotides at co ⁇ esponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the co ⁇ esponding position in the second sequence, then the molecules are identical at that position.
  • the determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • a prefe ⁇ ed, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877.
  • Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402.
  • PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.).
  • FASTA parameters see http://bioweb.pasteur.fr/docs/man man/fasta. I.html#sect2, the contents of which are inco ⁇ orated herein by reference.
  • the percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, only exact matches are counted.
  • a "chimeric protein” or “fusion protein” comprises all or part (preferably biologically active) of a polypeptide of the invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the same polypeptide of the invention).
  • a heterologous polypeptide i.e., a polypeptide other than the same polypeptide of the invention.
  • the term "operably linked” is intended to indicate that the polypeptide of the invention and the heterologous polypeptide are fused in-frame to each other.
  • the heterologous polypeptide can be fused to the N-terminus or C-terminus of the polypeptide of the invention.
  • One useful fusion protein is a GST fusion protein in which the polypeptide of the invention is fused to the C-terminus of GST sequences. Such fusion proteins can facilitate the purification of a recombinant polypeptide of the invention.
  • the fusion protein contains a heterologous signal sequence at its N-terminus.
  • the native signal sequence of a polypeptide of the invention can be removed and replaced with a signal sequence from another protein.
  • the gp67 secretory sequence of the baculovirus envelope protein can be used as a heterologous signal sequence (Current Protocols in Molecular Biology, Ausubel et al., eds., John Wiley & Sons, 1992).
  • Other examples of eukaryotic heterologous signal sequences include the secretory sequences of melittin and human placental alkaline phosphatase (Stratagene; La Jolla, California).
  • useful prokaryotic heterologous signal sequences include the phoA secretory signal (Sambrook et al., supra) and the protein A secretory signal (Pharmacia Biotech; Piscataway, New Jersey).
  • the fusion protein is an immunoglobulin fusion protein in which all or part of a polypeptide of the invention is fused to sequences derived from a member of the immunoglobulin protein family.
  • the immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a ligand (soluble or membrane-bound) and a protein on the surface of a cell (receptor), to thereby suppress signal transduction in vivo.
  • the immunoglobulin fusion protein can be used to affect the bioavailability of a cognate ligand of a polypeptide of the invention.
  • Inhibition of ligand/receptor interaction may be useful therapeutically, both for treating proliferative and differentiative disorders and for modulating (e.g. , promoting or inhibiting) cell survival.
  • the immunoglobulin fusion proteins of the invention can be used as immunogens to produce antibodies directed against a polypeptide of the invention in a subject, to purify ligands and in screening assays to identify molecules which inhibit the interaction of receptors with ligands.
  • Chimeric and fusion proteins of the invention can be produced by standard recombinant DNA techniques.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel et al., supra).
  • anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence
  • many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide).
  • a nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide of the invention.
  • a signal sequence of a polypeptide of the invention can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest.
  • Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein during secretion in one or more cleavage events.
  • Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway.
  • a nucleic acid sequence encoding a signal sequence of the invention can be operably linked in an expression vector to a protein of interest, such as a protein which is ordinarily not secreted or is otherwise difficult to isolate.
  • the signal sequence directs secretion of the protein, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence is subsequently or concu ⁇ ently cleaved.
  • the protein can then be readily purified from the extracellular medium by art recognized methods.
  • the signal sequence can be linked to the protein of interest using a sequence which facilitates purification, such as with a GST domain.
  • the signal sequences of the present invention can be used to identify regulatory sequences, e.g., promoters, enhancers, repressors. Since signal sequences are the most amino-terminal sequences of a peptide, it is expected that the nucleic acids which flank the signal sequence on its amino-terminal side will be regulatory sequences which affect transcription. Thus, a nucleotide sequence which encodes all or a portion of a signal sequence can be used as a probe to identify and isolate signal sequences and their flanking regions, and these flanking regions can be studied to identify regulatory elements therein.
  • regulatory sequences e.g., promoters, enhancers, repressors.
  • the present invention also pertains to variants of the polypeptides of the invention.
  • variants have an altered amino acid sequence which can function as either agonists (mimetics) or as antagonists.
  • Variants can be generated by mutagenesis, e.g. , discrete point mutation or truncation.
  • An agonist can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of the protein.
  • An antagonist of a protein can inhibit one or more of the activities of the naturally occurring form of the protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the protein of interest.
  • specific biological effects can be elicited by treatment with a variant of limited function. Treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein can have fewer side effects in a subject relative to treatment with the naturally occurring form of the protein.
  • Variants of a protein of the invention which function as either agonists (mimetics) or as antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the protein of the invention for agonist or antagonist activity.
  • a variegated library of variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display).
  • methods which can be used to produce libraries of potential variants of the polypeptides of the invention from a degenerate oligonucleotide sequence. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang (1983) Tetrahedron 39:3; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477).
  • libraries of fragments of the coding sequence of a polypeptide of the invention can be used to generate a variegated population of polypeptides for screening and subsequent selection of variants.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of the coding sequence of interest with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with SI nuclease, and ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which encodes N-terminal and internal fragments of various sizes of the protein of interest.
  • REM Recursive ensemble mutagenesis
  • polypeptides of the invention can exhibit post-translational modifications, including, but not limited to glycosylations, (e.g., N-linked or O-linked glycosylations), myristylations, palmitylations, acetylations and phosphorylations (e.g., serine/threonine or tyrosine).
  • glycosylations e.g., N-linked or O-linked glycosylations
  • myristylations e.g., palmitylations
  • acetylations e.g., serine/threonine or tyrosine
  • the TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 polypeptides of the invention exhibit reduced levels of O-linked glycosylation and/or N-linked glycosylation relative to endogenously expressed TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, MANGO 349 polypeptides.
  • the TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 polypeptides of the invention do not exhibit O-linked glycosylation or N-linked glycosylation.
  • polypeptides of the invention can, for example, include modifications that can increase such attributes as stability, half-life, ability to enter cells and aid in administration, e.g., in vivo administration of the polypeptides of the invention.
  • polypeptides of the invention can comprise a protein transduction domain of the HIV TAT protein as described in Schwarze, et al. (1999 Science 285:1569-1572), thereby facilitating delivery of polypeptides of the invention into cells.
  • An isolated polypeptide of the invention can be used as an immunogen to generate antibodies using standard techniques for polyclonal and monoclonal antibody preparation.
  • the full-length polypeptide or protein can be used or, alternatively, the invention provides antigenic peptide fragments for use as immunogens.
  • the antigenic peptide of a protein of the invention comprises at least 8 (preferably 10, 15, 20, or 30) amino acid residues of the amino acid sequence of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, and encompasses an epitope of the protein such that an antibody raised against the peptide forms a specific immune complex with the protein.
  • nucleic acid molecules of the invention are present as part of nucleic acid molecules comprising nucleic acid sequences that contain or encode heterologous (e.g. , vector, expression vector, or fusion protein) sequences. These nucleotides can then be used to express proteins which can be used as immunogens to generate an immune response, or more particularly, to generate polyclonal or monoclonal antibodies specific to the expressed protein.
  • An immunogen typically is used to prepare antibodies by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal).
  • a suitable subject e.g., rabbit, goat, mouse or other mammal.
  • An appropriate immunogenic preparation can contain, for example, recombinantly expressed or chemically synthesized polypeptide.
  • the preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent.
  • antibody refers to immunoglobulin molecules and immuno logically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site which specifically binds an antigen, such as a polypeptide of the invention, e.g., an epitope of a polypeptide of the invention.
  • a molecule which specifically binds to a given polypeptide of the invention is a molecule which binds the polypeptide, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin.
  • the invention provides polyclonal and monoclonal antibodies.
  • the term "monoclonal antibody” or “monoclonal antibody composition”, as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope.
  • Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a polypeptide of the invention as an immunogen.
  • Prefe ⁇ ed polyclonal antibody compositions are ones that have been selected for antibodies directed against a polypeptide or polypeptides of the invention.
  • Particularly prefe ⁇ ed polyclonal antibody preparations are ones that contain only antibodies directed against a polypeptide or polypeptides of the invention.
  • Particularly prefe ⁇ ed immunogen compositions are those that contain no other human proteins such as, for example, immunogen compositions made using a non-human host cell for recombinant expression of a polypeptide of the invention. In such a manner, the only human epitope or epitopes recognized by the resulting antibody compositions raised against this immunogen will be present as part of a polypeptide or polypeptides of the invention.
  • the antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide.
  • ELISA enzyme linked immunosorbent assay
  • the antibody molecules can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibodies specific for a protein or polypeptide of the invention can be selected for (e.g., partially purified) or purified by, e.g., affinity chromatography.
  • a recombinantly expressed and purified (or partially purified) protein of the invention is produced as described herein, and covalently or non-covalently coupled to a solid support such as, for example, a chromatography column.
  • the column can then be used to affinity purify antibodies specific for the proteins of the invention from a sample containing antibodies directed against a large number of different epitopes, thereby generating a substantially purified antibody composition, i.e., one that is substantially free of contaminating antibodies.
  • a substantially purified antibody composition is meant, in this context, that the antibody sample contains at most only 30% (by dry weight) of contaminating antibodies directed against epitopes other than those on the desired protein or polypeptide of the invention, and preferably at most 20%>, yet more preferably at most 10%>, and most preferably at most 5% (by dry weight) of the sample is contaminating antibodies.
  • a purified antibody composition means that at least 99% of the antibodies in the composition are directed against the desired protein or polypeptide of the invention.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96) or trioma techniques.
  • the technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al.
  • Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind the polypeptide of interest, e.g., using a standard ELISA assay.
  • a monoclonal antibody directed against a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide of interest.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP Phage Display Kit, Catalog No. 240612).
  • examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication No.
  • recombinant antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region. (See, e.g., Cabilly et al., U.S. Patent No. 4,816,567; and Boss et al., U.S. Patent No.
  • Humanized antibodies are antibody molecules from non- human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule.
  • CDRs complementarily determining regions
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rea ⁇ ange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • Lonberg and Huszar (1995, Int. Rev. Immunol. 13:65-93).
  • Completely human antibodies which recognize a selected epitope can be generated using a technique refe ⁇ ed to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a murine antibody
  • a completely human antibody recognizing the same epitope is used to guide the selection of a completely human antibody recognizing the same epitope.
  • An antibody directed against a polypeptide of the invention can be used to isolate the polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. Moreover, such an antibody can be used to detect the protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the polypeptide.
  • the antibodies can also be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 ⁇ 131j ) 35g or 3fj.
  • an antibody can be conjugated to a therapeutic moiety such as a cytotoxin, a therapeutic agent or a radioactive metal ion.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydro testosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g. , mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g. , mechlorethamine, thioepa chlor
  • daunorubicin (formerly daunomycin) and doxorubicin
  • antibiotics e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)
  • anti-mitotic agents e.g., vincristine and vinblastine
  • the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, ⁇ -interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-l”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), interleukin- 10 ("IL- 10"), interleukin- 12 (“IL- 12”), interferon
  • IL-l interleukin-1
  • IL-2 inter
  • an antibody of the invention can be conjugated to a second antibody to form an "antibody heteroconjugate" as described by Segal in U.S. Patent No. 4,676,980 or alternatively, the antibodies can be conjugated to form an "antibody heteropolymer” as described in Taylor et al, in U.S. Patent Nos. 5,470,570 and 5,487,890.
  • An antibody with or without a therapeutic moiety conjugated to it can be used as a therapeutic that is administered alone or in combination with cytotoxic factor(s) and/or cytokine(s).
  • the invention provides substantially purified antibodies or fragments thereof, including human or non-human antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide of the invention comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Acces
  • the invention provides human or non-human antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA
  • non-human antibodies can be goat, mouse, sheep, horse, chicken, rabbit, or rat antibodies.
  • the non-human antibodies of the invention can be chimeric and/or humanized antibodies.
  • the non-human antibodies of the invention can be polyclonal antibodies or monoclonal antibodies.
  • the invention provides monoclonal antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide of the invention comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291 , PTA 292,
  • the monoclonal antibodies can be human, humanized, chimeric and/or non-human antibodies.
  • the substantially purified antibodies or fragments thereof specifically bind to a signal peptide, a secreted sequence, an extracellular domain, a transmembrane or a cytoplasmic domain cytoplasmic membrane of a polypeptide of the invention.
  • the substantially purified antibodies or fragments thereof, the non-human antibodies or fragments thereof, and/or the monoclonal antibodies or fragments thereof, of the invention specifically bind to a secreted sequence, or alternatively, to an extracellular domain of the amino acid sequence of the invention. Examples of extracellular domains of the invention are shown in SEQ ID NOs:20, 21, 32, 41, 51, 89, 109, 112, 115, 136 or 233.
  • any of the antibodies of the invention can be conjugated to a therapeutic moiety or to a detectable substance.
  • detectable substances that can be conjugated to the antibodies of the invention are an enzyme, a prosthetic group, a fluorescent material, a luminescent material, a bioluminescent material, and a radioactive material.
  • the invention also provides a kit containing an antibody of the invention conjugated to a detectable substance, and instructions for use.
  • a pharmaceutical composition comprising an antibody of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition contains an antibody of the invention, a therapeutic moiety, and a pharmaceutically acceptable carrier.
  • Still another aspect of the invention is a method of making an antibody that specifically recognizes TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349, the method comprising immunizing a mammal with a polypeptide.
  • the polypeptide used as an immunogen comprises an amino acid sequence selected from the group consisting of: the amino acid sequence of any one of SEQ ID NOs:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 , or an amino acid sequence encoded by the cDNA of a clone deposited as ATCC® Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; a fragment of at least 15 amino acid residues of the amino acid sequence
  • a sample is collected from the mammal that contains an antibody that specifically recognizes the immunogen.
  • the polypeptide is recombinantly produced using a non-human host cell.
  • the antibodies can be further purified from the sample using techniques well known to those of skill in the art.
  • the method can further comprise producing a monoclonal antibody- producing cell from the cells of the mammal.
  • antibodies are collected from the antibody-producing cell.
  • vectors preferably expression vectors, containing a nucleic acid encoding a polypeptide of the invention (or a portion thereof).
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g. , bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • expression vectors are capable of directing the expression of genes to which they are operably linked.
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors).
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell.
  • the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed.
  • "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein.
  • the recombinant expression vectors of the invention can be designed for expression of a polypeptide of the invention in prokaryotic (e.g. , E. coli ) or eukaryotic cells (e.g., insect cells (using baculovirus expression vectors), yeast cells or mammalian cells). Suitable host cells are discussed further in Goeddel, supra.
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three pu ⁇ oses: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and
  • GST glutathione S-transferase
  • Suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) and pET 1 Id (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 60-89).
  • Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid t ⁇ -lac fusion promoter.
  • Target gene expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident ⁇ prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
  • One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 119-128).
  • Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al. (1992) Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • the expression vector is a yeast expression vector.
  • yeast S. cerivisae examples include pYepSecl (Baldari et al. (1987) EMBOJ. 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-943), pJRY88 (Schultz et al. (1987) Gene 54:113-123), pYES2 (Invitrogen Co ⁇ oration, San Diego, CA), and pPicZ (Invitrogen Co ⁇ , San Diego, CA).
  • the expression vector is a baculovirus expression vector.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBOJ. 6:187-195).
  • the expression vector's control functions are often provided by viral regulatory elements.
  • viral regulatory elements For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook et al., supra.
  • the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements are known in the art.
  • suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1 :268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J.
  • promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the ⁇ -fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
  • the invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operably linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to the mRNA encoding a polypeptide of the invention.
  • Regulatory sequences operably linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • a high efficiency regulatory region the activity of which can be determined by the cell type into which the vector is introduced.
  • host cell and "recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic (e.g., E. coli) or eukaryotic cell (e.g., insect cells, yeast or mammalian cells).
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have inco ⁇ orated the selectable marker gene will survive, while the other cells die).
  • an endogenous e.g., TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 and MANGO 34
  • nucleic acid within a cell, cell line or microorganism may be modified by inserting a DNA regulatory element heterologous to the endogenous gene of interest into the genome of a cell, stable cell line or cloned microorganism such that the inserted regulatory element is operatively linked with the endogenous gene (e.g., TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes) and controls, modulates or activates the endogenous gene.
  • a DNA regulatory element heterologous to the endogenous gene of interest e.
  • transcriptionally silent, endogenous TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes may be activated by insertion of a promiscuous regulatory element that works across cell types.
  • a heterologous regulatory element may be inserted into a stable cell line or cloned microorganism, such that it is operatively linked with and activates expression of endogenous TANGO 339, TANGO 353, TANGO 358, TANGO, 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes, using techniques, such as targeted homologous recombination, which are well known to those of skill in the art, and described e.g., in Chappel, U.S. Patent No. 5,272,071; PCT publication No. WO 91/06667, published May 16, 1991.
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce a polypeptide of the invention. Accordingly, the invention further provides methods for producing a polypeptide of the invention using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the polypeptide is produced. In another embodiment, the method further comprises isolating the polypeptide from the medium or the host cell.
  • the host cells of the invention can also be used to produce nonhuman transgenic animals.
  • a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which sequences encoding a polypeptide of the invention have been introduced.
  • Such host cells can then be used to create non-human transgenic animals in which exogenous sequences encoding a polypeptide of the invention have been introduced into their genome or homologous recombinant animals in which endogenous encoding a polypeptide of the invention sequences have been altered.
  • Such animals are useful for studying the function and/or activity of the polypeptide and for identifying and/or evaluating modulators of polypeptide activity.
  • a "transgenic animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene.
  • Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc.
  • a transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal.
  • an "homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
  • a transgenic animal of the invention can be created by introducing nucleic acid encoding a polypeptide of the invention (or a homologue thereof) into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal.
  • Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene.
  • a tissue-specific regulatory sequence(s) can be operably linked to the transgene to direct expression of the polypeptide of the invention to particular cells.
  • transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009, U.S. Patent No. 4,873,191 and in Hogan, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986) and Wakayama et al, (1999), Proc. Natl. Acad. Sci. USA, 96:14984-14989. Similar methods are used for production of other transgenic animals.
  • a transgenic founder animal can be identified based upon the presence of the transgene in its genome and/or expression of mRNA encoding the transgene in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying the transgene can further be bred to other transgenic animals carrying other transgenes.
  • a vector which contains at least a portion of a gene encoding a polypeptide of the invention into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the gene.
  • the vector is designed such that, upon homologous recombination, the endogenous gene is functionally disrupted (i.e., no longer encodes a functional protein; also refe ⁇ ed to as a "knock out" vector).
  • the vector can be designed such that, upon homologous recombination, the endogenous gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous protein).
  • the altered portion of the gene is flanked at its 5' and 3' ends by additional nucleic acid of the gene to allow for homologous recombination to occur between the exogenous gene carried by the vector and an endogenous gene in an embryonic stem cell.
  • the additional flanking nucleic acid sequences are of sufficient length for successful homologous recombination with the endogenous gene.
  • flanking DNA both at the 5' and 3' ends
  • flanking DNA both at the 5' and 3' ends
  • the vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced gene has homologously recombined with the endogenous gene are selected (see, e.g., Li et al. (1992) Cell 69:915).
  • the selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see, e.g., Bradley in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed. (IRL, Oxford, 1987) pp. 113-152).
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term.
  • Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene.
  • transgenic non-human animals can be produced which contain selected systems which allow for regulated expression of the transgene.
  • cre/loxP recombinase system of bacteriophage PI is the cre/loxP recombinase system of bacteriophage PI .
  • a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required.
  • Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
  • Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut et al. (1997) Nature 385:810-813 and PCT Publication NOS. WO 97/07668 and WO 97/07669.
  • compositions suitable for administration can be inco ⁇ orated into pharmaceutical compositions suitable for administration.
  • compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and abso ⁇ tion delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be inco ⁇ orated into the compositions.
  • the invention includes methods for preparing pharmaceutical compositions for modulating the expression or activity of a polypeptide or nucleic acid of the invention. Such methods comprise formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid of the invention. Such compositions can further include additional active agents. Thus, the invention further includes methods for preparing a pharmaceutical composition by formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid of the invention and one or more additional active compounds.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules,
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF; Parsippany, NJ) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by including in the composition an agent which delays abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by inco ⁇ orating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • the active compound e.g., a polypeptide or antibody
  • dispersions are prepared by inco ⁇ orating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the prefe ⁇ ed methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the pu ⁇ ose of oral therapeutic administration, the active compound can be inco ⁇ orated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
  • compositions can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g. , with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g. , with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Co ⁇ oration and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • the prefe ⁇ ed dosage is 0.1 mg/kg to 100 mg/kg of body weight (generally 10 mg/kg to 20 mg/kg). If the antibody is to act in the brain, a dosage of 50 mg/kg to 100 mg/kg is usually appropriate. Generally, partially human antibodies and fully human antibodies have a longer half-life within the human body than other antibodies. Accordingly, lower dosages and less frequent administration is often possible. Modifications such as lipidation can be used to stabilize antibodies and to enhance uptake and tissue penetration (e.g., into the brain). A method for lipidation of antibodies is described by Cruikshank et al. ((1997) J. Acquired Immune Deficiency Syndromes and Human Retrovirology 14:193).
  • Antibodies or antibodies conjugated to therapeutic moieties can be administered to an individual alone or in combination with cytotoxic factor(s), chemotherapeutic drug(s), and/or cytokine(s). If the latter, preferably, the antibodies are administered first and the cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) are administered thereafter within 24 hours.
  • the antibodies and cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) can be administered by multiple cycles depending upon the clinical response of the patient.
  • the antibodies and cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) can be administered by the same or separate routes, for example, by intravenous, intranasal or intramuscular administration.
  • Cytotoxic factors include, but are not limited to, TNF- ⁇ , TNF- ⁇ , IL-1, IFN- ⁇ and IL-2.
  • Chemotherapeutic drugs include, but are not limited to, 5-fluorouracil (5FU), vinblastine, actinomycin D, etoposide, cisplatin, methotrexate and doxorubicin.
  • Cytokines include, but are not limited to, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10 and IL-12.
  • a therapeutically effective amount of protein or polypeptide ranges from about 0.001 to 30 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
  • treatment of a subject with a therapeutically effective amount of a protein, polypeptide, or antibody can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with antibody, protein, or polypeptide in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the effective dosage of antibody, protein, or polypeptide used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of diagnostic assays as described herein.
  • the present invention encompasses agents which modulate expression or activity.
  • An agent may, for example, be a small molecule.
  • small molecules include, but are not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e,.
  • heteroorganic and organometallic compounds having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds. It is understood that appropriate doses of small molecule agents depends upon a number of factors within the ken of the ordinarily skilled physician, veterinarian, or researcher.
  • the dose(s) of the small molecule will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the small molecule to have upon the nucleic acid or polypeptide of the invention.
  • Exemplary doses include milligram or microgram amounts of the small molecule per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram.
  • appropriate doses of a small molecule depend upon the potency of the small molecule with respect to the expression or activity to be modulated. Such appropriate doses may be determined using the assays described herein.
  • a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • the nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (U.S. Patent 5,328,470) or by stereotactic injection (see, e.g., Chen et al. (1994) Proc. N ⁇ tl. Ac ⁇ d. Sci. USA 91 :3054-3057).
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • nucleic acid molecules, proteins, protein homologs, and antibodies described herein can be used in one or more of the following methods: a) screening assays; b) detection assays (e.g., chromosomal mapping, tissue typing, forensic biology); c) predictive medicine (e.g., diagnostic assays, prognostic assays, monitoring clinical trials, and pharmacogenomics); and d) methods of treatment (e.g., therapeutic and prophylactic).
  • polypeptides of the invention can to used to (i) modulate cellular proliferation; (ii) modulate cellular differentiation; and/or (iii) modulate cellular adhesion.
  • the isolated nucleic acid molecules of the invention can be used to express proteins (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect mRNA (e.g., in a biological sample) or a genetic lesion, and to modulate activity of a polypeptide of the invention.
  • the polypeptides of the invention can be used to screen drugs or compounds which modulate activity or expression of a polypeptide of the invention as well as to treat disorders characterized by insufficient or excessive production of a protein of the invention or production of a form of a protein of the invention which has decreased or abe ⁇ ant activity compared to the wild type protein.
  • the antibodies of the invention can be used to detect and isolate a protein of the and modulate activity of a protein of the invention.
  • This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
  • the invention provides a method (also refe ⁇ ed to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) which bind to polypeptide of the invention or have a stimulatory or inhibitory effect on, for example, expression or activity of a polypeptide of the invention.
  • modulators i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) which bind to polypeptide of the invention or have a stimulatory or inhibitory effect on, for example, expression or activity of a polypeptide of the invention.
  • the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of a polypeptide of the invention or biologically active portion thereof.
  • the test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound” library method; and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam (1997) Anticancer DrugDes. 12:145).
  • an assay is a cell-based assay in which a cell which expresses a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to the polypeptide determined.
  • the cell for example, can be a yeast cell or a cell of mammalian origin.
  • Determining the ability of the test compound to bind to the polypeptide can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the polypeptide or biologically active portion thereof can be determined by detecting the labeled compound in a complex.
  • test compounds can be labeled with 125 ⁇ 3 s, 14 or -1H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting.
  • test compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • the assay comprises contacting a cell which expresses a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the test compound to preferentially bind to the polypeptide or a biologically active portion thereof as compared to the known compound.
  • an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the polypeptide or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of the polypeptide or a biologically active portion thereof can be accomplished, for example, by determining the ability of the polypeptide protein to bind to or interact with a target molecule.
  • a target molecule is a molecule with which a selected polypeptide (e.g., a polypeptide of the invention) binds or interacts with in nature, for example, a molecule on the surface of a cell which expresses the selected protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule.
  • a target molecule can be a polypeptide of the invention or some other polypeptide or protein.
  • a target molecule can be a component of a signal transduction pathway which facilitates transduction of an extracellular signal (e.g., a signal generated by binding of a compound to a polypeptide of the invention) through the cell membrane and into the cell or a second intercellular protein which has catalytic activity or a protein which facilitates the association of downstream signaling molecules with a polypeptide of the invention.
  • Determining the ability of a polypeptide of the invention to bind to or interact with a target molecule can be accomplished by determining the activity of the target molecule.
  • the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (e.g. , intracellular
  • a reporter gene e.g., a regulatory element that is responsive to a polypeptide of the invention operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase
  • a cellular response for example, cellular differentiation, or cell proliferation.
  • an assay of the present invention is a cell- free assay comprising contacting a polypeptide of the invention or biologically active portion thereof with a test compound and determining the ability of the test compound to bind to the polypeptide or biologically active portion thereof. Binding of the test compound to the polypeptide can be determined either directly or indirectly as described above.
  • the assay includes contacting the polypeptide of the invention or biologically active portion thereof with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the test compound to preferentially bind to the polypeptide or biologically active portion thereof as compared to the known compound.
  • an assay is a cell-free assay comprising contacting a polypeptide of the invention or biologically active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the polypeptide or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of the polypeptide can be accomplished, for example, by determining the ability of the polypeptide to bind to a target molecule by one of the methods described above for determining direct binding. En an alternative embodiment, determining the ability of the test compound to modulate the activity of the polypeptide can be accomplished by determining the ability of the polypeptide of the invention to further modulate the target molecule.
  • the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as previously described.
  • the cell-free assay comprises contacting a polypeptide of the invention or biologically active portion thereof with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the polypeptide to preferentially bind to or modulate the activity of a target molecule.
  • the cell- free assays of the present invention are amenable to use of both a soluble form or the membrane-bound form of a polypeptide of the invention.
  • solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-octylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton X-100, Triton X-l 14, Thesit, Isotridecypoly(ethylene glycol ether)n, 3-[(3-cholamidopropyl)dimethylamminio]-l-propane sulfonate (CHAPS),
  • non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-octylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton X-100, Triton X-l 14, Thesit, Isotri
  • binding of a test compound to the polypeptide, or interaction of the polypeptide with a target molecule in the presence and absence of a candidate compound can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix.
  • glutathione-S-transferase fusion proteins or glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical; St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or A polypeptide of the invention, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above.
  • the complexes can be dissociated from the matrix, and the level of binding or activity of the polypeptide of the invention can be determined using standard techniques.
  • polypeptide of the invention or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated polypeptide of the invention or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g. , biotinylation kit, Pierce Chemicals; Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with the polypeptide of the invention or target molecules but which do not interfere with binding of the polypeptide of the invention to its target molecule can be derivatized to the wells of the plate, and unbound target or polypeptide of the invention trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the polypeptide of the invention or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the polypeptide of the invention or target molecule.
  • modulators of expression of a polypeptide of the invention are identified in a method in which a cell is contacted with a candidate compound and the expression of the selected mRNA or protein (i.e., the mRNA or protein co ⁇ esponding to a polypeptide or nucleic acid of the invention) in the cell is determined.
  • the level of expression of the selected mRNA or protein in the presence of the candidate compound is compared to the level of expression of the selected mRNA or protein in the absence of the candidate compound.
  • the candidate compound can then be identified as a modulator of expression of the polypeptide of the invention based on this comparison.
  • the candidate compound when expression of the selected mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of the selected mRNA or protein expression.
  • the candidate compound when expression of the selected mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of the selected mRNA or protein expression.
  • the level of the selected mRNA or protein expression in the cells can be determined by methods described herein.
  • a polypeptide of the inventions can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol Chem. 268:12046-12054; Bartel et al. (1993) Bio/Techniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and PCT Publication No.
  • binding proteins are also likely to be involved in the propagation of signals by the polypeptide of the inventions as, for example, upstream or downstream elements of a signaling pathway involving the polypeptide of the invention.
  • This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
  • Portions or fragments of the cDNA sequences identified herein (and the co ⁇ esponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. These applications are described in the subsections below.
  • Chromosome Mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. Accordingly, nucleic acid molecules described herein or fragments thereof, can be used to map the location of the co ⁇ esponding genes on a chromosome. The mapping of the sequences to chromosomes is an important first step in co ⁇ elating these sequences with genes associated with disease. Briefly, genes can be mapped to chromosomes by preparing PCR primers
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the nucleic acid sequences of the invention to design oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes. Other mapping strategies which can similarly be used to map a gene to its chromosome include in situ hybridization (described in Fan et al. (1990) Proc. Natl. Acad. Sci. USA 87:6223-27), pre-screening with labeled flow-sorted chromosomes (CITE), and pre-selection by hybridization to chromosome specific cDNA libraries.
  • in situ hybridization described in Fan et al. (1990) Proc. Natl. Acad. Sci. USA 87:6223-27
  • CITE labeled flow-sorted chromosomes
  • Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step.
  • FISH Fluorescence in situ hybridization
  • Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents co ⁇ esponding to noncoding regions of the genes actually are prefe ⁇ ed for mapping pu ⁇ oses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
  • differences in the D ⁇ A sequences between individuals affected and unaffected with a disease associated with a gene of the invention can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that D ⁇ A sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymo ⁇ hisms.
  • nucleic acid sequences disclosed herein can be used to perform searches against "mapping databases", e.g., BLAST-type search, such that the chromosome position of the gene is identified by sequence homology or identity with known sequence fragments which have been mapped to chromosomes.
  • mapping databases e.g., BLAST-type search
  • a polypeptide and fragments and sequences thereof and antibodies specific thereto can be used to map the location of the gene encoding the polypeptide on a chromosome. This mapping can be carried out by specifically detecting the presence of the polypeptide in members of a panel of somatic cell hybrids between cells of a first species of animal from which the protein originates and cells from a second species of animal and then determining which somatic cell hybrid(s) expresses the polypeptide and noting the chromosome(s) from the first species of animal that it contains. For examples of this technique, see Pajunen et al. (1988) Cytogenet. Cell Genet. 47:31-41 and Van Keuren et al. (1986) Hum. Genet. 74:34-40.
  • the presence of the polypeptide in the somatic cell hybrids can be determined by assaying an activity or property of the polypeptide, for example, enzymatic activity, as described in Bordelon-Riser et al. ( 919) Somatic Cell Genetics 5:597-613 and Owerbach et al. (1978; Proc. Natl. Acad. Sci. USA 75:5640-5644.
  • the nucleic acid sequences of the present invention can also be used to identify individuals from minute biological samples.
  • the United States military for example, is considering the use of restriction fragment length polymo ⁇ hism (RFLP) for identification of its personnel.
  • RFLP restriction fragment length polymo ⁇ hism
  • an individual's genomic D ⁇ A is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification.
  • This method does not suffer from the cu ⁇ ent limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult.
  • the sequences of the present invention are useful as additional DNA markers for RFLP (described in U.S. Patent 5,272,057).
  • sequences of the present invention can be used to provide an alternative technique which determines the actual base-by-base DNA sequence of selected portions of an individual's genome.
  • the nucleic acid sequences described herein can be used to prepare two PCR primers from the 5' and 3' ends of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it. Panels of co ⁇ esponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences.
  • the sequences of the present invention can be used to obtain such identification sequences from individuals and from tissue.
  • the nucleic acid sequences of the invention uniquely represent portions of the human genome.
  • allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases.
  • Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification pu ⁇ oses. Because greater numbers of polymo ⁇ hisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals.
  • the noncoding sequences of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128 can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers which each yield a noncoding amplified sequence of 100 bases.
  • a panel of reagents from the nucleic acid sequences described herein is used to generate a unique identification database for an individual, those same reagents can later be used to identify tissue from that individual.
  • positive identification of the individual, living or dead can be made from extremely small tissue samples.
  • DNA-based identification techniques can also be used in forensic biology. Forensic biology is a scientific field employing genetic typing of biological evidence found at a crime scene as a means for positively identifying, for example, a pe ⁇ etrator of a crime.
  • PCR technology can be used to amplify DNA sequences taken from very small biological samples such as tissues, e.g. , hair or skin, or body fluids, e.g. , blood, saliva, or semen found at a crime scene. The amplified sequence can then be compared to a standard, thereby allowing identification of the origin of the biological sample.
  • sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another "identification marker" (i.e. another DNA sequence that is unique to a particular individual).
  • an "identification marker” i.e. another DNA sequence that is unique to a particular individual.
  • actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments.
  • Sequences targeted to noncoding regions are particularly appropriate for this use as greater numbers of polymo ⁇ hisms occur in the noncoding regions, making it easier to differentiate individuals using this technique.
  • polynucleotide reagents include the nucleic acid sequences of the invention or portions thereof, e.g., fragments derived from noncoding regions having a length of at least 20 or 30 bases.
  • nucleic acid sequences described herein can further be used to provide polynucleotide reagents, e.g., labeled or labelable probes which can be used in, for example, an in situ hybridization technique, to identify a specific tissue, e.g., brain tissue. This can be very useful in cases where a forensic pathologist is presented with a tissue of unknown origin. Panels of such probes can be used to identify tissue by species and/or by organ type.
  • polynucleotide reagents e.g., labeled or labelable probes which can be used in, for example, an in situ hybridization technique, to identify a specific tissue, e.g., brain tissue. This can be very useful in cases where a forensic pathologist is presented with a tissue of unknown origin. Panels of such probes can be used to identify tissue by species and/or by organ type.
  • the present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trails are used for prognostic (predictive) pu ⁇ oses to thereby treat an individual prophylactically.
  • diagnostic assays for determining expression of a polypeptide or nucleic acid of the invention and/or activity of a polypeptide of the invention, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with abe ⁇ ant expression or activity of a polypeptide of the invention.
  • the invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention. For example, mutations in a gene of the invention can be assayed in a biological sample. Such assays can be used for prognostic or predictive pu ⁇ ose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with abe ⁇ ant expression or activity of a polypeptide of the invention.
  • Another aspect of the invention provides methods for expression of a nucleic acid or polypeptide of the invention or activity of a polypeptide of the invention in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (refe ⁇ ed to herein as "pharmacogenomics").
  • Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent).
  • Yet another aspect of the invention pertains to monitoring the influence of agents
  • An exemplary method for detecting the presence or absence of a polypeptide or nucleic acid of the invention in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting a polypeptide or nucleic acid (e.g., mRNA, genomic DNA) of the invention such that the presence of a polypeptide or nucleic acid of the invention is detected in the biological sample.
  • a prefe ⁇ ed agent for detecting mRNA or genomic DNA encoding a polypeptide of the invention is a labeled nucleic acid probe capable of hybridizing to mRNA or genomic DNA encoding a polypeptide of the invention.
  • the nucleic acid probe can be, for example, a full-length cDNA, such as the nucleic acid of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a polypeptide of the invention.
  • a full-length cDNA such as the nucleic acid of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a polypeptide of the invention.
  • a prefe ⁇ ed agent for detecting a polypeptide of the invention is an antibody capable of binding to a polypeptide of the invention, preferably an antibody with a detectable label.
  • Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used.
  • the term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • biological sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of a polypeptide of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence.
  • In vitro techniques for detection of genomic DNA include Southern hybridizations.
  • in vivo techniques for detection of a polypeptide of the invention include introducing into a subject a labeled antibody directed against the polypeptide.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • the biological sample contains protein molecules from the test subject.
  • the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.
  • a prefe ⁇ ed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
  • the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting a polypeptide of the invention or mRNA or genomic DNA encoding a polypeptide of the invention, such that the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide is detected in the biological sample, and comparing the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide in the control sample with the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide in the test sample.
  • kits for detecting the presence of a polypeptide or nucleic acid of the invention in a biological sample can be used to determine if a subject is suffering from or is at increased risk of developing a disorder associated with abe ⁇ ant expression of a polypeptide of the invention as discussed, for example, in sections above relating to uses of the sequences of the invention.
  • kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, neurological disorders, eye disorders and embryonic disorders, which are associated with abe ⁇ ant TANGO 339 expression.
  • kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g.
  • disorders associated with TANGO 339 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g. , B cell cancers stimulated by TNF), which are associated with abe ⁇ ant TANGO 353 expression.
  • apoptotic disorders e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus
  • cytotoxic disorders e.g., septic shock, cachexia
  • proliferative disorders e.g. , B cell cancers stimulated by TNF
  • kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders (e.g., thymic disorders) and embryonic disorders, which are associated with abe ⁇ ant TANGO 358 expression.
  • kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)).
  • immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders
  • disorders associated with decreased [x] activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF), which are associated with abe ⁇ ant TANGO 368 or TANGO 369 expression.
  • kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders (e.g., platelet disorders) endothelial disorders and embryonic disorders, which are associated with aberrant TANGO 402 expression.
  • kits can be used to determine if a subject is suffering from or is at risk for brain-related disorders such as cerebral edema, hydrocephalus, brain herniations, iatrogenic disease (due to, e.g., infection, toxins, or drugs), inflammations (e.g., bacterial and viral meningitis, encephalitis, and cerebral toxoplasmosis), cerebrovascular diseases (e.g., hypoxia, ischemia, and infarction, intracranial hemo ⁇ hage and vascular malformations, and hypertensive encephalopathy), and tumors (e.g., neuroglial tumors, neuronal tumors, tumors of pineal cells, meningeal tumors, primary and secondary lymphomas, intracranial tumors, and medulloblastoma), and to treat injury or trauma to the brain, which are associated with abe ⁇ ant MANGO 346 or MANGO 349 expression.
  • iatrogenic disease due to, e.g., infection,
  • kits can be used to determine if a subject is suffering from or is at risk for prostate-related disorders, (e.g. prostate cancer, prostatitis, benign prostatic hypertrophy, benign prostatic hype ⁇ lasia and atypical prostatic stromal lesions) which can be associated with abe ⁇ ant TANGO 365 or TANGO 383 expression.
  • prostate-related disorders e.g. prostate cancer, prostatitis, benign prostatic hypertrophy, benign prostatic hype ⁇ lasia and atypical prostatic stromal lesions
  • kits can be used to determine if a subject is suffering from or is at risk for endocrine-related disorders, e.g., whole animal homeostasis, appetite-related disorders, which are associated with abe ⁇ ant TANGO 393 expression.
  • the kit may comprise a labeled compound or agent capable of detecting the polypeptide or mRNA encoding the polypeptide in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample (e.g. , an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide). Kits may also include instructions for observing that the tested subject is suffering from or is at risk of developing a disorder associated with abe ⁇ ant expression of the polypeptide if the amount of the polypeptide or mRNA encoding the polypeptide is above or below a normal level.
  • a labeled compound or agent capable of detecting the polypeptide or mRNA encoding the polypeptide in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample (e.g. , an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA
  • the kit may comprise, for example: (1) a first antibody (e.g., attached to a solid support) which binds to a polypeptide of the invention; and, optionally, (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
  • a first antibody e.g., attached to a solid support
  • a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
  • the kit may comprise, for example: (1) an oligonucleotide, e.g., a detectably labeled oligonucleotide, which hybridizes to a nucleic acid sequence encoding a polypeptide of the invention or (2) a pair of primers useful for amplifying a nucleic acid molecule encoding a polypeptide of the invention.
  • the kit may also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent.
  • the kit may also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate).
  • the kit may also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained.
  • Each component of the kit is usually enclosed within an individual container and all of the various containers are within a single package along with instructions for observing whether the tested subject is suffering from or is at risk of developing a disorder associated with abe ⁇ ant expression of the polypeptide.
  • the methods described herein can furthermore be utilized as diagnostic or prognostic assays to identify subjects having or at risk of developing a disease or disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention.
  • the assays described herein such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention, such as a proliferative disorder, e.g., psoriasis or cancer, or an angiogenic disorder.
  • the prognostic assays can be utilized to identify a subject having or at risk for developing such a disease or disorder.
  • test sample refers to a biological sample obtained from a subject of interest.
  • a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing disorders such as disorders discussed, for example, in sections above relating to uses of the sequences of the invention.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders, neurological disorders and embryonic disorders, which are associated with abe ⁇ ant TANGO 339 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AEDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)).
  • immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AEDS)
  • inflammatory disorders e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheuma
  • Disorders associated with TANGO 339 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g. , B cell cancers stimulated by TNF), which are associated with abe ⁇ ant TANGO 353 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders (e.g., thymic disorders) and embryonic disorders, which are associated with abe ⁇ ant TANGO 358 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)).
  • immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)
  • inflammatory disorders e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoi
  • Disorders associated with TANGO 358 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin- dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF), which are associated with abe ⁇ ant TANGO 368 or TANGO 369 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders (e.g., platelet disorders), endothelial disorders and embryonic disorders, which are associated with abe ⁇ ant TANGO 402 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing brain-related disorders such as cerebral edema, hydrocephalus, brain herniations, iatrogenic disease (due to, e.g., infection, toxins, or drugs), inflammations (e.g., bacterial and viral meningitis, encephalitis, and cerebral toxoplasmosis), cerebrovascular diseases (e.g.
  • tumors e.g., neuroglial tumors, neuronal tumors, tumors of pineal cells, meningeal tumors, primary and secondary lymphomas, intracranial tumors, and meduUoblastoma
  • MANGO 346 or MANGO 349 expression e.g., hypoxia, ischemia, and infarction, intracranial hemo ⁇ hage and vascular malformations, and hypertensive encephalopathy
  • tumors e.g., neuroglial tumors, neuronal tumors, tumors of pineal cells, meningeal tumors, primary and secondary lymphomas, intracranial tumors, and meduUoblastoma
  • to treat injury or trauma to the brain which are associated with abe ⁇ ant MANGO 346 or MANGO 349 expression.
  • prognostic assays described herein can be used to identify a subject having or at risk of developing prostate-related disorders (e.g, prostate cancer, prostatis, benign prostatic hypertrophy, benign prostatic hype ⁇ lasmia and atypical prostatic stromal lesions).
  • prognostic assays described herein can be used to identify a subject having or at risk of developing endocrine-related disorders (e.g., animal homeostasis and appetite-related disorders), which are associated with abe ⁇ ant TANGO 393 expression.
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention.
  • an agent e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • agents e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • agents e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate
  • such methods can be used to determine whether a subject can be effectively treated with a specific agent or class of agents (e.g., agents of a type which decrease activity
  • the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention in which a test sample is obtained and the polypeptide or nucleic acid encoding the polypeptide is detected (e.g., wherein the presence of the polypeptide or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with abe ⁇ ant expression or activity of the polypeptide).
  • the methods of the invention can also be used to detect genetic lesions or mutations in a gene of the invention, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized abe ⁇ ant expression or activity of a polypeptide of the invention.
  • the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion or mutation characterized by at least one of an alteration affecting the integrity of a gene encoding the polypeptide of the invention, or the mis-expression of the gene encoding the polypeptide of the invention.
  • such genetic lesions or mutations can be detected by ascertaining the existence of at least one of: 1) a deletion of one or more nucleotides from the gene; 2) an addition of one or more nucleotides to the gene; 3) a substitution of one or more nucleotides of the gene; 4) a chromosomal rea ⁇ angement of the gene; 5) an alteration in the level of a messenger RNA transcript of the gene; 6) an abe ⁇ ant modification of the gene, such as of the methylation pattern of the genomic DNA; 7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of the gene; 8) a non-wild type level of a the protein encoded by the gene; 9) an allelic loss of the gene; and 10) an inappropriate post-translational modification of the protein encoded by the gene.
  • assay techniques known in the art which can be used for detecting lesions in a gene.
  • detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and
  • This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to the selected gene under conditions such that hybridization and amplification of the gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
  • nucleic acid e.g., genomic, mRNA or both
  • Alternative amplification methods include: self sustained sequence replication (Guatelli et al. (1990) Proc. Natl Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al. (1988) Bio/Technology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
  • mutations in a selected gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns.
  • sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA.
  • sequence specific ribozymes see, e.g., U.S. Patent No. 5,498,531 can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
  • genetic mutations can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density a ⁇ ays containing hundreds or thousands of oligonucleotides probes (Cronin et al. (1996) Human Mutation 7:244-255; Kozal et al. (1996) Nature Medicine 2:753-759).
  • a sample and control nucleic acids e.g., DNA or RNA
  • high density a ⁇ ays containing hundreds or thousands of oligonucleotides probes e.g., DNA or RNA
  • genetic mutations can be identified in two-dimensional arrays containing light-generated DNA probes as described in Cronin et al., supra.
  • a first hybridization a ⁇ ay of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear a ⁇ ays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization a ⁇ ay that allows the characterization of specific mutations by using smaller, specialized probe a ⁇ ays complementary to all variants or mutations detected.
  • Each mutation a ⁇ ay is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • any of a variety of sequencing reactions known in the art can be used to directly sequence the selected gene and detect mutations by comparing the sequence of the sample nucleic acids with the co ⁇ esponding wild-type (control) sequence.
  • Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995) Bio/Techniques 19:448), including sequencing by mass spectrometry ( see, e.g., PCT Publication No.
  • WO 94/16101 Cohen et al. (1996) Adv. Chromatogr. 36:127-162; and Griffin et al. (1993) Appl Biochem. Biotechnol. 38:147-159).
  • Other methods for detecting mutations in a selected gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242).
  • the technique of "mismatch cleavage” entails providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild-type sequence with potentially mutant RNA or DNA obtained from a tissue sample.
  • RNA/DNA duplexes can be treated with RNase to digest mismatched regions, and DNA/DNA hybrids can be treated with S 1 nuclease to digest mismatched regions.
  • either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397; Saleeba et al. (1992) Methods Enzymol. 217:286-295. In a prefe ⁇ ed embodiment, the control DNA or RNA can be labeled for detection.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in cDNAs obtained from samples of cells.
  • DNA mismatch repair enzymes
  • the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662).
  • a probe based on a selected sequence, e.g.
  • a wild-type sequence is hybridized to a cDNA or other DNA product from a test cell(s).
  • the duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039.
  • alterations in electrophoretic mobility will be used to identify mutations in genes.
  • SSCP Single-stranded DNA fragments of sample and control nucleic acids will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence, and the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • RNA rather than DNA
  • the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).
  • the movement of mutant or wild- type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495).
  • DGGE denaturing gradient gel electrophoresis
  • DNA will be modified to insure that it does not completely denature, for example by adding a 'GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys. Chem. 265:12753).
  • oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al. (1989) Proc. Natl. Acad. Sci. USA 86:6230).
  • Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention.
  • Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent or reduce polymerase extension (Prossner (1993) Tibtech 11 :238).
  • amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci. USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
  • the methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving a gene encoding a polypeptide of the invention.
  • any cell type or tissue e.g., preferably peripheral blood leukocytes, in which the polypeptide of the invention is expressed may be utilized in the prognostic assays described herein.
  • Agents, or modulators which have a stimulatory or inhibitory effect on activity or expression of a polypeptide of the invention as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders associated with abe ⁇ ant activity of the polypeptide.
  • the pharmacogenomics i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug
  • Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug.
  • the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of a polypeptide of the invention, expression of a nucleic acid of the invention, or mutation content of a gene of the invention in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
  • Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See, e.g., Linder (1997) Clin. Chem. 43(2):254-266.
  • two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body are refe ⁇ ed to as "altered drug action.” Genetic conditions transmitted as single factors altering the way the body acts on drugs are refe ⁇ ed to as "altered drug metabolism”. These pharmacogenetic conditions can occur either as rare defects or as polymo ⁇ hisms.
  • G6PD glucose-6-phosphate dehydrogenase deficiency
  • oxidant drugs anti-malarials, sulfonamides, analgesics, nitro urans
  • the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action.
  • drug metabolizing enzymes e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19
  • NAT 2 N-acetyltransferase 2
  • CYP2D6 and CYP2C19 cytochrome P450 enzymes
  • the gene coding for CYP2D6 is highly polymo ⁇ hic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, a PM will show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite mo ⁇ hine. The other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses.
  • the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
  • the activity of a polypeptide of the invention, expression of a nucleic acid encoding the polypeptide, or mutation content of a gene encoding the polypeptide in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
  • pharmacogenetic studies can be used to apply genotyping of polymo ⁇ hic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype.
  • Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of a polypeptide of the invention can be applied not only in basic drug screening, but also in clinical trials.
  • agents e.g., drugs, compounds
  • the effectiveness of an agent, as determined by a screening assay as described herein, to increase gene expression, protein levels or protein activity can be monitored in clinical trials of subjects exhibiting decreased gene expression, protein levels, or protein activity.
  • the effectiveness of an agent, as determined by a screening assay, to decrease gene expression, protein levels or protein activity can be monitored in clinical trials of subjects exhibiting increased gene expression, protein levels, or protein activity.
  • expression or activity of a polypeptide of the invention and preferably, that of other polypeptide that have been implicated in for example, a cellular proliferation disorder can be used as a marker of the immune responsiveness of a particular cell.
  • genes including those of the invention, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) which modulates activity or expression of a polypeptide of the invention (e.g., as identified in a screening assay described herein) can be identified.
  • an agent e.g., compound, drug or small molecule
  • a polypeptide of the invention e.g., as identified in a screening assay described herein
  • cells can be isolated and RNA prepared and analyzed for the levels of expression of a gene of the invention and other genes implicated in the disorder.
  • the levels of gene expression can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of a gene of the invention or other genes.
  • the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
  • the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of the polypeptide or nucleic acid of the invention in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level the of the polypeptide or nucleic acid of the invention in the post-administration samples; (v) comparing the level of the polypeptide or nucleic acid of the invention in the pre-administration sample with the level of the polypeptide or nucleic acid of the invention in the post-administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly.
  • an agent e.g., an agonist, antagonist,
  • increased administration of the agent may be desirable to increase the expression or activity of the polypeptide to higher levels than detected, i.e., to increase the effectiveness of the agent.
  • decreased administration of the agent may be desirable to decrease expression or activity of the polypeptide to lower levels than detected, i.e., to decrease the effectiveness of the agent.
  • the present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with abe ⁇ ant expression or activity of a polypeptide of the invention.
  • disorders characterized by abe ⁇ ant expression or activity of the polypeptides of the invention include proliferative disorders such as cancer.
  • the invention provides a method for preventing in a subject, a disease or condition associated with an abe ⁇ ant expression or activity of a polypeptide of the invention, by administering to the subject an agent which modulates expression or at least one activity of the polypeptide.
  • Subjects at risk for a disease which is caused or contributed to by abe ⁇ ant expression or activity of a polypeptide of the invention can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein.
  • Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the abe ⁇ ancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression.
  • an agonist or antagonist agent can be used for treating the subject.
  • an antagonist of a TANGO 339 protein may be used to treat an immunological disorder.
  • the appropriate agent can be determined based on screening assays described herein.
  • the modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of the polypeptide.
  • An agent that modulates activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of the polypeptide, a peptide, a peptidomimetic, or other small molecule.
  • the agent stimulates one or more of the biological activities of the polypeptide. Examples of such stimulatory agents include the active polypeptide of the invention and a nucleic acid molecule encoding the polypeptide of the invention that has been introduced into the cell.
  • the agent inhibits one or more of the biological activities of the polypeptide of the invention.
  • inhibitory agents include antisense nucleic acid molecules and antibodies.
  • These modulatory methods can be performed in vitro (e.g. , by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject).
  • the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by abe ⁇ ant expression or activity of a polypeptide of the invention.
  • the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g.
  • the method involves administering a polypeptide of the invention or a nucleic acid molecule of the invention as therapy to compensate for reduced or abe ⁇ ant expression or activity of the polypeptide.
  • Stimulation of activity is desirable in situations in which activity or expression is abnormally low or downregulated and/or in which increased activity is likely to have a beneficial effect. Conversely, inhibition of activity is desirable in situations in which activity or expression is abnormally high or upregulated and/or in which decreased activity is likely to have a beneficial effect.
  • Clones containing cDNA molecules encoding TANGO 339, TANGO 353, and TANGO 358 were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-292, as part of a composite deposit representing a mixture of three strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
  • an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOO ⁇ g/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure.
  • a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8%o agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows: TANGO 339 (EpT339) 2.7 kb TANGO 353 (EpT353) 1.3 kb TANGO 358 (EpT358) 1.6 kb
  • strains can be infe ⁇ ed from the fragments liberated.
  • Clones containing cDNA molecules encoding MANGO 346, TANGO 365, and TANGO 368 were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-291, as part of a composite deposit representing a mixture of three strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
  • an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOO ⁇ g/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure.
  • a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows:
  • TANGO 365 (EpT365): 1.4 kb TANGO 368 (EpT368): 1.0 kb
  • strains can be infe ⁇ ed from the fragments liberated.
  • Clones containing cDNA molecules encoding MANGO 349, TANGO 369, TANGO 383, and TANGO 393 (clones EpM349, EpT369, EpT383, and EpT393, respectively), were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-295, as part of a composite deposit representing a mixture of four strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
  • an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOO ⁇ g/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure.
  • a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows:
  • strains can be infe ⁇ ed from the fragments liberated.
  • Clones containing cDNA molecules encoding TANGO 402 were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-294, as part of a composite deposit representing a mixture of two strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
  • an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with 100 ⁇ g/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure.
  • a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions.
  • the digest liberates fragments as follows:
  • the identity of the strain containing TANGO 402 can be infe ⁇ ed from the liberation of a fragment of the above-identified size.

Description

SECRETED PROTEINS AND USES THEREOF
Cross Reference to Related Applications
This application is a continuation-in-part of U.S. patent application Serial No. 09/365,164, filed July 30, 1999, the contents of which are incorporated herein by reference in its entirety.
Background of the Invention
Many secreted proteins, for example, cytokines and cytokine receptors, play a vital role in the regulation of cell growth, cell differentiation, and a variety of specific cellular responses. A number of medically useful proteins, including erythropoietin, granulocyte- macrophage colony stimulating factor, human growth hormone, and various interleukins, are secreted proteins. Thus, an important goal in the design and development of new therapies is the identification and characterization of secreted and transmembrane proteins and the genes which encode them.
Many secreted proteins are receptors which bind a ligand and transduce an intracellular signal, leading to a variety of cellular responses. The identification and characterization of such a receptor enables one to identify both the ligands which bind to the receptor and the intracellular molecules and signal transduction pathways associated with the receptor, permitting one to identify or design modulators of receptor activity, e.g., receptor agonists or antagonists and modulators of signal transduction.
Summary of the Invention
The present invention is based, at least in part, on the discovery of cDNA molecules which encode the TANGO 339, TANGO 353, TANGO 358, TANGO 365,
TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 proteins, all of which are either wholly secreted or transmembrane proteins.
The TANGO 339 proteins are transmembrane 4 domain-containing polypeptides that exhibit homology to human CD9 antigen, a cell surface antigen associated with platelet activation and aggregation.
The TANGO 353, TANGO 358, and TANGO 365 proteins are transmembrane proteins.
The TANGO 368 proteins are secreted proteins encoded by sequences with homology to genomic sequences of the human T-cell receptor gamma VI gene region. The TANGO 383 proteins are transmembrane polypeptides with homology to retinopathy proteins. The TANGO 393 protein are transmembrane proteins with homology to proteins containing Leucine-rich repeats (LRR) such as the Leucine-Rich -2-Glycoprotein (LRG), SLIT- 1 , and Platelet Glycoprotein V (GPV) precursor.
The TANGO 402 proteins are homologous to the LOX-1 protein, which has been associated with low density lipoprotein metabolism and atherosclerosis.
The MANGO 346, MANGO 349, and TANGO 369 proteins are secreted proteins.
The TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 proteins, fragments, derivatives, and variants thereof are collectively referred to herein as "polypeptides of the invention" or "proteins of the invention." Nucleic acid molecules encoding the polypeptides or proteins of the invention are collectively referred to as "nucleic acids of the invention."
The nucleic acids and polypeptides of the present invention are useful as modulating agents in regulating a variety of cellular processes. Accordingly, in one aspect, this invention provides isolated nucleic acid molecules encoding a polypeptide of the invention or a biologically active portion thereof. The present invention also provides nucleic acid molecules which are suitable for use as primers or hybridization probes for the detection of nucleic acids encoding a polypeptide of the invention.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:2, the nucleotide sequence of the cDNA insert of an EpT339 clone deposited with the American Type Culture Collection (ATCC®) as Accession Number PTA-292, or a complement thereof.
The invention features nucleic acid molecules, which are at least 30%ι, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:27, SEQ ID NO:28, the nucleotide sequence of the cDNA insert of an EpT353 clone deposited with the ATCC® as Accession Number PTA-292, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:36, SEQ ID NO:37, the nucleotide sequence of the cDNA insert of an EpT358 clone deposited with the ATCC® as Accession Number PTA-292, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:44, SEQ ID NO:45, the nucleotide sequence of the cDNA insert of an EpT365 clone deposited with the ATCC® as Accession Number PTA-291, or a complement thereof.
The invention features nucleic acid molecules, preferably cDNA molecules, which are at least 98% identical to the nucleotide sequence of SEQ ID NO:52, SEQ ID NO:53, the nucleotide sequence of the cDNA insert of an EpT368 clone deposited with the
ATCC® as Accession Number PTA-291, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%,
45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of
SEQ ID NO:58, SEQ ID NO:59, the nucleotide sequence of the cDNA insert of an EpT369 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof.
The invention features nucleic acid molecules which are at least 40%, 45%, 50%,
55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID
NO:63, the nucleotide sequence of the cDNA insert of an EpT383 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules which are at least 65%, 75%,
80%, 85%, 90%, 95% or 98% identical to the nucleotide sequence of SEQ ID NO:64, the nucleotide sequence of the cDNA insert of an EpT383 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof. The invention features nucleic acid molecules which are at least 30%, 35%, 40%,
45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of
SEQ ID NO:73, SEQ ID NO:74, the nucleotide sequence of the cDNA insert of a human
EpT393 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof. The invention features nucleic acid molecules which are at least 30%, 35%, 40%,
45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of
SEQ ID NO:93, SEQ ID NO:94, the nucleotide sequence of a mouse EpT393 cDNA, or a complement thereof.
The invention features nucleic acid molecules which are at least 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO: 110,
SEQ ID NO:l 11, the nucleotide sequence of the cDNA insert of an EpT402 clone deposited with the ATCC® as Accession Number PTA-294, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%,
45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO: 123, SEQ ID NO: 124, the nucleotide sequence of the cDNA insert of an EpM346 clone deposited with the ATCC® as Accession Number PTA-291, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:128, SEQ ID NO:129, the nucleotide sequence of the cDNA insert of an EpM349 clone deposited with the ATCC® as Accession Number PTA-295, or a complement thereof.
The invention features nucleic acid molecules which are at least 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, a complement thereof, or the non-coding strand of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA-295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention. The invention features nucleic acid molecules of at least 480, 500, 550, 600, 650,
700, 750, 800, 850, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600 or 2700 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:l, the nucleotide sequence of an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. The invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100 contiguous nucleotides of nucleic acids 1 to 2102 of SEQ ID NO:l, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900 or 1000 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:2, or a complement thereof.
The invention features nucleic acid molecules of at least 575, 600, 650, 700, 800, 900, 1000, 1100 or 1200 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:27, the nucleotide sequence of an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. The invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 630 contiguous nucleotides of nucleic acids 1 to 634 of SEQ ID NO:27, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650 or 690 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:28, or a complement thereof. The invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550 or 560 contiguous nucleotides of nucleic acids 1 to 560 of SEQ ID NO:28, or a complement thereof. The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500 or 1600 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:36, the nucleotide sequence of an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least
20, 50, 100, 150, 200 or 240 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 37, or a complement thereof.
The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200, 250, 300, 340, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200 or 1300 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:44, the nucleotide sequence of an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, or 450 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:45, or a complement thereof.
The invention features nucleic acid molecules, preferably a cDNA molecule, which are at least 970 nucleotides of the nucleotide sequence the of the nucleotide sequence of SEQ ID NO:44, the nucleotide sequence of an EpT368 cDNA of ATCC® Accession Number PTA-291, or a complement thereof. The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1050 or 1100 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 58, the nucleotide sequence of an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150 or 174 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:59, or a complement thereof.
The invention features nucleic acid molecules of at least 510, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200 or 1300 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:63, the nucleotide sequence of an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 270, 300, 350, or 400 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 64, or a complement thereof.
The invention also features nucleic acid molecules comprising at least 20, 50, 100, 150, 200 or 250 contiguous nucleotides of nucleic acids 1 to 255 of SEQ ID NO:64, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 610 contiguous nucleotides of nucleic acids 775 to 1386 of SEQ ID NO:63, or a complement thereof.
The invention features nucleic acid molecules of at least 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600 or 1700 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 73, the nucleotide sequence of a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, or 1400 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:74, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least
20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200 or 1250 contiguous nucleotides of nucleotides 1 to 1250 of SEQ ID NO:73, or a complement thereof.
The invention features nucleic acid molecules of at least 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, or 1900 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 93, the nucleotide sequence of a mouse EpT393 cDNA, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950 or 984 contiguous nucleotides nucleotides of nucleic acids 1 to 984 of SEQ ID NO:93, or a complement thereof. The invention also features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250 or 292 contiguous nucleotides of the nucleic acids 1177 to 1469 of SEQ ID NO:93, or a complement thereof.
The invention also features nucleic acid molecules which include a fragment of at least 20,
50, 100, 150, 200, 250 or 280 contiguous nucleotides of the nucleic acids 1666 to 1946 of SEQ ID NO:93, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least
200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000,
1050, 1100, 1150, 1200, 1250, 1300, 1350, or 1400 contiguous nucleotide of the nucleotide sequence of SEQ ID NO:94, or a complement thereof. The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200 or
1300 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:l 10, the nucleotide sequence of an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least
20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 620 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:l 11, or a complement thereof.
The invention features nucleic acid molecules of at least 450, 500, 550, 600, 650,
700, 750, 800, 850, 900, 1000, 1050, 1100 or 1150 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 123, the nucleotide sequence of an EpM346 cDNA of
ATCC® Accession Number PTA-291, or a complement thereof.
The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
250, 300, 350, 400, 450, 500, 550, 600, 650, 700 or 800 contiguous nucleotides of nucleic acids 1 to 805 of SEQ ID NO: 123, or a complement thereof. The invention features nucleic acid molecules which include a fragment of at least
50, 75, 100, or 150 contiguous nucleotides of the nucleotide sequence of SEQ ID NO:124, or a complement thereof.
The invention features nucleic acid molecules of at least 20, 50, 100, 150, 200,
250, 300, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700,
2800, 2900, 3000, 3100, 3200, 3300, 3400, 3500 or 3600 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 128, the nucleotide sequence of an EpM349 cDNA of
ATCC® Accession Number PTA-295, or a complement thereof.
The invention features nucleic acid molecules which include a fragment of at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450 or 500 contiguous nucleotides of the nucleotide sequence of SEQ ID NO: 129, or a complement thereof. The invention features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 20, 50, 100, 150, 200, 250, 300, 400, 450, 500, 550, 600, 650 or more contiguous nucleotides identical to the nucleic acid sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or a complement thereof, or the non-coding strand of TANGO 339, TANGO 353, TANGO, 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:29, the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:38, the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:46, the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
The invention also features nucleic acid molecules, preferably cDNA molecules, which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 99% identical to the amino acid sequence of SEQ ID NO:54, the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:60, the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:65, the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA- 295, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:75, the amino acid sequence encoded by a human
EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:95, the amino acid sequence encoded by a mouse
EpT393 cDNA, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 26%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ED NO:l 12, the amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO: 125, the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 80%, 88%, 90%, 95% or 98% identical to the amino acid sequence of SEQ ID NO: 130, the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules which include a nucleotide sequence encoding a protein having an amino acid sequence that is at least 25% preferably 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA- 292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof, wherein the protein encoded by the nucleotide sequence also exhibits at least one structural and/or functional feature of a polypeptide of the invention.
In preferred embodiments, the nucleic acid molecules have the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA clones of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA- 295, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:3, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175, 200, 225, 250, 260 or 265 contiguous amino acids of SEQ ID NO:3, or the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:29, or a fragment thereof including at least 45, 50, 75, 100, 125, 150, 175, 200 or 225 contiguous amino acids of SEQ ID NO:29, or the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:38, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, or 80 contiguous amino acids of SEQ ID NO:38, or the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:46, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 90, 100, 125, 150 or 160 contiguous amino acids of SEQ ID NO:46, or the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
Also within the invention are nucleic acid molecules, preferably cDNA molecules, which encode a polypeptide having the amino acid sequence of SEQ ID NO:54, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 60, 70, 75 or 80 contiguous amino acids of SEQ ID NO:54, or the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:60, or a fragment thereof including at least 10, 15, 20, 25, 30, 50 or 55 contiguous amino acids of SEQ ID NO:60, or the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:65, or a fragment thereof including at least 90, 100, 110, 125 or 135 contiguous amino acids of SEQ ID NO:65, or the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:75, or a fragment thereof including at least 60, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400 or 450 contiguous amino acids of SEQ ID NO:75, or the amino acid sequence encoded by a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 95, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400 or 450 contiguous amino acids of SEQ ID NO:95, or the amino acid sequence encoded by a mouse EpT393 cDNA, or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO:l 12, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150, 175 or 200 contiguous amino acids of SEQ ID NO:l 12, he amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof. Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 125, or a fragment thereof including at least 10, 15, 20, 25, 30, 35, 40, 45, 50 or 55 contiguous amino acids of SEQ ID NO:125, or the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291 , or a complement thereof.
Also within the invention are nucleic acid molecules which encode a polypeptide having the amino acid sequence of SEQ ID NO: 130, or a fragment thereof including at least 10, 15, 20, 25, 30, 50, 75, 100, 125, 150 or 160 contiguous amino acids of SEQ ID NO: 130, or the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules which encode a polypeptide fragment of at least 10, 15, 25, 30, 50, 75, 100, 125, 150, 175, 200 or more contiguous amino acids of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA- 292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof, wherein the fragment exhibits at least one structural and/or functional feature of a polypeptide of the invention. The invention includes nucleic acid molecules which encode an allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof under stringent conditions. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:3, or the amino acid sequence encoded by an EpT339 cDNA of ATCC® Accession Number PTA-292.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:29, or the amino acid sequence encoded by an EpT353 cDNA of ATCC® Accession Number PTA-292.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:38, or the amino acid sequence encoded by an EpT358 cDNA of ATCC® Accession Number PTA-292. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:46 or the amino acid sequence encoded by an EpT365 cDNA of ATCC® Accession Number PTA-291. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:54, or the amino acid sequence encoded by an EpT368 cDNA of ATCC® Accession Number PTA-291.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least 30%, preferably 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:60, or the amino acid sequence encoded by an EpT369 cDNA of ATCC® Accession Number PTA-295.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 65%, preferably 70%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:65 or the amino acid sequence encoded by an EpT383 cDNA of ATCC® Accession Number PTA-295.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:75 or the amino acid sequence encoded by a human EpT393 cDNA of ATCC® Accession Number PTA-295.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%o, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:95 or the amino acid sequence encoded by a mouse EpT393 cDNA.
Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 26%, preferably 30%, 40%, 45%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO:l 12, or the amino acid sequence encoded by an EpT402 cDNA of ATCC® Accession Number PTA-294. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 125 or the amino acid sequence encoded by an EpM346 cDNA of ATCC® Accession Number PTA-291. Also within the invention are isolated polypeptides or proteins having an amino acid sequence that is at least about 30%, preferably 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the amino acid sequence of SEQ ID NO: 130 or the amino acid sequence encoded by an EpM349 cDNA of ATCC® Accession Number PTA-295.
The invention also features isolated polypeptides or proteins having an amino acid sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95% or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 52, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:3, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 1 or 2, a complement thereof, or the non-coding strand of an EpT339 cDNA of ATCC® Accession Number PTA-292. Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:29, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:27 or 28, a complement thereof, or the non-coding strand of an EpT353 cDNA of ATCC® Accession Number PTA-292.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:38, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:36 or 37, a complement thereof, or the non-coding strand of an EpT358 cDNA of ATCC® Accession Number PTA-292. Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:46, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:45, a complement thereof, or the non-coding strand of an EpT365 cDNA of ATCC® Accession Number PTA-291.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:54, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:52 or 53, a complement thereof, or the non-coding strand of an EpT368 cDNA of ATCC® Accession Number PTA-291.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:60, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 58 or 59, a complement thereof, or the non-coding strand of an EpT369 cDNA of ATCC® Accession Number PTA-295. Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 65%, preferably 70%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:65, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:64, a complement thereof, or the non-coding strand of an EpT383 cDNA of ATCC® Accession Number PTA-295.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 75, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:74, a complement thereof, or the non-coding strand of a human EpT393 cDNA of ATCC® Accession Number PTA 295. Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:96, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:95, a complement thereof, or the non-coding strand of a mouse EpT393 cDNA.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about preferably 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO:l 12, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l 10 or 111, a complement thereof, or the non-coding strand of an EpT402 cDNA of ATCC® Accession Number PTA-294.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 125, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 124, a complement thereof, or the non-coding strand of an EpM346 cDNA of ATCC® Accession Number PTA-291.
Also within the invention are isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to the nucleic acid sequence encoding SEQ ID NO: 130, and isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 129, a complement thereof, or the non-coding strand of an EpM349 cDNA of ATCC® Accession Number PTA-295. The invention features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 30%, preferably 35%, 40%, 45%, 50%, 55%, 65%, 75%, 85%, 95%, or 98% identical to the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, a complement thereof, or the non-coding strand of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA- 291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA- 295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
The invention also features isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence that is at least about 25%, preferably 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%, 85%, 95% or 98% identical to a nucleic acid sequence encoding SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, isolated polypeptides or proteins which are encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, a complement thereof, or the non-coding strand of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of ATCC® Accession Number PTA-291 , Accession Number PTA-292, Accession Number PTA-294 or
Accession Number PTA-295, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention.
Also within the invention are polypeptides which are allelic variants of a polypeptide that includes the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC®
Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, respectively, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule having the sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129, or a complement thereof under stringent conditions. Also within the invention are polypeptides which are allelic variants of a polypeptide that includes the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125 or 130, or the amino acid sequence encoded by a cDNA of ATCC® Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, respectively, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule having the sequence of SEQ ID NO: 1, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129, or a complement thereof under stringent conditions, wherein the polypeptides or proteins also exhibit at least one structural and/or functional feature of a polypeptide of the invention. The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l or 2, or an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 480, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600 or 2700 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, an EpT339 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. In another embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900 or 1000 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:2 or nucleic acids 1 to 2100 of SEQ ID NO:l, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:27, or an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 575, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150 or 1200 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:27 or 28, an EpT353 cDNA of ATCC® Accession Number PTA- 292, or a complement thereof. In another embodiment, the nucleic acid molecules are at least 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650 or 690 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:28, an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. In yet another embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550 or 560 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising nucleotides 1 to 500 of SEQ ID NO:28, an EpT353 cDNA of ATCC® Accession Number PTA-292, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:36 or 37, or an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 50, 100, 150, 200 or 240 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:36 or 37, an EpT358 cDNA of ATCC® Accession Number PTA-292, or a complement thereof. The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:44, or an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, or 1150 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:44, an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof. In another embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450 or 490 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:45, an EpT365 cDNA of ATCC® Accession Number PTA-291, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:58 or 59, or an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 20, 50, 100, 150 or 174 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:58 or 59, an EpT369 cDNA of ATCC® Accession Number PTA-295, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 63, or an EpT383 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, or 600 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides of SEQ ID NO:63, or an EpT 283 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. Preferably, such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 250 and/or 800 to 1386 of SEQ ID NO:63.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 73, or a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. In other embodiments, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350 or 1386 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides from of SEQ ID NO:73, a human EpT393 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. Preferably, such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 1250 of SEQ ID NO:73.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:93, or a mouse EpT393 cDNA, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800 or 1900 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotides of SEQ ID NO:93, a mouse EpT393 cDNA or a complement thereof. Preferably, such nucleic acids hybridize under these conditions to at least a portion of nucleotides 1 to 950 and or 1200 to 1800 of SEQ ID NO:93.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l 10 or 111, or an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600 or 620 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 110 or 111, an EpT402 cDNA of ATCC® Accession Number PTA-294, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 123, or an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100 or 1150 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 123, an EpM346 cDNA of ATCC® Accession Number PTA-291, or a complement thereof. In another embodiment, the nucleic acid molecules are at least 50, 75, 100, 125, 150 or 175 contiguous nucleotides in length and hybridize under stringent conditions to the nucleotide sequence of SEQ ID NO: 124, or a complement thereof.
The invention also features nucleic acid molecules that hybridize under stringent conditions to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 128, or an EpM349 cDNA of ATCC® Accession Number PTA-295, or a complement thereof. In one embodiment, the nucleic acid molecules are at least 20, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2700, 2800, 2900, 3000, 3100, 3200, 3300, 3400, 3500, or 3600 contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO: 128, an EpM349 cDNA of ATCC® Accession Number PTA- 295, or a complement thereof.
The invention also features nucleic acid molecules at least 15, preferably 50, 75, 100, 150, 200, 250, 300, 350, 400, 500, 600 or more contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or a nucleotide sequence of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof. The invention also features nucleic acid molecules at least 15, preferably 50, 75, 100, 150, 200, 250, 300, 350, 400, 500, 600 or more contiguous nucleotides in length and hybridize under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or a nucleotide sequence of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 cDNA of Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294 or Accession Number PTA-295, or a complement thereof, wherein such nucleic acid molecules encode polypeptides or proteins that exhibit at least one structural and/or functional feature of a polypeptide of the invention.
In prefeπed embodiments, the isolated nucleic acid molecules encode a cytoplasmic, transmembrane, or extracellular domain of a polypeptide of the invention.
In one embodiment, the invention provides an isolated nucleic acid molecule which is antisense to the coding strand of a nucleic acid of the invention. Another aspect of the invention provides vectors, e.g., recombinant expression vectors, comprising a nucleic acid molecule of the invention. In another embodiment, the invention provides host cells containing such a vector or engineered to contain and/or express a nucleic acid molecule of the invention. The invention also provides methods for producing a polypeptide of the invention by culturing, in a suitable medium, a host cell of the invention containing a recombinant expression vector encoding a polypeptide of the invention such that the polypeptide of the invention is produced.
Another aspect of this invention features isolated or recombinant proteins and polypeptides of the invention. Preferred proteins and polypeptides possess at least one biological activity possessed by the corresponding naturally-occurring human polypeptide. An activity, a biological activity, or a functional activity of a polypeptide or nucleic acid of the invention refers to an activity exerted by a protein, polypeptide or nucleic acid molecule of the invention on a responsive cell as determined in vivo or in vitro, according to standard techniques. Such activities can be a direct activity, such as an association with or an enzymatic activity on a second protein, or an indirect activity, such as a cellular signaling activity mediated by interaction of the protein with a second protein.
For TANGO 339 or modulators thereof, biological activities include, e.g., (1) the ability to modulate (this term, as used herein, includes, but is not limited to, stabilize, promote, inhibit or disrupt) the development, differentiation, proliferation and/or activity of immune cells (e.g., B-lymphocyte function); (2) the ability to modulate the development and progression of cancer (e.g. lymphomas and/or melanoma-associated cancer); (3) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein- ligand interactions, e.g., in receptor-ligand recognition; (4) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; (5) the ability to modulate hematopoietic processes; (6) the ability to modulate platelet activation and aggregation; (7) the ability to modulate intracellular signaling cascades
(e.g., signal transduction cascades); (8) the ability to modulate intercellular signaling (e.g., in the nervous system); (9) the ability modulate the development, differentiation, proliferation and/or activity of neuronal cells and glial cells (e.g., oligodendrocytes and astrocytes); (10) the ability to modulate the development, differentiation and activity of eye structures, such as the retina (e.g., the ability to modulate photoreceptor disk morphogenesis); and (11) the ability to modulate the development of organs, tissues and/or cells in an embryo and/or fetus.
For TANGO 353 or modulators thereof, biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of immune cells, such as lymphocytes (e.g., T cells and B cells); (2) ability to modulate cell proliferation, e.g., abnormal cell proliferation;(3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. , in receptor-ligand recognition.
For TANGO 358 or modulators thereof, biological activities include, e.g., (1) the ability to modulate development, differentiation, maturation, proliferation and/or activity of immune cells such as thymocytes, e.g., T- lymphocytes; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition. For TANGO 365 or modulators thereof, biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of prostate cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
For TANGO 368 or modulators thereof, biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of cells, such as immune cells, e.g., natural killer cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
For TANGO 369 or modulators thereof, biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of cells, such as immune cells, e.g., natural killer cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling (e.g., in the immune system); (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; and (6) the ability to modulate, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition.
For TANGO 383 or modulators thereof, biological activities include, e.g., (1) the ability to modulate, e.g. , stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of prostate cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; and (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions.
For TANGO 393 or modulators thereof, biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions e g-, homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) the ability to modulate the proliferation, differentiation and/or activity of hypothalamus cells; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate intercellular signaling; and (5) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions. For TANGO 402 or modulators thereof, biological activities include, e.g., (1) the ability to modulate development, differentiation, proliferation and/or activity of immune cells (e.g., leukocytes and macrophages), endothelial cells and smooth muscle cells; (2) the ability to modulate the host immune response; (3) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (4) the ability to modulate the development of organs, tissues and/or cells of the embryo and/or fetus; (5) the ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions; (6) the ability to modulate atherosclerosis, e.g., the initiation and progression of atherosclerosis; (7) the ability to modulate low-density lipoproteins e.g., the ability to modulate levels, metabolism and/or cellular uptake of oxidized low-density lipoprotein (Ox-LDL), the ability to bind to Ox-LDL, and the ability to modulate Ox-LDL activity in cells; (8) the ability to modulate atherogenesis; and (9) the ability to modulate inflammatory functions e.g., by modulating leukocyte adhesion to extracellular matrix and/or endothelial cells; (10) the ability to bind proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (11) the ability to modulate internalization of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (12) the ability to modulate degradation, e.g., proteolytic degradation, of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (13) the ability to modulate, e.g., increase, uptake of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low- density lipoproteins, by cells, e.g., macrophages and muscle cells, e.g., smooth muscle cells; (14) the ability to modulate, e.g., prevent, lipid deposition, e.g., in arteries, and thus modulate, e.g., prevent, intimal thickening; (15) the ability to modulate, e.g., induce or prevent, changes in cells, e.g., transformation of cells (e.g., macrophages and smooth muscle cells) into foam cells and functional alteration of cells (e.g., endothelial cells, e.g., intimal neovascular endothelial cells); (16) the ability to bind and phagocytose cells, e.g., aged and apoptotic cells; (17) the ability to remove debris, e.g., apoptotic cells, from blood vessel walls; (18) the ability to modulate homeostasis, e.g., vascular homeostasis, e.g., by modulating, e.g., preventing the impairment of, nitric oxide production; (19) the ability to modulate, e.g., inhibit, the expression of molecules, e.g., adhesion molecules (e.g., leukocyte adhesion molecules) and growth factors (e.g., smooth-muscle growth factors); (20) the ability to alter, e.g., increase, expression in response to stimuli, e.g., TNF, shear stress, and pathophysiological stimuli relevant to disorders (e.g., atherosclerosis and inflammation); (21) the ability to form, e.g., stabilize, promote, facilitate, inhibit, or disrupt, cell to cell and cell to blood product interaction, e.g., between leukocytes and platelets or leukocytes and vascular endothelial cells; and (22) the ability to recognize large molecules, e.g., carbohydrates.
For MANGO 346 or modulators thereof, biological activities include, e.g., (1) the ability to modulate, e.g. , stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); (5) the ability to modulate neural signaling; (6) the ability to modulate intercellular signaling (e.g., in the neural system); and (7) ability to modulate cell-cell interactions and/or cell-extracellular matrix interactions.
For MANGO 349 or modulators thereof, biological activities include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades); and (5) the ability to modulate intercellular signaling (e.g., in the immune system).
In one embodiment, a polypeptide of the invention has an amino acid sequence sufficiently identical to an identified domain of a polypeptide of the invention. As used herein, the term "sufficiently identical" refers to a first amino acid or nucleotide sequence which contains a sufficient or minimum number of identical or equivalent (e.g., with a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have or encode a common structural domain and/or common functional activity. For example, amino acid or nucleotide sequences which contain or encode a common structural domain having about 60% identity, preferably 65% identity, more preferably 75%, 85%, 95%, 98% identity.
In one embodiment, a TANGO 339 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain, transmembrane 4 domain, a transmembrane 4-like domain, a peripherin/rom-1 domain, a peripherin/rom-1-like domain, and an intracellular or cytoplasmic domain. In one embodiment, a TANGO 353 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain and an intracellular or cytoplasmic domain.
In one embodiment, an TANGO 358 includes at least one or more of the following domains: a signal sequence, an extracellular domain, a transmembrane domain, and an intracellular or cytoplasmic domain.
In one embodiment, a TANGO 365 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, at least one transmembrane domain and an intracellular or cytoplasmic domain.
In one embodiment, a TANGO 368 protein includes at least a signal peptide. In one embodiment, a TANGO 369 protein includes at least a signal peptide. In one embodiment, a TANGO 383 protein includes at least one or more of the following domains: a signal sequence, at least one transmembrane domain, an intracellular or cytoplasmic domain, and an extracellular domain.
In one embodiment, a TANGO 393 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, and a transmembrane domain, a leucine-rich repeat domain and an intracellular or cytoplasmic domain.
In one embodiment, a TANGO 402 protein includes at least one or more of the following domains: a signal sequence, an extracellular domain, a C-type lectin domain, a C-type lectin-like domain, a transmembrane domain, and an intracellular or cytoplasmic domain.
In one embodiment, a MANGO 346 protein includes at least a signal sequence.
In one embodiment, a MANGO 349 protein includes at least a signal sequence.
In one embodiment, the isolated polypeptide of the invention lacks both a transmembrane and a cytoplasmic domain. In another embodiment, the polypeptide lacks both a transmembrane domain and a cytoplasmic domain and is soluble under physiological conditions.
The polypeptides of the present invention, or biologically active portions thereof, can be operably linked to a heterologous amino acid sequence to form fusion proteins. The invention further features antibodies that specifically bind a polypeptide of the invention such as monoclonal or polyclonal antibodies.
In addition, the polypeptides of the invention or biologically active portions thereof, or antibodies of the invention, can be incorporated into pharmaceutical compositions, which optionally include pharmaceutically acceptable carriers.
In another aspect, the present invention provides methods for detecting the presence, activity or expression of a polypeptide of the invention in a biological sample by contacting the biological sample with an agent capable of detecting an indicator of the presence, activity or expression such that the presence activity or expression of a polypeptide of the invention is detected in the biological sample.
In another aspect, the invention provides methods for modulating activity of a polypeptide of the invention comprising contacting a cell with an agent that modulates (inhibits or stimulates) the activity or expression of a polypeptide of the invention such that activity or expression in the cell is modulated. In one embodiment, the agent is an antibody that specifically binds to a polypeptide of the invention. In another embodiment, the agent is a fragment of a polypeptide of the invention or a nucleic acid molecule encoding such a polypeptide fragment. In another embodiment, the agent modulates expression of a polypeptide of the invention by modulating transcription, splicing, or translation of an mRNA encoding a polypeptide of the invention. In yet another embodiment, the agent is a nucleic acid molecule having a nucleotide sequence that is antisense to the coding strand of an mRNA encoding a polypeptide of the invention.
The present invention also provides methods to treat a subject having a disorder characterized by aberrant activity of a polypeptide of the invention or abeπant expression of a nucleic acid of the invention by administering an agent which is a modulator of the activity of a polypeptide of the invention or a modulator of the expression of a nucleic acid of the invention to the subject. In one embodiment, the modulator is a protein of the invention. In other embodiments, the modulator is a polypeptide (e.g., an antibody or a fragment of a polypeptide of the invention), a peptidomimetic, or other small molecule (e.g., a small organic molecule).
The present invention also provides diagnostic assays for identifying the presence or absence of a genetic lesion or mutation characterized by at least one of: (i) abeπant modification or mutation of a gene encoding a polypeptide of the invention, (ii) mis- regulation of a gene encoding a polypeptide of the invention, and (iii) abeπant post- translational modification of the invention wherein a wild-type form of the gene encodes a protein having the activity of the polypeptide of the invention. In another aspect, the invention provides a method for identifying a compound that binds to or modulates the activity of a polypeptide of the invention. In general, such methods entail, measuring a biological activity of the polypeptide in the presence and absence of a test compound and identifying those compounds which alter the activity of the polypeptide. The invention also features methods for identifying a compound which modulates the expression of a polypeptide or nucleic acid of the invention by measuring the expression of the polypeptide or nucleic acid in the presence and absence of the compound.
In another aspect, the invention provides human or non-human antibodies or fragments thereof which specifically bind to a protein of the invention.
In a prefeπed embodiment, an antibody or a fragment thereof, i.e., human and non- human antibodies or fragments thereof and/or monoclonal antibodies or fragments thereof of the invention, specifically bind to an extracellular domain having the amino acid sequence of SEQ ID NO:20, 21, 32, 41, 51, 89, 109, 112, 115, 136 or 233. Any of the antibodies of the invention can be conjugated to a therapeutic moiety or to a detectable substance. Non- limiting examples of detectable substances that can be conjugated to the antibodies of the invention are an enzyme, a prosthetic group, a fluorescent material, a luminescent material, a bioluminescent material, and a radioactive material.
The invention also provides a kit containing an antibody of the invention and instructions for use. Such kits can also comprise an antibody of the invention conjugated to a detectable substance and instructions for use. Still another aspect of the invention is a pharmaceutical composition comprising an antibody of the invention and a pharmaceutically acceptable carrier. In prefeπed embodiments, the pharmaceutical composition contains an antibody of the invention, a therapeutic moiety, and a pharmaceutically acceptable carrier.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
Brief Description of the Drawings FIGURE 1 depicts the cDNA sequence of human TANGO 339 (SEQ ID NO: 1) and the predicted amino acid sequence of human TANGO 339 (SEQ ID NO:3). The open reading frame of SEQ ID NO:l extends from nucleotide 210 to nucleotide 1019 of SEQ ID NO:l (SEQ ID NO:2).
FIGURE 2 depicts a hydropathy plot of human TANGO 339. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and N-glycosylation site (Ngly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 42 of SEQ ID NO:3; SEQ ID NO:5) on the left from the mature protein (amino acids 43 to 270 of SEQ ID NO:3; SEQ ID NO:4) on the right.
FIGURE 3 depicts an alignment of the amino acid sequence of human CD9 antigen (SEQ ID NO:24; Accession Number NM 001769) and the amino acid sequence of human TANGO 339 (SEQ ID NO:3). The amino acid sequences of human CD9 antigen and human TANGO 339 are 24.1% identical. This alignment was performed using the ALIGN alignment program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4.
FIGURE 4 depicts an alignment of the nucleotide sequence of the coding region of human CD9 antigen (SEQ ID NO:25; Accession Number NM__001769) and the nucleotide sequence of the coding region of human TANGO 339 (SEQ ID NO:l). The nucleotide sequences of the coding regions of human CD9 antigen and human TANGO 339 are 45.9% identical. This alignment was performed using the ALIGN alignment program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4.
FIGURE 5 depicts the cDNA sequence of human TANGO 353 (SEQ ID NO:27) and the predicted amino acid sequence of human TANGO 353 (SEQ ID NO:29). The open reading frame of human TANGO 353 extends from nucleotide 76 to nucleotide 765 of SEQ ID NO:27 (SEQ ID NO:28).
FIGURE 6 depicts a hydropathy plot of human TANGO 353. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and potential N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 14 of SEQ ID NO:29; SEQ ID NO:31) on the left from the mature protein (amino acids 15 to 230 of SEQ ID NO:29; SEQ ID NO:30) on the right.
FIGURE 7 depicts a cDNA sequence of human TANGO 358 (SEQ ID NO:36) and the predicted amino acid sequence of human TANGO 358 (SEQ ID NO:38). The open reading frame of human TANGO 358 extends from nucleotide 184 to 429 of SEQ ID NO:36 (SEQ ID NO:37).
FIGURE 8 depicts a hydropathy plot of human TANGO 358. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 42 of SEQ ID NO:36; SEQ ID NO:40) on the left from the mature protein (amino acids 43 to 82 of SEQ ID NO:36; SEQ ID NO:39) on the right. FIGURE 9 depicts the cDNA sequence of human TANGO 365 (SEQ ID NO:44) and the predicted amino acid sequence of human TANGO 365 (SEQ ID NO:46). The open reading frame of SEQ ID NO:44 extends from nucleotide 56 to nucleotide 550 (SEQ ID NO:45).
FIGURE 10 depicts a hydropathy plot of human TANGO 365. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (Cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 36 of SEQ ID NO:46; SEQ ID NO:47) preceding the mature protein (amino acids 37 to 165 of SEQ ID NO:46; SEQ ID NO:48) on the right. FIGURE 11 depicts the cDNA sequence of human TANGO 368 (SEQ ID NO: 52) and the predicted amino acid sequence of TANGO 368 (SEQ ID NO: 54). The open reading frame of human TANGO 368 extends from nucleotide 152 to nucleotide 328 of SEQ ID NO:52 (SEQ ID NO:53). FIGURE 12 depicts a hydropathy plot of human TANGO 368. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (Cys) and potential N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 27 of SEQ ID NO:54; SEQ ID NO:56) on the left from the mature protein (amino acids 28 to 59 of SEQ ID NO:54; SEQ ID NO:55) on the right. FIGURE 13 depicts a local alignment of the nucleotide sequence of full-length human TANGO 368 (SEQ ID NO:52) and a fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57). The nucleotide sequence of human TANGO 368 and the human T-cell receptor gamma VI gene region are 99.3 % identical for a 973 bp overlap. This alignment was performed using the LALIGN program with a PAM120 scoring matrix, a gap length penalty of 12 and a gap penalty of 4.
FIGURE 14 depicts a cDNA sequence of human TANGO 369 (SEQ ID NO: 58) and the predicted amino acid sequence of human TANGO 369 (SEQ ID NO:60). The open reading frame of human TANGO 369 extends from nucleotide 162 to 335 of SEQ ID NO:58 (SEQ ED NO:61).
FIGURE 15 depicts a hydropathy plot of human TANGO 369. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:60; SEQ ID NO:62) on the left from the mature protein (amino acids 27 to 58 of SEQ ID NO:60; SEQ ID NO:61) on the right. FIGURE 16 depicts the cDNA sequence of human TANGO 383 (SEQ ID NO:63) and the predicted amino acid sequence of human TANGO 383 (SEQ ID NO:65). The open reading frame of SEQ ED NO:63 extends from nucleotide 104 to nucleotide 523 (SEQ ID NO:64).
FIGURE 17 depicts a hydropathy plot of human TANGO 383. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 20 of SEQ ID NO: 65; SEQ ID NO:66) on the left from the mature protein (amino acids 21 to 140 of SEQ ID NO:65; SEQ ID NO:67) on the right. FIGURE 18 depicts an alignment of the amino acid sequence of TANGO 383
(SEQ ID NO:65) and the amino acid sequence of Neuronal Thread Protein AD7C-NTP (SEQ ID NO:72). The alignments demonstrates that the amino acid sequences of TANGO 383 and Neuronal Thread Protein AD7C-NTP are 52% identical. This alignment was performed using the ProDom NCBI-BLASTP2 program with graphical output using the following settings: Matrix: BLOSUM62; Expect: 0.1; Filter: none.
FIGURE 19 depicts the cDNA sequence of human TANGO 393 (SEQ ID NO:73) and the predicted amino acid sequence of human TANGO 393 (SEQ ID NO:75). The open reading frame of SEQ ID NO: 73 extends from nucleotide 40 to nucleotide 1458 (SEQ ID NO:74). FIGURE 20 depicts a hydropathy plot of human TANGO 393. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and N-glycosylation sites (Ngly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:75; SEQ ID NO:76) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:75; SEQ ID NO:77) on the right.
FIGURE 21 depicts the cDNA sequence of mouse TANGO 393 (SEQ ID NO:93) and the predicted amino acid sequence of mouse TANGO 393 (SEQ ID NO:95). The open reading frame of SEQ ID NO:93 extends from nucleotide 226 to nucleotide 1644 (SEQ ID NO:94).
FIGURE 22 depicts a hydropathy plot of mouse TANGO 393. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and N-glycosylation sites (N-Gly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:95; SEQ ID NO:96) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:95; SEQ ID NO:97) on the right.
FIGURE 23 depicts an alignment of the open reading frames of human TANGO 393 (SEQ ID NO:74) and mouse TANGO 393 (SEQ ID NO:94) demonstrating an identity of 82.8%. The algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989) FIGURE 24 depicts an alignment of the immature proteins of human TANGO 393 (SEQ ID NO:75) and mouse TANGO 393 (SEQ ID NO:95) demonstrating an identity of 89.2%. The algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989) FIGURE 25 depicts the cDNA sequence of human TANGO 402 (SEQ ED NO : 110) and the predicted amino acid sequence of human TANGO 402 (SEQ ID NO: 112). The open reading frame of human TANGO 402 extends from nucleotide 87 to nucleotide 707 of SEQ ID NO: 110 (SEQ ID NO: 111).
FIGURE 26 depicts a hydropathy plot of human TANGO 402. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) and N-glycosylation (Ngly) sites are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 50 of SEQ ID NO: 112; SEQ ID NO: 114) on the left from the mature protein (amino acids 51 to 207 of SEQ ED NO:112; SEQ ID NO:113) on the right.
FIGURE 27 depicts an alignment of the amino acid sequence of human TANGO 402 (SEQ ID NO:l 12) and the amino acid sequence of human LOX-1 (SEQ ID NO: 122; Accession Number AB010710). The alignment demonstrates that the amino acid sequences of human TANGO 402 and human LOX-1 are 25.1% identical. This alignment was performed using the ALIGN program with a PAM120 scoring matrix, a gap length penalty of 12 and a gap penalty of 4.
FIGURE 28 depicts an alignment of the nucleotide sequences of the open reading frames of human TANGO 402 (SEQ ID NO: 111) and human LOX-1 (SEQ ID NO: 121 ; Accession Number AB010710). The alignment of the open reading frame of human TANGO 402 and that of human LOX-1 demonstrates that those two coding regions are 42.0 % identical. An alignment demonstrates that the nucleotide sequences of the cDNA of human TANGO 402 and human LOX-1 are 44.0 % identical. The alignments were performed using the ALIGN program with a PAM120 scoring matrix, a gap length penalty of 12, and a gap penalty of 4. FIGURE 29 depicts the cDNA sequence of human MANGO 346 (SEQ ED
NO: 123) and the predicted amino acid sequence of human MANGO 346 (SEQ ID NO: 125). The open reading frame of SEQ ID NO: 123 extends from nucleotide 319 to nucleotide 498 (SEQ ID NO: 124).
FIGURE 30 depicts a hydropathy plot of human MANGO 346. Relatively hydrophobic regions of the protein are above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 19 of SEQ ID NO: 125; SEQ ID NO: 126) on the left from the mature protein (amino acids 20 to 60 of SEQ ID NO: 125; SEQ ID NO: 127) on the right. FIGURE 31 depicts the cDNA sequence of human MANGO 349 (SEQ ID
NO: 128) and the predicted amino acid sequence of human MANGO 349 (SEQ ID NO: 130). The open reading frame of SEQ ED NO:128 extends from nucleotide 221 to nucleotide 721 (SEQ ED NO: 129).
FIGURE 32 depicts a hydropathy plot of human MANGO 349. Relatively hydrophobic regions of the protein are shown above the dashed horizontal line, and relatively hydrophilic regions of the protein are below the dashed horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:130; SEQ ID NO:131) on the left from the mature protein (amino acids 27 to 167 of SEQ ID NO: 130; SEQ ID NO: 132) on the right.
Detailed Description of the Invention
TheTANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 and MANGO 349 proteins and nucleic acid molecules comprise families of molecules having certain conserved structural and functional features. As used herein, the terms "family" or "families" are intended to mean two or more proteins or nucleic acid molecules having a common structural domain and having sufficient amino acid or nucleotide sequence identity as defined herein. Family members can be from either the same or different species. For example, a family can comprises two or more proteins of human origin, or can comprise one or more proteins of human origin and one or more of non-human origin. Members of the same family may also have common structural domains.
For example, TANGO 339 proteins, TANGO 353 proteins, TANGO 358 proteins, TANGO 365 proteins, TANGO 368 proteins, TANGO 369 proteins, TANGO 383 proteins, TANGO 393 proteins, TANGO 402 proteins, MANGO 346 proteins and MANGO 349 proteins of the invention can have signal sequences. As used herein, a "signal sequence" includes a peptide of at least about 15 or 20 amino acid residues in length which occurs at the N-terminus of secretory and membrane-bound proteins and which contains at least about 70% hydrophobic amino acid residues such as alanine, leucine, isoleucine, phenylalanine, proline, tyrosine, tryptophan, or valine. In a prefeπed embodiment, a signal sequence contains at least about 10 to 40 amino acid residues, preferably about 19-34 amino acid residues, and has at least about 60-80%, more preferably 65-75%, and more preferably at least about 70% hydrophobic residues. A signal sequence serves to direct a protein containing such a sequence to a lipid bilayer. Thus, in one embodiment, a TANGO 339 protein contains a signal sequence of about amino acids 1 to 42 of SEQ ID NO:3 (SEQ ID NO:5).
In another embodiment, a TANGO 353 protein contains a signal sequence of about amino acids 1 to 14 of SEQ ID NO:37 (SEQ ID NO:31). In another embodiment, a TANGO 358 protein contains a signal sequence at about amino acids 1 to 42 of SEQ ID NO:38 (SEQ ID NO:40). In another embodiment, a TANGO 365 protein contains a signal sequence of about amino acids 1 to 36 of SEQ ID NO:46 (SEQ ID NO:47). In another embodiment, a TANGO 368 protein contains a signal sequence of about amino acids 1 to 27 of SEQ ID NO:54 (SEQ ID NO:56). In another embodiment, a TANGO 369 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ED NO:60 (SEQ ED NO:62). In another embodiment, a TANGO 383 protein contains a signal sequence of about amino acids 1 to 20 of SEQ ID NO:65 (SEQ ED NO:66). In another embodiment, human TANGO 393 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ID NO:75 (SEQ ID NO:76). In another embodiment, mouse TANGO 393 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ED NO:95 (SEQ ED NO:96). In another embodiment, a TANGO 402 protein contains a signal sequence of about amino acids 1 to 50 of SEQ ID NO: 112 (SEQ ED NO: 114). In another embodiment, a MANGO 346 protein contains a signal sequence of about amino acids 1 to 19 of SEQ ED NO:125 (SEQ ID NO:126). In another embodiment, a MANGO 349 protein contains a signal sequence of about amino acids 1 to 26 of SEQ ID NO: 130 (SEQ ID NO: 131). The signal sequence is usually cleaved during processing of the mature protein. In the case of, e.g. , transmembrane 4-type proteins, the signal peptide is generally not cleaved, but becomes a transmembrane-anchoring domain of the polypeptide.
A TANGO 339 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. In one embodiment, a TANGO 339 protein contains extracellular domains at about amino acid residues 43 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO:20 and SEQ ID
NO:21, respectively), transmembrane domains at about amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO:15, SEQ ID NO:16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at about amino acid residues 85 to 92 and 255 to 270 of SEQ ID NO:3 (SEQ ID NO:22 and SEQ ID NO:23, respectively). In this embodiment, the mature TANGO 339 protein coπesponds to amino acids 43 to 270 of SEQ ED NO:3 (SEQ ED NO:4). In another embodiment, a TANGO 339 protein contains extracellular domains at about amino acid residues 1 to 16, 85 to 92, and 255 to 270 of SEQ ID NO:3 (SEQ ED NO:l 1, SEQ ED NO: 12, and SEQ ED NO: 13, respectively), transmembrane domains at about amino acid residues 17 to 41, 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ED NO:14, SEQ ID NO:15, SEQ ID NO:16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at about amino acid residues 42 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO: 18 and SEQ ID NO: 19, respectively). In this embodiment, the mature TANGO 339 protein coπesponds to amino acids 1 to 270 of SEQ ID NO:3.
A TANGO 339 family member can include a signal sequence. In certain embodiment, a TANGO 339 family member has the amino acid sequence of SEQ ID
NO:3, and the signal sequence is located at amino acids 1 to 40, 1 to 41, 1 to 42, 1 to 43 or 1 to 44. In such embodiments of the invention, the domains and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 40 results in an extracellular domain consisting of amino acids 41 to 61 of SEQ ID NO: 3 and the mature TANGO 339 protein coπesponding to amino 41 to 270.
A TANGO 339 family member can include one or more transmembrane 4 or transmembrane 4-like domains. A transmembrane 4 domain typically has the following consensus sequence: G-xxx-[LIVMF]-xx-[GSA]-[LIVMF][LIVMF]-G-C-x-[GA]-[STA]- xx-[EG]-xx-[CWN]-[LIVM][LIVM], wherein G is a glycine residue, "x" is any amino acid, [L1VMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue. A transmembrane 4 domain is a characteristic of transmembrane 4 superfamily members which include, for example, CD9 antigen, CD37, CD53, CD63, CD81, and CD82. Transmembrane 4 proteins have the following characteristics: they are type III membrane proteins, which contain an N-terminal membrane-anchoring domain that is not cleaved during biosynthesis and that functions both as a translocation signal and as a membrane anchor; they contain a total of four transmembrane domains and at least seven conserved cysteine residues; and they are approximately 218 to 284 amino acid residues.
A transmembrane 4-like domain as described herein can have the following consensus sequence: G-xxx-[LIVMF]-xx-[GSA]-[LIVMF]-x-G-C-x-[GA]-[STA]-xx- [EG]-xx-[CWN]-[LIVM][LEVM], wherein G is a glycine residue, "x" is any amino acid, [LIVMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue.
In one embodiment, a TANGO 339 family member has the amino acid sequence of SEQ ID NO:3 and, preferably, a transmembrane 4 domain-like consensus sequence is located at about amino acid positions 69 to 91 of SEQ ED NO:3 (SEQ ED NO:7). In another embodiment, a TANGO 339 family member has the amino acid sequence of SEQ ID NO: 3 and, preferably, a transmembrane 4-like domain is located at about amino acid positions 68 to 260 of SEQ ED NO:3 (SEQ ID NO:6). En another embodiment, a TANGO 339 family member includes one or more transmembrane 4-like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 69 to 91 of SEQ ID NO:3 (SEQ ID NO:7). In yet another embodiment, a TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ID NO:6).
In another embodiment, a TANGO 339 family member includes one or more transmembrane 4-like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 69 to 91 of SEQ ID NO:3 (SEQ ID NO:7), and has at least one TANGO 339 biological activity as described herein. In yet another embodiment a TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ED NO:6), and has at least one TANGO 339 biological activity as described herein.
En another embodiment, the transmembrane 4-like domain of TANGO 339 is a transmembrane 4 domain, which has the following consensus sequence: G-xxx-[LEVMF]- xx-[GSA]-[LIVMF][LIVMF]-G-C-x-[GA]-[STA]-xx-[EG]-xx-[CWN]-[LIVM][LIVM], wherein G is a glycine residue, "x" is any amino acid, [LIVMF] is a leucine, isoleucine, valine, methionine or phenylalanine residue, [GA] is either a glycine or an alanine residue, [STA] is a serine, threonine or alanine residue, [EG] is either a glutamic acid or glycine residue, [CWN] is cysteine, tryptophan or asparagine residue. In this embodiment, a
TANGO 339 family member includes one or more transmembrane 4-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 68 to 261 of SEQ ID NO:3 (SEQ ID NO:6). In another embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 or peripherin/rom-1 -like domains. A peripherin/rom-1 domain typically has the following consensus sequence: D-G-V-P-F-S-C-C-N-P-x-S-P-R-P-C, wherein D is an aspartic acid residue, G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, S is a serine residue, C is a cysteine residue, N is an asparagine residue, x is any amino acid, and R is an arginine residue. Peripherin/rom-1 domains are characteristic of retinal-specific integral membrane proteins that are located at the rims of the photoreceptor disks and that function in disk morphogenesis. Peripherin (or RDS) and rom-1 are examples of proteins that contain the peripherin/rom-1 domain. Defects in the peripherin gene have been shown to cause various diseases, including autosomal dominant retinitis pigmentosa, autosomal dominant punctata albescens, and butterfly-shaped pigment dystrophy.
A peripherin/rom-1 -like domain as described herein has the following consensus sequence: G-V-P-F-S-C-C-x-P, wherein G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, and C is a cysteine residue. In one embodiment, a TANGO 339 family member has the amino acid sequence of SEQ ID NO:3 and, preferably, a peripherin/rom-1 -like domain consensus sequence is located at about amino acid positions 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9). In another embodiment, a TANGO 339 family member has the amino acid sequence of SEQ ID NO:31 and, preferably, a peripherin/rom-1 -like domain is located at about amino acid positions 18 to 270 of SEQ ED NO:3 (SEQ ID NO: 8). In another embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 -like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9). In another embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 -like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 15%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 18 to 270 of SEQ ID NO:3 (SEQ ID NO:8).
In another embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 -like domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%), more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 181 to 189 of SEQ ID NO:3 (SEQ ID NO:9), and has at least one TANGO 339 biological activity as described herein. In yet another embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 -like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 18 to 270 of SEQ ID NO:3 (SEQ ED NO:8), and has at least one TANGO 339 biological activity as described herein.
En another embodiment, the peripherin/rom-1 -like domain of TANGO 339 is a peripherin/rom-1 domain, which has the following consensus sequence: D-G-V-P-F-S-C- C-N-P-x-S-P-R-P-C, wherein D is an aspartic acid residue, G is a glycine residue, V is a valine residue, P is a proline residue, F is a phenylalanine residue, S is a serine residue, C is a cysteine residue, N is an asparagine residue, x is any amino acid, and R is an arginine residue. In this embodiment, a TANGO 339 family member includes one or more peripherin/rom-1 -like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 18 to 270 of SEQ ID NO:3 (SEQ ID NO:8).
A TANGO 353 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. Thus, in one embodiment, an TANGO 353 protein contains an extracellular domain of about amino acids 1 to 116 of SEQ ID NO:29, or a mature extracellular domain of about amino acids 15 to 116 of SEQ ID NO:29 (SEQ ID NO:32). In another embodiment, a TANGO 353 protein contains a transmembrane domain of about amino acids 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33). In another embodiment, a TANGO 353 protein contains a cytoplasmic domain of about amino acids 142 to 230 of SEQ ID NO:29 (SEQ ED NO:34). En yet another embodiment, a TANGO 353 protein is a mature protein containing an extracellular, transmembrane and cytoplasmic domain of about amino acids 15 to 230 of SEQ ID NO:29 (SEQ ID NO:30). A TANGO 353 family member can include a signal sequence. In certain embodiments, a TANGO 353 family member has the amino acid sequence of SEQ ID NO:29, and the signal sequence is located at amino acids 1 to 12, 1 to 13, 1 to 14, 1 to 15 or 1 to 16. In such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 12 results in an extracellular domain consisting of amino acids 13 to 116 of SEQ ID NO:29 and the mature TANGO 353 protein coπesponding to amino 13 to 230.
A TANGO 358 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. In one embodiment, a TANGO 358 protein contains an extracellular domain at amino acids 1 to about 49 of SEQ ID NO:38 or a mature extracellular domain at about amino acid residues 43 to 49 of SEQ ID NO:38 (SEQ ED NO:41), a transmembrane domain at about amino acid residues 50 to 66 of SEQ ID NO:38 (SEQ ID NO:42), and a cytoplasmic domain at about amino acid residues 67 to 82 of SEQ ID NO:38 (SEQ ID NO:43). A TANGO 358 family member can include a signal sequence. In certain embodiment, a TANGO 358 family member has the amino acid sequence of SEQ ID NO:38, and the signal sequence is located at amino acids 1 to 40, 1 to 41, 1 to 42, 1 to 43 or 1 to 44. In such embodiments of the invention, the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 40 results in an extracellular domain consisting of amino acids 41 to 50 of SEQ ID NO:38 and the mature TANGO 368 protein coπesponding to amino 41 to 82.
A TANGO 365 family member can include a signal sequence. In certain embodiments, a TANGO 365 family member has the amino acid sequence of SEQ ED NO:46, and the signal sequence is located at amino acids 1 to 34, 1 to 35, 1 to 36, 1 to 37 or 1 to 38. En such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 36 results in a mature TANGO 365 protein coπesponding to amino 37 to 165 of SEQ ID NO:46 (SEQ ID NO:47).
A TANGO 365 family member can include one or more of the following domains: (1) an extracellular domain; (2) two transmembrane domains; and (3) a cytoplasmic domain. Thus, in one embodiment, a TANGO 365 protein contains an extracellular domain of about amino acids 95 to 165 of SEQ ED NO:46 (SEQ ED NO:51), or a mature extracellular domain of about amino acids 30 to 246 of SEQ ID NO:46. In another embodiment, a TANGO 365 protein contains a first transmembrane domain of about amino acids 52 to 70 of SEQ ED NO:46 (SEQ ED NO:49). In another embodiment, an protein contains a cytoplasmic domain of about amino acids 71 to 77 of SEQ ID NO:46 (SEQ ID NO: 133). In another embodiment, a TANGO 365 protein contains a second transmembrane domain of about amino acids 78 to 94 of SEQ ID NO:46 (SEQ ED NO:50). En yet another embodiment, a TANGO 365 protein is a mature protein containing an extracellular domain, two transmembrane domains and a cytoplasmic domain of about amino acids 37 to 165 of SEQ ID NO:46 (SEQ ID NO:48).
A TANGO 368 family member can include a signal sequence. In certain embodiments, a TANGO 368 family member has the amino acid sequence of SEQ ID NO:54, and the signal sequence is located at amino acids 1 to 25, 1 to 26, 1 to 27, 1 to 28 or 1 to 29. In such embodiments of the invention, the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 27 results in a mature TANGO 368 protein coπesponding to amino 28 to 59 of SEQ ID NO:54 (SEQ ED NO:55). A TANGO 369 family member can include a signal sequence. En certain embodiments, a TANGO 369 family member has the amino acid sequence of SEQ ED NO:60, and the signal sequence is located at amino acids 1 to 24, 1 to 25, 1 to 26, 1 to 27 or 1 to 28. In such embodiments of the invention, the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 26 results in a mature TANGO 368 protein coπesponding to amino 27 to 58 of SEQ ID NO:60 (SEQ ED NO:61).
A TANGO 383 family member can include a signal sequence. En certain embodiments, a TANGO 383 family member has the amino acid sequence of SEQ ED NO:65, and the signal sequence is located at amino acids 1 to 18, 1 to 19, 1 to 20, or 1 to 21. In such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 20 results in a mature TANGO 383 protein coπesponding to amino 21 to 140 of SEQ ID NO:65.
A TANGO 383 family member can include one or more of the following domains: (1) an extracellular domain; (2) two transmembrane domains; and (3) a cytoplasmic domain. In one embodiment, a TANGO 383 protein contains a cytoplasmic domain of about amino acids 21 to 49 of SEQ ID NO:65. In another embodiment, a TANGO 383 protein contains a first transmembrane domain of about amino acids 50 to 70 of SEQ ID NO:65 (SEQ ID NO:68). In another embodiment, a TANGO 383 protein contains an extracellular domain of about amino acids 71 to 115 of SEQ ID NO:65 (SEQ ED NO:70). En another embodiment, a TANGO 383 protein contains a second transmembrane domain of about amino acids 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69). In yet another embodiment, a TANGO 383 protein is a mature protein containing an extracellular domain, two transmembrane domains and a cytoplasmic domain of about amino acids 21 to 140 of SEQ ED NO:65 (SEQ ED NO:67). In another example, a TANGO 393 family member can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain; and (4) a leucine-rich domain. In one embodiment, a TANGO 393 protein contains an extracellular domain at amino acids 1 to about 447 of SEQ ID NO: 75 or a mature extracellular domain at about amino acid residues 27 to 447 of SEQ ED NO: 75 (SEQ ED NO:89), a transmembrane domain at about amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a cytoplasmic domain at about amino acid residues 468 to 473 of SEQ ID NO:75 (SEQ ID NO: 134). In another embodiment, a TANGO 393 family member contains an extracellular domain at amino acids 1 to about 26 of SEQ ID NO:95 or a mature extracellular domain at about amino acid residues 27 to 449 of SEQ ID NO:95 (SEQ ID NO: 109), a transmembrane domain at about amino acid residues 450 to 467 of SEQ ID NO:95 (SEQ ID NO:98), and a cytoplasmic domain at about amino acid residues 468 to 473 of SEQ ID NO:95 (SEQ ID NO:35).
A TANGO 393 family member can include one or more leucine-rich-repeat (LRR) domains. A leucine-rich-repeat domain typically has the following degenerate consensus sequence: x-L-x-x-L-x-L-x-x-[NCT]-x-L-x-x-x-L-x-x-x-x-L-x-x-L, wherein L is a leucine residue and can be replaced by any aliphatic residue,"x" is any amino acid, and [NCT] is either an asparagine, cysteine or threonine, respectively. Leucine-rich-repeat domains most frequently appear in tandem repeats. The degenerate leucine-rich-repeat domains are characteristic of a diverse set of signaling proteins that are involved in cell signaling, cell growth and cell differentiation. Defects in leucine-rich-repeat genes have been shown to cause various diseases which include but are not limited to Bernard-Soulier disease, a bleeding disorder. Furthermore, leucine-rich-repeat genes are involved in the pathogenesis of diseases, for example, the leucine-rich-repeat of type- 1 human immunodeficiency virus Rev protein is the trans-activating region of the virus (Kobe and Deisenhofer, 1994, TIBS, 19:415-421).
In one embodiment, a TANGO 393 family member has the amino acid sequence of SEQ ED NO:76 and, preferably, a leucine-rich-repeat domain consensus sequence is located at about amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 (SEQ ID NO:76), SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively. In another embodiment, a TANGO 393 family member has the amino acid sequence of SEQ ID NO:95 and, preferably, a leucine-rich-repeat domain is located at about amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively. In another embodiment, a TANGO 393 family member includes one or more leucine-rich-repeat domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 of SEQ ID NO:76 (SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively). In another embodiment, a TANGO 393 family member includes one or more leucine-rich-repeat domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively. In another embodiment, a TANGO 393 family member includes one or more leucine-rich-repeat domain consensus sequences having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of human TANGO 393 of SEQ ID NO:76 (SEQ ID NO:79, 80, 81, 83, 83, 84, 85, 86, 87 and 88, respectively), and has at least one TANGO 393 biological activity as described herein. In yet another embodiment, a TANGO 393 family member includes one or more leucine-rich-repeat domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acid positions 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and/or 260 to 310 of mouse TANGO 393 (SEQ ID NO:95), SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively, and has at least one TANGO 393 biological activity as described herein. A TANGO 402 family member can include of one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. In one embodiment, a TANGO 402 protein contains an extracellular domain at amino acids 1 to about 133 of SEQ ID NO:l 12 or a mature extracellular domain at about amino acid residues 51 to 133 of SEQ ED NO: 112 (SEQ ID NO:l 15), a transmembrane domain at about amino acid residues 134 to 151 of SEQ ED NO:112 (SEQ ED NO: 116), and a cytoplasmic domain at about amino acid residues 152 to 207 of SEQ ED NO:l 12 (SEQ ID NO: 117).
A TANGO 402 family member can include a signal sequence. In certain embodiments, a TANGO 402 family member has the amino acid sequence of SEQ ID NO:l 12, and the signal sequence is located at amino acids 1 to 48, 1 to 49, 1 to 50, 1 to 51 or 1 to 52. In such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 48 results in an extracellular domain consisting of amino acids 49 to 133 of SEQ ID NO:l 12 and the mature TANGO 402 protein coπesponding to amino 49 to 207 of SEQ ID NO:l 12.
A TANGO 402 family member can include a C-type lectin domain or a C-type lectin-like domain.
A C-type lectin domain typically has the following consensus sequence: C- [LIVMFATG]-x(5,12)-[WL]-x-[DNSR]-x(2)-C-x(5,6)-[FYWLIVSTA]-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, x is any amino acid and the number in parentheses indicates the number of amino acids, [WL] is either a tryptophan or leucine residue, [DNSR] is a aspartic acid, asparagine, serine or arginine residue, [FYWLEVSTA] is a phenylalanine, tyrosine, tryptophan, leucine, isoleucine, valine, serine, threonine or alanine residue, and [LIVSTA] is a leucine, isoleucine, valine, serine, threonine or alanine residue. C-type lectin domains contain four cysteines, which are involved in two disulfide bonds, and are about 110 to 130 amino acid residues. C-type lectin domains typically function as calcium-dependent carbohydrate-recognition domains and have been found in various proteins including, but not limited to, asialoglycoprotein receptors (ASGPR), pulmonary surfactant-associated protein A (SP-A), mannan-binding proteins, L-selectin, neurocan, and tetranectin. These proteins have various functions including, for example, cell adhesion (i.e., L-selectin). ASGPR mediates the endocytosis of plasma glycoprotein to which the terminal salic acid-residue in their carbohydrated moieties has been removed. SP-A binds to surfactant phospholipids and contributes to lower the surface tension at the air-liquid interface in the alveoli of the lung.
A C-type lectin-like domain as described herein has the following consensus sequence: C-[LIVMFATG]-x-(5,12)-[DNSR]-x(2)-C-x(5,6)-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, "x" is any amino acid and the number in parentheses indicates the number of amino acids [DNSR] is an aspartic acid, asparagine, serine or arginine residue, and [LIVSTA] is a leucine, isoleucine, valine, serine, threonine or alanine residue. In one embodiment, a TANGO 402 family member has the amino acid sequence of SEQ ID NO: 112 and, preferably, a C-type lectin-like domain is located at about amino acid positions 104 to 193 of SEQ ID NO:l 12 (SEQ ID NO:l 18), wherein the consensus sequence is at about amino acid positions 172 to 193 of SEQ ID NO: 112 (SEQ ID NO: 119).
In another embodiment, a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118).
In another embodiment, a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118), and has at least one TANGO 402 biological activity as described herein.
In another embodiment, a TANGO 402 family member includes one or more C- type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118) and includes a cysteine residue N-terminal to the consensus sequence. In yet another embodiment, a TANGO 402 family member includes one or more C-type lectin-like domain having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO:l 12 (SEQ ID NO:l 18), includes a cysteine residue N-terminal to the consensus sequence, and has at least one TANGO 402 biological activity as described herein.
In another embodiment, the C-type lectin-like domain of TANGO 402 is a C-type lectin domain, which has the following consensus sequence: C-[LIVMFATG]-x(5,12)- [WL]-x-[DNSR]-x(2)-C-x(5,6)-[FYWLEVSTA]-[LIVSTA]-C, wherein C is a cysteine residue, [LIVMFATG] is a leucine, isoleucine, methionine, phenylalanine, alanine, threonine or glycine residue, x is any amino acid, [WL] is either a tryptophan or leucine residue, [DNSR] is a aspartic acid, asparagine, serine or arginine residue, [FYWLIVSTA] is a phenylalanine, tyrosine, tryptophan, leucine, isoleucine, valine, serine, threonine or alanine residue, and [LIVSTA] is a leucine, isoleucine, valine, serine, threonine or alanine residue. In this embodiment, a TANGO 402 family member includes one or more C-type lectin-like domains having an amino acid sequence that is at least about 55%, preferably at least about 65%, more preferably at least 75%, yet more preferably at least about 85%, and most preferably at least about 95% identical to amino acids 104 to 193 of SEQ ID NO: 112 (SEQ ID NO: 118).
A MANGO 346 family member can include a signal sequence. In certain embodiments, a MANGO 346 family member has the amino acid sequence of SEQ ID NO:125, and the signal sequence is located at amino acids 1 to 17, 1 to 18, 1 to 19, 1 to 20 or 1 to 21. In such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 19 results in the mature MANGO 346 protein coπesponding to amino 20 to 60 (SEQ ID NO: 127). A MANGO 349 family member can include a signal sequence. In certain embodiments, a MANGO 349 family member has the amino acid sequence of SEQ ID NO: 130, and the signal sequence is located at amino acids 1 to 24, 1 to 25, 1 to 26, 1 to 27 or 1 to 28. In such embodiments of the invention, the extracellular domain and the mature protein resulting from cleavage of such signal peptides are also included herein. For example, the cleavage of a signal sequence consisting of amino acids 1 to 26 results in the mature MANGO 349 protein coπesponding to amino 27 to 167 of SEQ ID NO: 130 (SEQ ID NO: 132).
Various features of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 are summarized below.
Human TANGO 339
A cDNA encoding human TANGO 339 was identified by analyzing the sequences of clones present in a human fetal library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthgalOOgOl, encoding full-length human TANGO 339. The human TANGO 339 cDNA of this clone is 2715 nucleotides long (Figure 1; SEQ ID NO:l). The open reading frame of this cDNA, nucleotides 210 to 1019 of SEQ ID NO:l (SEQ ID NO:2), encodes a 270 amino acid transmembrane protein (Figure 1; SEQ ID NO:3).
Figure 2 depicts a hydropathy plot of human TANGO 339. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues
(cys) and N-glycosylation site are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 339 includes a 42 amino acid signal peptide (amino acid 1 to amino acid 42 of SEQ ID NO:3; SEQ ID NO: 5) preceding the mature human TANGO 339 protein (coπesponding to amino acid 43 to amino acid 270 of SEQ ID NO:3; SEQ ID NO:4). In instances wherein the signal peptide is cleaved, the molecular weight of human TANGO 339 protein without post-translational modifications is 30.7 kDa prior to the cleavage of the signal peptide, and 25.6 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 56, 67 and 72 of SEQ ID NO: 3 indicates that there can be alternative forms of human TANGO 339 of 215 amino acids of SEQ ID NO:3, 204 amino acids of SEQ ID NO:3, and 199 amino acids of SEQ ID NO:3, respectively.
Human TANGO 339 protein is a transmembrane protein that contains extracellular domains at amino acid residues 43 to 61 and 116 to 232 of SEQ ED NO:3 (SEQ ED NO:20 and SEQ ID NO:21, respectively), transmembrane domains at amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 15, SEQ ED NO: 16 and SEQ ID NO: 17, respectively), and cytoplasmic domains at amino acid residues 85 to 92 and 255 to 270 of SEQ ID NO:3 (SEQ ID NO:22 and SEQ ID NO:23, respectively). In instances wherein the signal peptide is not cleaved, human TANGO 339 has extracellular domains at amino acid residues 1 to 16, 85 to 92, and 255 to 270 of SEQ ID NO:3 (SEQ ID NO: 11, SEQ ED NO: 12, and SEQ ED NO: 13, respectively), transmembrane domains at amino acid residues 17 to 41, 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, respectively), and cytoplasmic domains of amino acid residues 42 to 61 and 116 to 232 of SEQ ID NO:3 (SEQ ID NO: 18 and SEQ ID NO: 19, respectively).
Alternatively, in another embodiment, a human TANGO 339 protein contains cytoplasmic domains at amino acid residues 43 to 61 and 116 to 232 of SEQ ID NO: 3, transmembrane domains at amino acid residues 62 to 84, 93 to 115, and 233 to 254 of SEQ ID NO:3 (SEQ ID NO: 15, SEQ ID NO: 16 and SEQ ID NO: 17, respectively), and extracellular domains at amino acid residues 85 to 92 and 255 to 270 of SEQ ED NO:3.
En one embodiment of a nucleotide sequence of human TANGO 339, the nucleotide at position 29 is adenine (A)(SEQ ED NO:2). En this embodiment, the amino acid at position 10 is lysine (K)(SEQ ED NO:3). In an alternative embodiment, a species variant of human TANGO 339 has a nucleotide at position 29 which is guanine (G)(SEQ ID NO: 136). In this embodiment, the amino acid at position 10 is arginine (R)(SEQ ID NO:137), i.e., a conservative substitution.
En another embodiment of a nucleotide sequence of human TANGO 339, the nucleotide at position 59 is thymine (T)(SEQ ED NO:2). En this embodiment, the amino acid at position 20 is phenylalanine (F)(SEQ ID NO:3). In an alternative embodiment, a species variant of human TANGO 339 has a nucleotide at position 59 which is adenine (A)(SEQ ID NO: 138). In this embodiment, the amino acid at position 20 is tyrosine (Y)(SEQ ID NO:139), i.e., a conservative substitution.
In another embodiment of a nucleotide sequence of human TANGO 339, the nucleotide at position 119 is cytosine (C)(SEQ ID NO:2). In this embodiment, the amino acid at position 40 is alanine (A)(SEQ ID NO:3). In an alternative embodiment, a species variant of human TANGO 339 has a nucleotide at position 119 which is thymine (T)(SEQ ID NO: 140). In this embodiment, the amino acid at position 40 is valine (V)(SEQ ID NO: 141), i.e., a conservative substitution. In another embodiment of a nucleotide sequence of human TANGO 339, the nucleotide at position 180 is cytosine (C)(SEQ ID NO:2). In this embodiment, the amino acid at position 60 is aspartate (D)(SEQ ID NO:3). In an alternative embodiment, a species variant of human TANGO 339 has a nucleotide at position 180 which is guanine (G)(SEQ ID NO: 142). In this embodiment, the amino acid at position 60 is glutamate (E)(SEQ ID NO: 143), i.e., a conservative substitution.
Human TANGO 339 includes a transmembrane 4-like domain (at amino acids 68 to 260 of SEQ ID NO:3; SEQ ID NO:6) and a peripherin /rom-1-like domain (at amino acids 18 to 270 of SEQ ID NO:3; SEQ ID NO:8).
Human TANGO 339 has an N-glycosylation site with the sequence NCSG (at amino acid residues 169 to 172 of SEQ ED NO:3). Two protein kinase C phosphorylation sites are present in human TANGO 339. The first has the sequence SEK (at amino acid residues 42 to 44 of SEQ ED NO:3) and the second has the sequence SYR (at amino acid residues 133 to 135 of SEQ ED NO:3). Human TANGO 339 has three casein kinase EE phosphorylation sites. The first has the sequence SYRD (at amino acid residues 133 to 136 of SEQ ID NO:3), the second has the sequence SKWD (at amino acid residues 210 to 213 of SEQ ID NO:3), and the third has the sequence SDIE (at amino acid residues 259 to 262 of SEQ ED NO:3). Six N-myristylation sites are present in human TANGO 339. The first has the sequence GCVGAL (at amino acid residues 79 to 84 of SEQ ID NO:3), the second has the sequence GASYSR (at amino acid residues 172 to 177 of SEQ ID NO:3), the third has the sequence GVPFSC (at amino acid residues 181 to 186 of SEQ ED NO:3), the fourth has the sequence GCEQAL (at amino acid residues 220 to 225 of SEQ ED NO:3), the fifth has the sequence GVFIAI (at amino acid residues 238 to 243 of SEQ ID NO:3), and the sixth has the sequence GIFLAR (at amino acid residues 250 to 255 of SEQ ID NO:3). Human TANGO 339 has a prokaryotic membrane lipoprotein lipid attachment site with the sequence VVMFTLGFAGC (at amino acid residues 70 to 80 of SEQ ID NO:3; SEQ ID NO:10).
The human TANGO 339 gene maps to human chromosome 10 between markers D10S201 and D10S551. As retinal G protein coupled receptor and pulmonary-associated protein Al map to this region of chromosome 10, TANGO 339 nucleic acids, proteins and modulators thereof can be used to diagnose disorders associated with G protein coupled receptors and/or modulate G protein coupled receptor-related processes, e.g., retinal processes and/or pulmonary-related processes.
Figure 3 shows an alignment of the human TANGO 339 amino acid sequence (SEQ ID NO:3) with the human CD9 antigen amino acid sequence (SEQ ID NO:24; Accession Number NM_001769). The alignment shows that there is a 24.1% overall amino acid sequence identity between human TANGO 339 and human CD9 antigen. The CD9 antigen is a widely expressed cell surface glycoprotein that has been shown to be involved in such processes as cell activation, proliferation, and adhesion. For example, CD9 antigen expression on platelets mediates platelet activation and aggregation. CD9 antigen has also been shown to be expressed by neural cells and can play a role in intercellular signaling in the nervous system, in particular, controlling cellular attraction or repulsion in guiding neural growth to target points. Further, the CD9 antigen has been shown to associate with beta 1 integrins and other transmembrane 4 superfamily members, including CD81 and CD82. As such TANGO 339 proteins, nucleic acids and modulators thereof could be useful in modulating cellular interaction such as between immune cells, and also can be involved in modulating intercellular signaling, such as neural cell intercellular signaling.
Figure 4 shows an alignment of the nucleotide sequence of human CD9 antigen coding region (SEQ ED NO:25; Accession Number NM_001769) and the nucleotide sequence of human TANGO 339 coding region (SEQ ID NO:2). The alignment shows a 45.9 % overall sequence identity between the two nucleotide sequences. The full-length human CD9 antigen nucleic acid sequence (SEQ ID NO:26; Accession Number NP_001760) and human TANGO 339 cDNA (SEQ ID NO:l) have an overall sequence identity of 30.3%.
Clone EpT339, which encodes human TANGO 339, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-292. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 339 Nucleic acids, Polypeptides. and Modulators Thereof
As TANGO 339 was originally found in a human fetal library, TANGO 339 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of cells, tissues and/or organs, e.g., the proliferation of tissues and cells of lymphoid origin and neural origin. TANGO 339 nucleic acids, proteins and modulators thereof can be used to treat immune related disorders, e.g., immunodeficiency disorders (e.g., HIV), viral disorders, cancers, autoimmune disorders, (e.g., arthritis and graft rejection) and inflammatory disorders (e.g. , bacterial or viral infection, psoriasis, septicemia, arthritis, allergic reactions). TANGO 339 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the development of cells, tissues and/or organs in the embryo and/or fetus.
In light of the fact that TANGO 339 has characteristics of transmembrane 4 proteins, TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate (e.g., stabilize, promote, inhibit or disrupt) cellular activation, cellular proliferation, motility, and differentiation. For example, such TANGO 339 compositions and modulators thereof can be used to modulate binding to extracellular matrix (ECM)- associated factors such as integrins and can function to modulate ligand binding to cell surface receptors. In further light of the fact that TANGO 339 has characteristics of transmembrane 4 proteins, TANGO 339 nucleic acids, proteins and modulators thereof can be used to modulate disorders associated with abeπant signal transduction in response to ECM- associated proteins and cell surface receptors such as other transmembrane 4 proteins. TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development and progression of proliferative disorders, e.g., neoplasms or tumors
(such as carcinomas, sarcomas, adenomas or myeloid lymphomas) associated with cancer, (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leimyosarcoma, rhabdothehosarcoma, colon sarcoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hematoma, bile duct carcinoma, melanoma, choriocarcinoma, semicoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependynoma, pinealoma, hemangioblastoma, retinoblastoma; leukemias, e.g. acute lymphocytic leukemia and acute myelocytic leukemia (myelolastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphoma
(Hodgkin's disease and non-Hodgkin's diseases), multiple myeloma and Waldenstrom's macroglobulinemia.
TANGO 339 proteins exhibit similarity to human CD9 antigen, a member of the transmembrane 4 superfamily. In light of this, TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate platelet activation and aggregation. For example, antagonists to TANGO 339 action, such as peptides, antibodies or small molecules that decrease or block TANGO 339 binding to extracellular matrix components (e.g., integrins) or that prevent TANGO 339 signaling, can be used as platelet activation and aggregation blockers. In another example, agonists that mimic TANGO 339 activity, such as peptides, antibodies or small molecules, can be used to induce platelet activation and aggregation. Antibodies may activate or inhibit the cell adhesion, proliferation and activation, and may help in treating inflammation, cancer, cardiovascular disease or stroke by affecting these cellular processes. TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate platelet-related processes and disorders, e.g., Glanzmann's thromboasthemia, which is a bleeding disorder characterized by failure of platelet aggregation in response to cell stimuli.
In further light of the fact that TANGO 339 proteins exhibit similarity to human CD9 antigen, TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling in the nervous system. The CD9 antigen, which is expressed at the surface of central nervous system (CNS) mature myelin, may modulate intercellular signal transduction and enhance myelin membrane adhesion to extracellular matrices at very late stages of development, thereby playing a role in the maintenance of the entire myelin sheath.
In light, in part, of the fact that TANGO 339 proteins contain peripherin/rom-1 -like domains, TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development and function of the eye, such as retinal development and function, (e.g., photoreceptor disk morphogenesis). TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to treat eye diseases and/or disorders, e.g., autosomal dominant retinitis pigmentosa, autosomal dominant punctata albescens, butterfly-shaped pigment dystrophy, cataracts, macular degeneration, myopia, stigmatism and retinoblastoma.
As TANGO 339 maps to a region of chromosome 10 which encodes polypeptides expressed in the lung, TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate the development, differentiation and activity of pulmonary structures, e.g., lung. TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate or treat pulmonary disorders, such as atelectasis, pulmonary congestion or edema, cystic fibrosis, chronic obstructive airway disease (e.g., emphysema, chronic bronchitis, bronchial asthma, and bronchiectasis), diffuse interstitial diseases (e.g., sarcoidosis, pneumoconiosis, hypersensitivity pneumonitis, Goodpasture's syndrome, idiopathic pulmonary hemosiderosis, pulmonary alveolar proteinosis, desquamative interstitial pneumonitis, chronic interstitial pneumonia, fibrosing alveolitis, hamman-rich syndrome, pulmonary eosinophilia, diffuse interstitial fibrosis, Wegener's granulomatosis, lymphomatoid granulomatosis, and lipid pneumonia), lung cancer or tumors (e.g., bronchogenic carcinoma, bronchiolovlveolar carcinoma, bronchial carcinoid, hamartoma, and mesenchymal tumors). As TANGO 339 nucleic acids exhibit homology to a human brain EST (Accession
Number Q59384, disclosed in Patent No. WP 93/16178), TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in the development, differentiation and activity of the brain, including, but not limited to development, differentiation and activation of neuronal cells and glial cells (e.g., oligodendrocytes astrocytes), and amelioration of one or more symptoms associated with abnormal function of such cell types. TANGO 339 nucleic acids, proteins and modulators thereof can be utilized to treat neural diseases and/or disorders, e.g. epilepsy, spinal cord injuries, infarction, infection, malignancy, paraneoplastic syndromes, neuropsychiatric disorders (e.g., schizophrenia, depression, anxiety disorders, and anorexia nervosa), and neurodegenerative diseases including, but not limited to, Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis and progressive supra-nuclear palsy.
TANGO 339 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the brain) and/or cells (e.g., neurons) in which TANGO 339 is expressed. TANGO 339 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping. Human TANGO 353
A cDNA encoding human TANGO 353 was identified by analyzing the sequences of clones present in a mixed lymphocyte reaction library for sequences that encode a wholly secreted or transmembrane protein. This analysis led to the identification of a clone, jthLa03 lgl2 encoding full-length human TANGO 353. The human TANGO 353 cDNA of this clone is 1239 nucleotides long (Figure 5; SEQ ID NO:27). The open reading frame of this cDNA, nucleotides 76 to 765 of SEQ ID NO:27 (SEQ ID NO:28), encodes a 230 amino acid transmembrane protein (Figure 5; SEQ ID NO:29). Figure 6 depicts a hydropathy plot of human TANGO 353. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and N-glycosylation sites (NGly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right. The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein
Engineering 10:1-6) predicted that human TANGO 353 includes a 14 amino acid signal peptide (amino acid 1 to amino acid 14 of SEQ ID NO:29; SEQ ID NO:31) preceding the mature human TANGO 353 protein (coπesponding to amino acid 15 to amino acid 230 of SEQ ID NO:29; SEQ ID NO:30). The molecular weight of human TANGO 353 protein without post-translational modifications is 24.8 kDa prior to the cleavage of the signal peptide and 23.3 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 39, 170 and 184 of SEQ ID NO:29 indicates that there can be alternative forms of human TANGO 353 of 192 amino acids of SEQ ED NO:29, 61 amino acids of SEQ ID NO:29, and 47 amino acids of SEQ ID NO:29, respectively. Human TANGO 353 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. The human TANGO 353 protein contains an extracellular domain at amino acid residues 15 to 116 of SEQ ID NO:29 (SEQ ID NO:32), a transmembrane domain at amino acid residues 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33), and a cytoplasmic domain at amino acid residues 142 to 230 of SEQ ID NO:29 (SEQ ID NO:34).
Alternatively, in another embodiment, a human TANGO 353 protein contains a cytoplasmic domain at amino acid residues 15 to 116 of SEQ ID NO:29, a transmembrane domain at amino acid residues 117 to 141 of SEQ ID NO:29 (SEQ ID NO:33), and an extracellular domain at amino acid residues 142 to 230 of SEQ ID NO:29. In one embodiment of a nucleotide sequence of human TANGO 353, the nucleotide at position 68 is thymine (T)(SEQ ID NO:28). In this embodiment, the amino acid at position 23 is valine (V)(SEQ ID NO:29). In an alternative embodiment, a species variant of human TANGO 353 has a nucleotide at position 68 which is cytosine (C)(SEQ ID NO: 144). In this embodiment, the amino acid at position 23 is alanine (A)(SEQ ID NO:145), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 353, the nucleotide at position 77 is adenine (A)(SEQ ID NO:28). In this embodiment, the amino acid at position 26 is tyrosine (Y)(SEQ ID NO:29). In an alternative embodiment, a species variant of human TANGO 353 has a nucleotide at position 77 which is thymine (T)(SEQ ID NO: 146). In this embodiment, the amino acid at position 26 is phenylalanine (F)(SEQ ID NO: 147), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 353, the nucleotide at position 203 is guanine (G)(SEQ ID NO:28). In this embodiment, the amino acid at position 68 is arginine (R)(SEQ ID NO:29). In an alternative embodiment, a species variant of human TANGO 353 has a nucleotide at position 203 which is adenine (A)(SEQ ID NO: 148). In this embodiment, the amino acid at position 68 is histidine (H)(SEQ ID NO: 149), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 353, the nucleotide at position 309 is cytosine (C)(SEQ ID NO:28). In this embodiment, the amino acid at position 103 is aspartate (D)(SEQ ID NO:29). In an alternative embodiment, a species variant of human TANGO 353 has a nucleotide at position 309 which is guanine (G)(SEQ ID NO: 150). In this embodiment, the amino acid at position 103 is glutamate (E)(SEQ ID NO: 151), i.e., a conservative substitution. Four N-glycosylation sites are present in human TANGO 353. The first has the sequence NFTL (at amino acid residues 48 to 51 of SEQ ID NO:29), the second has the sequence NLSG (at amino acid residues 73 to 76 of SEQ ID NO:29), the third has the sequence NQSQ (at amino acid residues 97 to 100 of SEQ ID NO:29), and the fourth has the sequence NVSF (at amino acid residues 109 to 112 of SEQ ED NO:29). Human TANGO 353 has one cAMP- and cGMP-dependent protein kinase phosphorylation site with the sequence KRPT (at amino acid residues 209 to 212 of SEQ ED NO:29). Five protein kinase C phosphorylation sites are present in human TANGO 353. The first has the sequence SIR (at amino acid residues 19 to 21 of SEQ ID NO:29), the second has the sequence SSK (at amino acid residues 78 to 80 of SEQ ID NO:29), the third has the sequence SAK (at amino acids 180 to 182 of SEQ ID NO:29), the fourth has the sequence TRK (at amino acid residues 207 to 209 of SEQ ED NO:29), and the fifth has the sequence TFR (at amino acid residues 225 to 227 of SEQ ID NO:29). Human TANGO 353 has four casein kinase II phosphorylation sites. The first has the sequence SSQE (at amino acid residues 28 to 31 of SEQ ID NO:29), the second has the sequence TMPE (at amino acid residues 183 to 186 of SEQ ID NO:29), the third has the sequence TLDD (at amino acid residues 191 to 194 of SEQ ID NO:29), and the fourth has the sequence SSPE (at amino acid residues 216 to 219 of SEQ ID NO:29). Human TANGO 353 has two N- myristylation sites. The first has the sequence GNFPGA (at amino acid residues 42 to 47 of SEQ ID NO:29) and the second has the sequence GVTFNL (at amino acid residues 69 to 74 of SEQ ID NO:29). Clone EpT353, which encodes human TANGO 353, was deposited with the
American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-292. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 353 Nucleic acids, Polypeptides, and Modulators Thereof
As TANGO 353 was originally found in a mixed lymphocyte library, TANGO 353 nucleic acids, proteins, and modulators thereof can be utilized to diagnose disorders and/or modulate processes involved in lymphocyte development, differentiation and activity, including, but not limited to development, differentiation and activation of T cells, including T helper, T cytotoxic and non-specific T killer cell types and subtypes, and B cells, immune functions associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types. Such disorders can include, but are not limited to, autoimmune disorders (e.g., autoimmune thyroiditis, Type I diabetes mellitus, insulin-resistant diabetes, autoimmune anemia, multiple sclerosis, rheumatoid arthritis, lupus or sclerodoma, allergy, including allergic rhinitis and food allergies, asthma, psoriasis, graft rejection, transplantation rejection, graft versus host disease, pathogenic susceptibilities), inflammatory disorders (e.g., bacterial or viral infections, wound healing and inflammatory bowel disease and arthritis), apoptotic disorders, and cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF).
Other TANGO 353 associated disorders can include TNF related disorders (e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), immunological differentiative and apoptotic disorders (e.g., hyper- proliferative syndromes such as systemic lupus erythematosus (lupus)), and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer). Modulators of TANGO 353 expression and/or activity can be used to treat such disorders.
As TANGO 353 is expressed in mixed lymphocyte cultures, and hence likely expressed in bone marrow, TANGO 353 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders associated with cells in the bone maπow and/or modulate the proliferation, differentiation, and/or function of cells that appear in the bone maπow, e.g., stem cells (e.g., hematopoietic stem cells), and blood cells, e.g., erythrocytes, platelets, and leukocytes. Thus TANGO 353 nucleic acids, proteins, and modulators thereof can be used to treat bone maπow, blood, and hematopoietic associated diseases and disorders, e.g., acute myeloid leukemia, hemophilia, leukemia, anemia (e.g., sickle cell anemia), and thalassemia.
As TANGO 353 is a transmembrane protein, TANGO 353 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling cascades, or alternatively.
TANGO 353 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., spleen) and/or cells (e.g., lymphocytes) in which TANGO 353 is expressed. TANGO 353 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g. , in radiation hybrid mapping.
Human TANGO 358
A cDNA encoding human TANGO 358 was identified by analyzing the sequences of clones present in a fetal thymus library for sequences that encode a wholly secreted or transmembrane protein. This analysis led to the identification of a clone, jthTbl28c07 encoding full-length human TANGO 358. The human TANGO 358 cDNA of this clone is 1608 nucleotides long (Figure 7; SEQ ED NO:36). The open reading frame of this cDNA, nucleotides 184 to 429 of SEQ ID NO:36 (SEQ ID NO:37), encodes an 82 amino acid transmembrane protein (Figure 7; SEQ ID NO:38).
Figure 8 depicts a hydropathy plot of human TANGO 358. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues
(cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10: 1-6) predicted that human TANGO 358 includes a 42 amino acid signal peptide (amino acid 1 to amino acid 42 of SEQ ID NO:36; SEQ ID NO:40) preceding the mature human TANGO 358 protein (coπesponding to amino acid 43 to amino acid 82 of SEQ ID NO:36; SEQ ID NO:39). The molecular weight of human TANGO 358 protein without post-translational modifications is 9.5 kDa prior to the cleavage of the signal peptide and 4.5 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 17, 20 and 63 of SEQ ID NO:38 indicates that there can be alternative forms of human TANGO 358 of 66 amino acids of SEQ ED NO:38, 63 amino acids of SEQ ED NO:38, and 20 amino acids of SEQ ED NO:38, respectively.
Human TANGO 358 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. The human TANGO 358 protein contains an extracellular domain at amino acid residues 43 to 49 of SEQ ID NO:38 (SEQ ED NO:41), a transmembrane domain at amino acid residues 50 to 66 of SEQ ED NO:38 (SEQ ED NO:42), and a cytoplasmic domain at amino acid residues 67 to 82 of SEQ ED NO:38 (SEQ ED NO:43). Alternatively, in another embodiment, a human TANGO 358 protein contains a cytoplasmic domain at amino acid residues 43 to 49 of SEQ ID NO:38, a transmembrane domain at amino acid residues 50 to 66 of SEQ ED NO:38 (SEQ ED NO:42), and an extracellular domain at amino acid residues 67 to 82 of SEQ ID NO:38. Further, human TANGO 358 has a protein kinase C phosphorylation site with the sequence SIK (at amino acid residues 45 to 47 of SEQ ID NO:38). In one embodiment of a nucleotide sequence of human TANGO 358, the nucleotide at position 20 is adenine (A)(SEQ ID NO:37). In this embodiment, the amino acid at position 7 is histidine (H)(SEQ ID NO:38). In an alternative embodiment, a species variant of human TANGO 358 has a nucleotide at position 20 which is guanine (G)(SEQ ID NO: 152). In this embodiment, the amino acid at position 7 is arginine (R)(SEQ ID NO: 153), i. e. , a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 358, the nucleotide at position 35 is thymine (T)(SEQ ID NO:37). In this embodiment, the amino acid at position 12 is valine (V)(SEQ ID NO:38). En an alternative embodiment, a species variant of human TANGO 358 has a nucleotide at position 35 which is cytosine (C)(SEQ ID NO:154). In this embodiment, the amino acid at position 12 is alanine (A)(SEQ ID NO:155), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 358, the nucleotide at position 85 is thymine (T)(SEQ ID NO:37). In this embodiment, the amino acid at position 29 is serine (S)(SEQ ID NO:38). In an alternative embodiment, a species variant of human TANGO 358 has a nucleotide at position 85 which is adenine (A)(SEQ ID NO: 156). In this embodiment, the amino acid at position 29 is threonine (T)(SEQ ED NO:157), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 358, the nucleotide at position 91 is cytosine (C)(SEQ ID NO:37). In this embodiment, the amino acid at position 31 is glutamine (Q)(SEQ ID NO:38). En an alternative embodiment, a species variant of human TANGO 358 has a nucleotide at position 91 which is guanine (G)(SEQ ED NO: 158). In this embodiment, the amino acid at position 31 is glutamate (E)(SEQ ID NO: 159), i.e., a conservative substitution.
Clone EpT358, which encodes human TANGO 358, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999, and assigned Accession Number PTA-292. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 358 Nucleic acids. Polypeptides, and Modulators Thereof
As TANGO 358 was originally found in a fetal thymus library, TANGO 358 nucleic acids, proteins, and modulators thereof can be used to diagnose thymus associated disorders. TANGO 358 nucleic acids, proteins, and modulators thereof can also be used modulate the proliferation, development, differentiation, maturation and/or function of thymocytes, e.g., modulate development and maturation of T-lymphocytes. TANGO 358 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes such as the ability to modulate host immune response by, e.g., modulating the formation of and/or binding to immune complexes, and modulating the positive and negative selection of thymocytes. Such TANGO 358 compositions and modulators thereof can be utilized, e.g., to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to infection and autoimmune disorders (e.g., insulin-dependent mellitus, multiple sclerosis, systemic lupus, erythematosus, sjogren's syndrome, autoimmune thyroiditis, idiotpathic Addison's disease, vitiligo, Grave's disease, idiopathic thrombocytopenia purpura, rheumatoid arthritis, and scleroderma). TANGO 358 nucleic acids, proteins and modulators thereof can also be utilized to treat viral infections, inflammatory immune disorders and immune-related cancers including but not limited to, leukemia (e.g., acute leukemia, chronic leukemia, Hodgkin's disease non-Hodgkin's lymphoma ,and multiple myeloma). Disorders associated with TANGO 358 activity, including those which TANGO 358 proteins, nucleic acids and modulators thereof may be an antagonist can be used to treat include immune disorders, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)). Disorders associated with modulated TANGO 358 activity can also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF).
In light of the fact that TANGO 358 was isolated from a thymus library, TANGO 358 proteins, nucleic acids and modulators thereof can be used to treat disorders that include TNF-related disorders (e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer). Modulators of TANGO 358 expression and/or activity can be used to treat such disorders.
As TANGO 358 is a transmembrane protein, TANGO 358 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extra-cellular matrix.
TANGO 358 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., T-lymphocytes) in which TANGO 358 is expressed. TANGO 358 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 365
A cDNA encoding TANGO 365 was identified by analyzing the sequences of clones present in a human prostate fibroblast library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthqc001g06, encoding full-length Human TANGO 365. The TANGO 365 cDNA of this clone is 1338 nucleotides long (Figure 9; SEQ ID NO:44). The open reading frame of this cDNA, nucleotides 56 to 550 of SEQ ID NO:44 (SEQ ID NO:45), encodes a 165 amino acid transmembrane protein (Figure 9; SEQ ID NO:46). Figure 10 depicts a hydropathy plot of human TANGO 365. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 36 of SEQ ID NO:46; SEQ ID NO:47) on the left from the mature protein (amino acids 37 to 165 of SEQ ID NO:46; SEQ ID NO:48) on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 365 includes a 36 amino acid signal peptide (amino acid 1 to amino acid 36 of SEQ ID NO:46; SEQ ID NO:47) preceding the mature protein (coπesponding to amino acid 37 to amino acid 165 of SEQ ED NO:46; SEQ ED NO:48). The molecular weight of TANGO 365 protein without post-translational modifications is 17.4 kDa prior to the cleavage of the signal peptide, 13.6 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 16, 35 and 81 of SEQ ED NO:46 indicates that there can be alternative forms of human TANGO 365 of 150 amino acids of SEQ ED NO:46, 131 amino acids of SEQ ID NO:46, and 65 amino acids of SEQ ED NO:46, respectively.
Human TANGO 365 is a transmembrane protein which can include one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. The human TANGO 365 protein contains two extracellular domains; one at amino acid residues 37 to 51 of SEQ ID NO:46 (SEQ ED NO:232); and a second at amino acid residues 95 to 165 of SEQ ID NO:46 (SEQ ID NO:51), two hydrophobic transmembrane domains; one at amino acids 52 to 70 of SEQ ED NO:46 (SEQ ID NO:49); and a second at amino acids 78 to 94 of SEQ ID NO:46 (SEQ ID NO:50), and a cytoplasmic domain at amino acid residues 71 to 77 of SEQ ID NO:46 (SEQ ID NO:234). Alternatively, in another embodiment, a human TANGO 365 protein contains two cytoplasmic domains; one at amino acid residues 37 to 51 of SEQ ID NO:46; and a second at amino acid residues 95 to 165 of SEQ ID NO:46, two hydrophobic transmembrane domains; one at amino acids 52 to 70 of SEQ ID NO:46 (SEQ ID NO:49); and a second at amino acids 78 to 94 of SEQ ED NO:46 (SEQ ED NO:50), and an extracellular domain at amino acid residues 71 to 77 of SEQ ED NO:46.
In one embodiment of a nucleotide sequence of human TANGO 365, the nucleotide at position 14 is cytosine (C)(SEQ ID NO:45). In this embodiment, the amino acid at position 5 is alanine (A)(SEQ ID NO:46). In an alternative embodiment, a species variant of human TANGO 365 has a nucleotide at position 14 which is thymidine (T)(SEQ ID NO: 160). In this embodiment, the amino acid at position 5 is valine (V)(SEQ ID NO: 161), i.e., a conservative substitution. In one embodiment of a nucleotide sequence of human TANGO 365, the nucleotide at position 41 is guanine (G)(SEQ ID NO:45). In this embodiment, the amino acid at position 14 is arginine (R)(SEQ ID NO:46). En an alternative embodiment, a species variant of human TANGO 365 has a nucleotide at position 41 which is adenine (A)(SEQ ID NO: 162). In this embodiment, the amino acid at position 14 is histidine (H)(SEQ ID NO: 163), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 365, the nucleotide at position 59 is cytosine (C)(SEQ ID NO:45). In this embodiment, the amino acid at position 20 is threonine (T)(SEQ ID NO:46). En an alternative embodiment, a species variant of human TANGO 365 has a nucleotide at position 59 which is guanine (G)(SEQ ED NO: 164). In this embodiment, the amino acid at position 20 is serine (S)(SEQ ID NO: 165), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 365, the nucleotide at position 115 is adenine (A)(SEQ ID NO:45). In this embodiment, the amino acid at position 39 is asparagine (N)(SEQ ID NO:46). In an alternative embodiment, a species variant of human TANGO 365 has a nucleotide at position 115 which is guanine (G)(SEQ ID NO: 166). In this embodiment, the amino acid at position 39 is aspartate (D)(SEQ ID NO: 167), i.e., a conservative substitution.
One protein kinase C phosphorylation site is present in human TANGO 365. The site has the sequence SLR and is found (at amino acids 139 to 141 of SEQ ID NO:46). The TANGO 365 protein has four N-myristoylation sites. The first has the sequence GGTRCR and is found (at amino acids 18 to 23 of SEQ ID NO:46), the second has the sequence GTSMAC and is found (at amino acids 32 to 37 of SEQ ID NO:46), the third has the sequence GAACSL and is found (at amino acids 87 to 92 of SEQ ED NO:46), and the fourth has the sequence GSSDSS and is found (at amino acids 144 to 149 of SEQ ID NO:46). Human TANGO 365 also has an amidation site which has the sequence of LGRR (at amino acids 69 to 72 of SEQ ID NO:46).
Clone EpT365, which encodes human TANGO 365, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-291. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112. Uses of TANGO 365 Nucleic acids, Polypeptides. and Modulators Thereof
TANGO 365 was identified as being expressed in a prostate fibroblast library. In light of this, TANGO 365 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate processes involved in prostate development, differentiation and activity, including, but not limited to development, and differentiation and activation of prostate tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types. Such disorders can include, but are not limited to, malignant or benign prostate cell growth. Such disorders can include, but are not limited to, malignant or benign prostate cell growth. The TANGO 365 proteins can be used to treat subjects with or without prostate cancer e.g., prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato-rectal fistulas, atypical prostatic stromal lesions.
TANGO 365 nucleic acids, proteins, and modulators thereof can also be used to treat disorders of the cells and tissues in which it is expressed. As TANGO 365 is a transmembrane protein, proteins, nucleic acids and modulators thereof can be used to diagnose disorders and/or modulate intercellular signaling processes by disrupting or enhancing ligand-receptor or cell interaction with the extracellular matrix. Further, TANGO 365 could be used in detection and diagnostic assays to assay for normal or inappropriate expression of TANGO 365 proteins in abeπantly growing cells.
TANGO 365 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the prostate) and/or cells (e.g., fibroblasts) in which TANGO 365 is expressed. TANGO 365 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 368
A cDNA encoding human TANGO 368 was identified by analyzing the sequences of clones present in a natural killer cell library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthta080f06, encoding full-length human TANGO 368. The human TANGO 368 cDNA of this clone is 983 nucleotides long (Figure 11; SEQ ID NO:52). The open reading frame of this cDNA, nucleotides 152 to 328 of SEQ ID NO:52 (SEQ ID NO:53), encodes a 59 amino acid secreted protein (Figure 11; SEQ ID NO:54).
Figure 12 depicts a hydropathy plot of human TANGO 368. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and N-glycosylation sites (NGly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 368 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 27 of SEQ ED NO:54; SEQ ID NO:56) preceding the mature human TANGO 368 protein (coπesponding to amino acid 28 to amino acid 59 of SEQ ED NO:54; SEQ ED NO:55). The molecular weight of TANGO 368 protein without post-translational modifications is 6.5 kDa prior to the cleavage of the signal peptide and 3.5 kDa after cleavage of the signal peptide.
In one embodiment of a nucleotide sequence of human TANGO 368, the nucleotide at position 8 is cytosine (C)(SEQ ID NO:53). In this embodiment, the amino acid at position 3 is threonine (T)(SEQ ID NO:54). En an alternative embodiment, a species variant of human TANGO 368 has a nucleotide at position 8 which is guanine (G)(SEQ ED NO: 168). En this embodiment, the amino acid at position 3 is serine (S)(SEQ ID NO: 169), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 368, the nucleotide at position 10 is cytosine (C)(SEQ ID NO:53). In this embodiment, the amino acid at position 4 is glutamine (Q)(SEQ ID NO:54). In an alternative embodiment, a species variant of human TANGO 368 has a nucleotide at position 10 which is guanine (G)(SEQ ID NO: 170). In this embodiment, the amino acid at position 4 is glutamate (E)(SEQ ID NO: 171), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 368, the nucleotide at position 16 is cytosine (C)(SEQ ID NO:53). In this embodiment, the amino acid at position 6 is leucine (L)(SEQ ID NO:54). In an alternative embodiment, a species variant of human TANGO 368 has a nucleotide at position 16 which is guanine (G)(SEQ ID NO: 172). In this embodiment, the amino acid at position 6 is valine (V)(SEQ ID NO:173), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 368, the nucleotide at position 110 is adenine (A)(SEQ ID NO:53). In this embodiment, the amino acid at position 37 is histidine (H)(SEQ ID NO:54). In an alternative embodiment, a species variant of human TANGO 368 has a nucleotide at position 110 which is guanine (G)(SEQ ID NO: 174). In this embodiment, the amino acid at position 37 is arginine (R)(SEQ ID NO:175), i.e., a conservative substitution. Human TANGO 368 has an N-glycosylation site with the sequence NFTC (at amino acid residues 40 to 43 of SEQ ID NO: 54), a protein kinase C phosphorylation site with the sequence SLK (at amino acid residues 24 to 26 of SEQ ID NO:54), and a casein kinase II phosphorylation site with the sequence TQPE (at amino acid residues 27 to 30 of SEQ ID NO:54).
Figure 13 depicts a local alignment of the nucleotide sequence of full length human TANGO 368 (SEQ ID NO:52) and a fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57), which maps to a region of human chromosome 7. The full-length nucleic acid sequence of human TANGO 368 (SEQ ID NO:52) has 99.3% identity to a 973 bp fragment of the human T-cell receptor gamma VI gene region (Accession Number AF057177; SEQ ID NO:57). Northern blots were performed to analyze the expression of human TANGO 368 mRNA in human tissues. A weak signal was observed in the spleen and lymph node, however, no expression was detected in the thymus, peripheral blood leukocytes or fetal liver.
Clone EpT368, which encodes human TANGO 368, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-291. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 368 Nucleic acids. Polypeptides. and Modulators Thereof
As TANGO 368 was originally found in a natural killer cell library, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of immune cells, such as lymphocytes, e.g., natural killer cells, T-cells and B-cells. TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes e.g., the host immune response by, for example, modulating the formation of and/or binding to immune complexes, detection and defense against surface antigens and bacteria, and immune surveillance for rapid removal or pathogens. Such TANGO 368 nucleic acids, proteins and modulators thereof can be utilized, e.g., to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to viral or bacterial infection, autoimmune disorders (e.g., Grave's disease, Hashimoto's disease, and arthritis), immunodeficiency disorders (e.g., HIV, and inflammatory disorders (e.g., asthma, arthritis, psoriasis, septicemia, inflammatory bowel disease and allergies). As TANGO 368 exhibits expression in the spleen, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the spleen, e.g., cells of the splenic connective tissue, e.g., splenic smooth muscle cells and/or endothelial cells of the splenic blood vessels. TANGO 368 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., regenerated or phagocytized within the spleen, e.g., erythrocytes and/or B and T lymphocytes and macrophages. Thus, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to treat spleen, e.g., the fetal spleen, associated diseases and disorders. Examples of splenic diseases and disorders include e.g., splenic lymphoma and/or splenomegaly, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
As TANGO 368 exhibits expression in the lymph nodes, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the lymph node, e.g., cells of the lymph node connective tissue, e.g., lymph node smooth muscle cells and/or endothelial cells of the lymph node blood vessels. TANGO 368 nucleic acids, proteins, and modulators thereof can also be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., phagocytized within the lymph node, e.g., erythrocytes and/or B and T lymphocytes and macrophages. Thus, TANGO 368 nucleic acids, proteins, and modulators thereof can be used to treat lymph node associated diseases and disorders. Examples of lymph node diseases and disorders include e.g., lymphadenopathy, lymphoma, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream. In light of the fact that TANGO 368 is homologous to the T-cell receptor gamma
(TCRγ) locus, TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate the recognition of antigens in association with the major histocompatibility complex. TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate diseases and/or disorders associated with abeπant TCR-MHC interactions. Further, TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to modulate cell-cell receptor interactions.
As TANGO 368 exhibits homology to human T-cell receptor gamma VI gene region (Accession Numbers AF057177), which maps to a region of chromosome 7, TANGO 368 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders and/or modulate diseases associated with that region of chromosome 7, e.g., Stiff-Mann syndrome. As TANGO 368 is a secreted protein and thus likely a signaling molecule, TANGO 368 nucleic acids, proteins or modulators thereof, can be used to modulate TANGO 368 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. , in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
TANGO 368 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., natural killer cells) in which TANGO 368 is expressed. TANGO 368 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 369 A cDNA encoding human TANGO 369 was identified by analyzing the sequences of clones present in a natural killer cell library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthta088h08, encoding full-length human TANGO 369. The human TANGO 369 cDNA of this clone is 1119 nucleotides long (Figure 14; SEQ ID NO:58). The open reading frame of this cDNA, nucleotides 162 to 335 of SEQ ID NO:58 (SEQ ID NO:59), encodes a 58 amino acid secreted protein (Figure 14; SEQ ID NO:60).
Figure 15 depicts a hydropathy plot of human TANGO 369. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence on the left from the mature protein on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 369 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ED NO:60; SEQ ID NO:62) preceding the mature human TANGO 369 protein (coπesponding to amino acid 27 to amino acid 58 of SEQ ED NO:60; SEQ ED NO:61). The molecular weight of TANGO 369 protein without post-translational modifications is 6.8 kDa prior to the cleavage of the signal peptide and 3.7 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 17 and 25 of SEQ ID NO:60 indicates that there can be alternative forms of human TANGO 369 of 42 amino acids of SEQ ED NO:60, and 34 amino acids of SEQ ED NO:60, respectively. In one embodiment of a nucleotide sequence of human TANGO 369, the nucleotide at position 58 is cytosine (C)(SEQ ID NO:59). In this embodiment, the amino acid at position 20 is leucine (L)(SEQ ID NO:54). In an alternative embodiment, a species variant of human TANGO 369 has a nucleotide at position 58 which is guanine (G)(SEQ ID NO: 176). In this embodiment, the amino acid at position 20 is valine (V)(SEQ ID NO: 177), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 369, the nucleotide at position 68 is guanine (G)(SEQ ID NO:59). In this embodiment, the amino acid at position 23 is arginine (R)(SEQ ID NO:60). In an alternative embodiment, a species variant of human TANGO 369 has a nucleotide at position 68 which is adenine (A)(SEQ ID NO: 178). In this embodiment, the amino acid at position 23 is lysine (K)(SEQ ID NO: 179), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 369, the nucleotide at position 70 is thymine (T)(SEQ ID NO:59). In this embodiment, the amino acid at position 24 is leucine (L)(SEQ ID NO:60). In an alternative embodiment, a species variant of human TANGO 369 has a nucleotide at position 70 which is adenine (A)(SEQ ID NO: 180). In this embodiment, the amino acid at position 24 is threonine (T)(SEQ ID NO: 181), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 369, the nucleotide at position 120 is guanine (G)(SEQ ID NO:59). In this embodiment, the amino acid at position 40 is glutamate (E)(SEQ ID NO:60). In an alternative embodiment, a species variant of human TANGO 369 has a nucleotide at position 120 which is cytosine (C)(SEQ ID NO: 182). In this embodiment, the amino acid at position 40 is aspartate (D)(SEQ ID NO: 183), i.e., a conservative substitution. Northern blots were performed to analyze the expression of human TANGO 369 mRNA in human tissues. A very weak signal was observed in the spleen and lymph node, however, no expression was detected in the thymus, peripheral blood leukocytes or fetal liver.
Clone EpT369, which encodes human TANGO 369, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-295. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112. Uses of TANGO 369 Nucleic acids, Polypeptides. and Modulators Thereof
As TANGO 369 was originally found in a natural killer cell library, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, development, differentiation, and/or function of lymphocytes, e.g., natural killer cells. TANGO 369 nucleic acids, proteins and modulators thereof can be utilized to modulate immune-related processes, e.g., the host immune response by, for example, modulating the formation of and/or binding to immune complexes, detection and defense against surface antigens and bacteria, and immune surveillance for rapid removal or pathogens. Such TANGO 369 compositions and modulators thereof can be utilized, eg-, to ameliorate incidence of any symptoms associated with disorders that involve such immune-related processes, including, but not limited to viral or bacterial infection, autoimmune disorders (e.g., Grave's disease, Hashimoto's disease, arthritis, graft rejection), and inflammatory disorders (e.g., bacterial or viral infection, psoriasis, allergies and inflammatory bowel diseases). As TANGO 369 exhibits expression in the spleen, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the spleen, e.g., cells of the splenic connective tissue, e.g., splenic smooth muscle cells and/or endothelial cells of the splenic blood vessels. TANGO 369 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., regenerated or phagocytized within the spleen, e.g., erythrocytes and/or B and T lymphocytes and macrophages. Thus, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to treat spleen, e.g., the fetal spleen, associated diseases and disorders. Examples of splenic diseases and disorders include e.g., splenic lymphoma and/or splenomegaly, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
As TANGO 369 exhibits expression in the lymph nodes, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of cells that form the lymph node, e.g., cells of the lymph node connective tissue, e.g., lymph node smooth muscle cells and/or endothelial cells of the lymph node blood vessels. TANGO 369 nucleic acids, proteins, and modulators thereof can also be used to modulate the proliferation, differentiation, and/or function of cells that are processed, e.g., phagocytized within the lymph node, e.g., erythrocytes and/or B and T lymphocytes and macrophages. Thus, TANGO 369 nucleic acids, proteins, and modulators thereof can be used to treat lymph node associated diseases and disorders. Examples of lymph node diseases and disorders include e.g., lymphadenopathy, lymphoma, and/or phagocytotic disorders, e.g., those inhibiting macrophage engulfrnent of bacteria and viruses in the bloodstream.
TANGO 369 is associated with immune cells. As such, immune disorders associated TANGO 369 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate or treat immune disorders that include, but are not limited to, immune proliferative disorders (e.g., carcinoma, lymphoma, e.g., follicular lymphoma), and disorders associated with fighting pathogenic infections, e.g. , bacterial (e.g., chlamydia) infection, parasitic infection, and viral infection (e.g., HSV infection), and pathogenic disorders associated with immune disorders (e.g., immunodeficiency disorders, such as HIV).
Other immune disorders associated with TANGO 369 activity, for which TANGO 369 nucleic acids, proteins and modulators thereof can be used to modulate, identify, diagnose or treat, include, e.g., autoimmune disorders, such as arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders, such as bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis), apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF). Other TANGO 369 associated immune disorders include TNF related disorders
(e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer). TANGO 369 nucleic acids, proteins and modulators thereof can be used to treat such disorders. As TANGO 369 is a secreted protein, TANGO 369 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extracellular matrix.
As TANGO 369 is a secreted protein and thus likely a signaling molecule, TANGO 369 nucleic acids, proteins or modulators thereof can be used TANGO 369 biological activities, which can also include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades). TANGO 369 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the thymus) and/or cells (e.g., natural killer cells) in which TANGO 369 is expressed. TANGO 369 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 383
A cDNA encoding human TANGO 383 was identified by analyzing the sequences of clones present in a human prostate epithelium cDNA library. This analysis led to the identification of a clone, jthqb083bl0, encoding full-length TANGO 383. The human cDNA of this clone is 1386 nucleotides long (Figure 16; SEQ ID NO:63). The open reading frame of this cDNA, nucleotides 104 to 523 of SEQ ID NO:63 (SEQ ID NO:64), encodes a 140 amino acid TANGO 383 transmembrane protein (Figure 16; SEQ ID NO:65).
Figure 17 depicts a hydropathy plot of human TANGO 383. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 20 of SEQ ID NO:65; SEQ ID NO:66) on the left from the mature protein (amino acids 21 to 140 of SEQ ID NO:65; SEQ ID NO:67) on the right.
The signal peptide prediction program SIGNALP (Nielsen, et al. (1997) Protein Engineering 10:1-6) predicted that TANGO 383 includes a 20 amino acid signal peptide (amino acid 1 to amino acid 20 of SEQ ID NO:65; SEQ ED NO:66) preceding the mature protein (coπesponding to amino acid 21 to amino acid 140 of SEQ ID NO:65; SEQ ED NO:67). The molecular weight of TANGO 383 without post-translational modifications is 14.9 kDa prior to the cleavage of the signal peptide, 12.7 kDa after cleavage of the signal peptide.
TANGO 383 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain. The TANGO 383 protein contains an extracellular domain at amino acids 71 to 115 of SEQ ID NO:65 (SEQ ID NO:70), a first transmembrane domain at amino acid residues 50 to 70 of SEQ ID NO:65 (SEQ ED NO:68), a second transmembrane domain at amino acid residues 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69), a first cytoplasmic domain at amino acid residues 21 to 49 of SEQ ID NO:65 (SEQ ED NO:235) and a second cytoplasmic domain at amino acid residues 134 to 140 of SEQ ID NO:65 (SEQ ID NO: 136). Alternatively, in another embodiment, a TANGO 383 protein contains a cytoplasmic domain at amino acids 71 to 115 of SEQ ID NO: 65, a first transmembrane domain at amino acid residues 50 to 70 of SEQ ID NO:65 (SEQ ID NO:68), a second transmembrane domain at amino acid residues 116 to 133 of SEQ ID NO:65 (SEQ ID NO:69), a first extracellular domain at amino acid residues 21 to 49 of SEQ ID NO:65 and a second extracellular domain at amino acid residues 134 to 140 of SEQ ID NO:65.
En one embodiment of a nucleotide sequence of human TANGO 383, the nucleotide at position 4 is cytosine (C)(SEQ ID NO:64). In this embodiment, the amino acid at position 2 is leucine (L)(SEQ ID NO:65). In an alternative embodiment, a species variant of human TANGO 383 has a nucleotide at position 4 which is adenine (A)(SEQ ID NO: 184). In this embodiment, the amino acid at position 2 is isoleucine (I)(SEQ ED NO: 185), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 383, the nucleotide at position 8 is guanine (G)(SEQ ID NO:64). In this embodiment, the amino acid at position 3 is serine (S)(SEQ ID NO:65). In an alternative embodiment, a species variant of human TANGO 383 has a nucleotide at position 8 which is cytosine (C)(SEQ ID NO: 186). In this embodiment, the amino acid at position 3 is threonine (T)(SEQ ID NO: 187), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 383, the nucleotide at position 17 is adenine (A)(SEQ ID NO:64). In this embodiment, the amino acid at position 6 is lysine (K)(SEQ ID NO:65). In an alternative embodiment, a species variant of human TANGO 383 has a nucleotide at position 17 which is guanine (G)(SEQ ID NO: 188). In this embodiment, the amino acid at position 6 is arginine (R)(SEQ ID NO: 189), i.e., a conservative substitution. In one embodiment of a nucleotide sequence of human TANGO 383, the nucleotide at position 57 is cytosine (C)(SEQ ID NO:64). In this embodiment, the amino acid at position 19 is aspartate (D)(SEQ ID NO:65). In an alternative embodiment, a species variant of human TANGO 383 has a nucleotide at position 57 which is guanine (G)(SEQ ID NO: 190). In this embodiment, the amino acid at position 19 is glutamate (E)(SEQ ID NO: 191), i.e., a conservative substitution.
One protein kinase C phosphorylation site is present in TANGO 383, and has the sequence SPR (at amino acids 21 to 24 of SEQ ID NO:65). TANGO 383 has one casein kinase II phosphorylation site which has the sequence SKAE (at amino acids 42 to 45 of SEQ ID NO:65). TANGO 383 has three N-myristylation sites. The first has the sequence GVELAS (at amino acids 24 to 29 of SEQ ID NO:65), the second has the sequence GAVLAH (at amino acids 84 to 89 of SEQ ED NO:65), and the third has the sequence GSSDSH (at amino acids 96 to 101 of SEQ ID NO:65). TANGO 383 has a consensus tyrosine phosphorylation site which has the amino acid sequence RGKREAGLY and (at amino acids 33 to 41 of SEQ ID NO:65). TANGO 383 also has an amidation site with the sequence RGKR (at amino acids 33-36 of SEQ ID NO:65). Figure 18 depicts an alignment of the amino acid sequence of TANGO 383 (SEQ
ID NO:65) and the amino acid sequence of Neuronal Thread Protein AD7C-NTP (SEQ ID NO:72). The alignments demonstrates that the amino acid sequences of TANGO 383 and Neuronal Thread Protein AD7C-NTP are 52% identical. This alignment was performed using the ProDom NCBI-BLASTP2 program with graphical output using the following settings: Matrix: BLOSUM62; Expect: 0.1; Filter: none.
Thus, TANGO 383 exhibits homology to neural thread proteins which are phospho-proteins expressed in the central nervous system which are phosphorylated during neuritic sprouting. Therefore, TANGO 383 nucleic acids, proteins and modulators thereof may be used to diagnose disorders and/or inhibit or modulate neurodegenerative sprouting and synaptic disassociation associated with, e.g., Alzheimer's disease, and other diseases in neural tissue as discussed below.
Clone EpT383, which encodes human TANGO 383, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-295. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 383 Nucleic acids. Polypeptides. and Modulators Thereof
As TANGO 383 was originally found in a prostate epithelium library, TANGO 383 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of prostate cells. TANGO 383 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in prostate development, differentiation and activity, including, but not limited to development, and differentiation and activation of prostate tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types or disorders associated with such cell types. Such disorders can include, but are not limited to, malignant or benign prostate cell growth or inflammatory disorders (e.g. , prostatitis, benign prostatic hypertrophy, benign prostatic hyperplasia (BPH), prostatic paraganglioma, prostate adenocarcinoma, prostatic intraepithelial neoplasia, prostato-rectal fistulas, atypical prostatic stromal lesions).
TANGO 383 exhibits homology to neural thread proteins which are phospho- proteins expressed in the central nervous system which are phosphorylated during neuritic sprouting. Therefore, TANGO 383 nucleic acids, proteins and modulators thereof may be used to diagnose disorders and/or inhibit or modulate neurodegenerative sprouting and synaptic disassociation associated with, e.g., Alzheimer's disease. TANGO 383 nucleic acids, proteins and modulators thereof may also be utilized to diminish the effects of stroke and other neural damage, e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra- nuclear palsy, and other dementias.
As TANGO 383 is a transmembrane protein, TANGO 383 nucleic acids, proteins and modulators thereof can be utilized to modulate intercellular signaling pathways, for example by disrupting ligand-receptor interactions or cellular interactions with the extracellular matrix.
As TANGO 383 is a transmembrane protein and thus likely a signaling molecule, TANGO 383 nucleic acids, proteins or modulators thereof, activities can include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades). TANGO 383 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the prostate) and/or cells (e.g., epithelial cells) in which TANGO 383 is expressed. TANGO 383 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 393
A cDNA encoding human TANGO 393 was identified by analyzing the sequences of clones present in a human fetal hypothalamus cDNA library for sequences containing signal peptides. This analysis led to the identification of a clone, jthhb039f09, encoding full-length human TANGO 393. The human cDNA of this clone is 1778 nucleotides long (Figure 19; SEQ ID NO:73). The open reading frame of this cDNA, nucleotides 40 to 1458 of SEQ ED NO:75 (SEQ ED NO:74), encodes a 473 amino acid human TANGO 393 transmembrane protein (Figure 19; SEQ ID NO:75).
Figure 20 depicts a hydropathy plot of human TANGO 393. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues
(cys) and potential N-glycosylation sites are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:75; SEQ ED NO:76) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:75; SEQ ID NO:77) on the right. The signal peptide prediction program SIGNALP (Nielsen, et al. (1997) Protein
Engineering 10:1-6) predicted that human TANGO 393 includes an 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:75; SEQ ID NO:76) preceding the mature protein (coπesponding to amino acid 27 to amino acid 473 of SEQ ED NO:75; SEQ ID NO:77). The molecular weight of human TANGO 393 without post-translational modifications is 50.7 kDa prior to the cleavage of the signal peptide, 47.8 kDa after cleavage of the signal peptide. The presence of a methionine residue at position 229 of SEQ ID NO: 75 indicates that there can be alternative forms of human TANGO 393 of 245 amino acids of SEQ ID NO:75.
Human TANGO 393 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; and (3) a cytoplasmic domain; and (4) a leucine-rich domain. The human TANGO 393 protein contains an extracellular domain at amino acids 27 to 447 of SEQ ID NO:75 (SEQ ID NO:89), a transmembrane domain at amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a cytoplasmic domain at amino acid residues 468 to 473 of SEQ ED NO:75 (SEQ ED NO: 134).
Alternatively, in another embodiment, a human TANGO 393 protein contains a cytoplasmic domain at amino acids 27 to 447 of SEQ ID NO:75, a transmembrane domain at amino acid residues 448 to 467 of SEQ ID NO:75 (SEQ ID NO:78), and a extracellular domain at amino acid residues 468 to 473 of SEQ ID NO:75. In one embodiment of a nucleotide sequence of human TANGO 393, the nucleotide at position 5 is adenine (A)(SEQ ID NO:74). In this embodiment, the amino acid at position 2 is lysine (K)(SEQ ID NO:75). In an alternative embodiment, a species variant of human TANGO 393 has a nucleotide at position 5 which is guanine (G)(SEQ ID NO: 192). In this embodiment, the amino acid at position 2 is arginine (R)(SEQ ID NO: 193), i. e. , a conservative substitution. In one embodiment of a nucleotide sequence of human TANGO 393, the nucleotide at position 17 is cytosine (C)(SEQ ID NO:74). In this embodiment, the amino acid at position 6 is alanine (A)(SEQ ID NO:75). In an alternative embodiment, a species variant of human TANGO 393 has a nucleotide at position 17 which is thymidine (T)(SEQ ED NO: 194). In this embodiment, the amino acid at position 6 is valine (V)(SEQ ED NO: 195), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 393, the nucleotide at position 55 is cytosine (C)(SEQ ID NO:74). In this embodiment, the amino acid at position 19 is glutamine (Q)(SEQ ID NO:75). In an alternative embodiment, a species variant of human TANGO 393 has a nucleotide at position 55 which is guanine (G)(SEQ ID NO: 196). In this embodiment, the amino acid at position 19 is glutamate (E)(SEQ ID NO: 197), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 393, the nucleotide at position 118 is adenine (A)(SEQ ID NO:74). In this embodiment, the amino acid at position 40 is threonine (T)(SEQ ID NO:75). In an alternative embodiment, a species variant of human TANGO 393 has a nucleotide at position 118 which is thymine (T)(SEQ ID NO: 198). In this embodiment, the amino acid at position 40 is serine (S)(SEQ ID NO: 199), i.e., a conservative substitution.
Human TANGO 393 has LRR from amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and 260 to 310 of SEQ ID NO:75 (SEQ ID NO:79, 80, 81, 82, 83, 84, 85, 86, 87 and 88, respectively). These repeats are spaced in beta-alpha folds in the structure of the protein, so as to create a hydrophobic face that induces particular folding of the protein.
Human TANGO 393 has five N-glycosylation sites. The first has a sequence of NLTI (at amino acids 82-85 of SEQ ID NO:75), the second has a sequence of NLTH (at amino acids 179 to 182 of SEQ ID NO:75), the third has a sequence of NLSA (at amino acids 237 to 240 of SEQ ID NO:75), the fourth has a sequence of NGSG (at amino acids 372 to 375 of SEQ ID NO:75), and the fifth has a sequence of NRTR (at amino acids 423 to 426 of SEQ ID NO:75). Human TANGO 393 has one Glycosaminoglycan attachment site, the sequence of which is SGGG (at amino acids 436 to 439 of SEQ ID NO:75).
Human TANGO 393 has one cAMP and cGMP-dependent protein kinase phosphorylation site, the sequence of which is KRAS (at amino acids 2 to 5 of SEQ ID NO:75). Human TANGO 393 has five protein kinase C phosphorylation sites, where the first has a sequence SQR of (at amino acids 59 to 61 of SEQ ID NO:75), the second has a sequence SFR of (at amino acids 76 to 78 of SEQ ID NO:75), the third has a sequence TFR of (at amino acids 173 to 175 of SEQ ID NO:75), the fourth has a sequence TGR of (at amino acids 321 to 323 of SEQ ID NO:75), and the fifth has a sequence SRK of (at amino acids 420 to 422 of SEQ ID NO:75). Human TANGO 393 has five casein kinase II phosphorylation sites, where the first has a sequence of TFRD (at amino acids 173 to 176 of SEQ ID NO:75), the second has a sequence of SVPE (at amino acids 192 to 195 of SEQ ID NO:75), the third has a sequence of SSSE (at amino acids 281 to 284 of SEQ ID NO:75), the fourth has a sequence of TDEE (at amino acids 325 to 328 of SEQ ID NO: 75), and the fifth has a sequence of SVLE (at amino acids 345 to 348 of SEQ ED NO: 75). Human TANGO 393 has eleven N-myristylation sites, where the first has the sequence GACVCY (at amino acids 29 to 34 of SEQ ID NO:75), the second has the sequence GFPAAS (at amino acids 54 to 59 of SEQ ED NO:75), and the third has the sequence GNRESH (at amino acids 66 to 71 of SEQ ED NO:75), the fourth has the sequence GLFRGL (at amino acids 148 to 153 of SEQ ED NO:75), and the fifth has the sequence GNRISS (at amino acids 187 to 192 of SEQ ED NO:75), the sixth has the sequence GCAVAT (at amino acids 308 to 313 of SEQ ED NO:75), and the seventh has the sequence GLPKCC (at amino acids 331 to 336 of SEQ ED NO:75), the eighth has the sequence GTLPGS (at amino acids 385 to 390 of SEQ ED NO:75), and the ninth has the sequence GQAGSG (at amino acids 432 to 437 of SEQ ED NO:75), the tenth has the sequence GGGTGD (at amino acids 438 to 443 of SEQ ED NO:75), and the eleventh has the sequence GALPSL (at amino acids 448 to 453 of SEQ ED NO:75). Human TANGO 393 has a Leucine zipper pattern which has the amino acid sequence
LHLDRCGLQELGPGLFRGLAAL (at amino acids 135 to 156 of SEQ ID NO:75).
Human TANGO 393 maps by homology to ESTs to Chromosome 22 between D22S420 and D22S446.
Clone EpT393, which encodes human TANGO 393, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-295. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. § 112.
Mouse TANGO 393
A cDNA encoding mouse TANGO 393 was identified in an analysis of a fetal hypothalamus library for screening encoding signal peptides. This analysis led to the identification of a clone, jtmoa038d08, encoding full-length mouse TANGO 393. The mouse cDNA of this clone is 1946 nucleotides long (Figure 21; SEQ ID NO:93). The open reading frame is from nucleotides 226 to 1644 of SEQ ED NO:93 (SEQ ED NO:94), encodes a 473 amino acid mouse TANGO 393 transmembrane protein (Figure 21; SEQ ED NO:95).
Figure 22 depicts a hydropathy plot of mouse TANGO 393. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and potential N-glycosylation sites are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 26 of SEQ ID NO:95; SEQ ID NO:96) on the left from the mature protein (amino acids 27 to 473 of SEQ ID NO:95; SEQ ID NO:97) on the right.
The signal peptide prediction program SIGNALP (Nielsen, et al. (1997) Protein Engineering 10:1-6) predicted that mouse TANGO 393 includes an 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:95; SEQ ID NO:96) preceding the mature protein (coπesponding to amino acid 27 to amino acid 473 of SEQ ID NO:95; SEQ ID NO:97). The molecular weight of mouse TANGO 393 without post-translational modifications is 51.0 kDa prior to the cleavage of the signal peptide, 48.1 kDa after cleavage of the signal peptide. The presence of a methionine residue at positions 229, 240 and 247 of SEQ ID NO: 95 indicates that there can be alternative forms of mouse TANGO 393 of 245 amino acids of SEQ ID NO:95, 234 amino acids of SEQ ID NO:95, and 227 amino acids of SEQ ID NO:95, respectively.
Mouse TANGO 393 is a transmembrane protein which contains one or more of the following domains: (1) an extracellular domain; (2) a transmembrane domain; (3) a cytoplasmic domain; and (4) leucine-rich repeat domain. The mouse TANGO 393 protein contains an extracellular domain at amino acids 27 to 449 of SEQ ID NO:95 (SEQ ID NO: 109), a transmembrane domain at amino acid residues 450 to 467 of SEQ ID NO:75 (SEQ ID NO:98), and a cytoplasmic domain at amino acid residues 468 to 473 of SEQ ED NO:95 (SEQ ID NO:135).
Alternatively, in another embodiment, a mouse TANGO 393 protein contains a cytoplasmic domain at amino acids 27 to 449 of SEQ ID NO:95, a transmembrane domain at amino acid residues 450 to 467 of SEQ ID NO:75 (SEQ ED NO:98), and an extracellular domain at amino acid residues 468 to 473 of SEQ ED NO:95.
In one embodiment of a nucleotide sequence of mouse TANGO 393, the nucleotide at position 5 is adenine (A)(SEQ ID NO:94). In this embodiment, the amino acid at position 2 is lysine (K)(SEQ ID NO:95). En an alternative embodiment, a species variant of mouse TANGO 393 has a nucleotide at position 5 which is guanine (G)(SEQ ED NO:200). In this embodiment, the amino acid at position 2 is arginine (R)(SEQ ID NO:201), i.e., a conservative substitution.
En one embodiment of a nucleotide sequence of mouse TANGO 393, the nucleotide at position 59 is cytosine (C)(SEQ ID NO:94). In this embodiment, the amino acid at position 20 is alanine (A)(SEQ ID NO:95). In an alternative embodiment, a species variant of mouse TANGO 393 has a nucleotide at position 59 which is thymidine (T)(SEQ ID NO:202). In this embodiment, the amino acid at position 20 is valine (V)(SEQ ID NO:203), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of mouse TANGO 393, the nucleotide at position 118 is adenine (A)(SEQ ID NO:94). In this embodiment, the amino acid at position 40 is threonine (T)(SEQ ID NO:95). In an alternative embodiment, a species variant of mouse TANGO 393 has a nucleotide at position 118 which is thymidine (T)(SEQ ID NO:204). In this embodiment, the amino acid at position 40 is serine (S)(SEQ ID NO:205), i.e., a conservative substitution. In one embodiment of a nucleotide sequence of mouse TANGO 393, the nucleotide at position 178 is cytosine (C)(SEQ ID NO:94). In this embodiment, the amino acid at position 60 is glutamine (Q)(SEQ ID NO:95). In an alternative embodiment, a species variant of mouse TANGO 393 has a nucleotide at position 178 which is guanine (G)(SEQ ID NO:206). In this embodiment, the amino acid at position 60 is glutamate (E)(SEQ ED NO:207), i.e., a conservative substitution.
Mouse TANGO 393 has five N-glycosylation sites. The first has a sequence of NLTI (at amino acids 82-85 of SEQ ID NO:95), the second has a sequence of NLTH (at amino acids 179 to 182 of SEQ ID NO:95), the third has a sequence of NLSM (at amino acids 237 to 240 of SEQ ID NO:95), the fourth has a sequence of NGSG (at amino acids 372 to 375), and the fifth has a sequence of NRTR (at amino acids 423 to 426 of SEQ ID NO:95). Mouse TANGO 393 has one Glycosaminoglycan attachment site, the sequence of which is SGTG (at amino acids 439 to 442 of SEQ ID NO:95). Mouse TANGO 393 has one cAMP- and cGMP-dependent protein kinase phosphorylation site, the sequence of which is KRAS (at amino acids 2 to 5 of SEQ ID NO:95). Mouse TANGO 393 has four protein kinase C phosphorylation sites, where the first has a sequence SQR of (at amino acids 59 to 61 of SEQ ID NO:95), the second has a sequence SCR of (at amino acids 79 to 81 of SEQ ID NO:95), the third has a sequence TFR of (at amino acids 173 to 175 of SEQ ID NO:95), and the fourth has a sequence SRK of (at amino acids 420 to 422 of SEQ ID NO:95). Mouse TANGO 393 has eight casein kinase II phosphorylation sites, where the first has a sequence of TLLE (at amino acids 105 to 108 of SEQ ID NO:95), the second has a sequence of TFRD (at amino acids 173 to 176 of SEQ ID NO:95), the third has a sequence of SVPE (at amino acids 192 to 195 of SEQ ID NO:95), the fourth has a sequence of SSSE (at amino acids 281 to 284 of SEQ ID NO:95), the fifth has a sequence of SDLE (at amino acids 304 to 307 of SEQ ID NO:95), the sixth has a sequence of TDEE (at amino acids 325 to 328 of SEQ ID NO:95), the seventh has a sequence of SVLE (at amino acids 345 to 348 of SEQ ID NO:95), and the eighth has a sequence of SS AE (at amino acids 389 to 392 of SEQ ID NO:95). Mouse TANGO 393 has ten N-myristylation sites, where the first has the sequence GACVCY (at amino acids 29 to 34 of SEQ ID NO:95), the second has the sequence GIPAAS (at amino acids 54 to 59 of SEQ ID NO:95), and the third has the sequence GN ISH (at amino acids 66 to 71 of SEQ ED NO:95), the fourth has the sequence GLFRGL (at amino acids 148 to 153 of SEQ ID NO:95), and the fifth has the sequence GCAVAS (at amino acids 308 to 313 of SEQ ED NO:95), the sixth has the sequence GTLPSS (at amino acids 385 to 390 of SEQ ED NO:95), and the seventh has the sequence GLPTTG (at amino acids 407 to 412 of SEQ ID NO:95), the eighth has the sequence GQAGSG (at amino acids 432 to 437 of SEQ ED NO:95), and the ninth has the sequence GTGDAE (at amino acids 440 to 445 of SEQ ED NO:95), and the tenth has the sequence GALPAL (at amino acids 448 to 453 of SEQ ID NO:95). Mouse TANGO 393 has a prokaryotic membrane lipoprotein lipid attachment site with the sequence of SHVPAASFQSC (at amino acids 70 to 80 of SEQ ID NO:95). Mouse TANGO 393 has a Leucine zipper pattern which has the amino acid sequence LHLDRCGLRELGPGLFRGLAAL (at amino acids 135 to 156 of SEQ ID NO:95).
Mouse TANGO 393 has LRR from amino acids 26 to 57, 58 to 81, 82 to 105, 106 to 130, 131 to 154, 155 to 178, 179 to 202, 203 to 226, 227 to 250, and 260 to 310 of SEQ ID NO:95 (SEQ ID NO:99, 100, 101, 102, 103, 104, 105, 106, 107 and 108, respectively). These repeats are spaced in beta-alpha folds in the structure of the protein, so as to create a hydrophobic face that induces particular folding of the protein.
Figure 23 depicts an alignment of the open reading frames of human TANGO 393 (SEQ ID NO:74) and mouse TANGO 393 (SEQ ID NO:94) demonstrating an identity of 82.8%. The algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989) Figure 24 depicts an alignment of the immature proteins of human TANGO 393
(SEQ ID NO:75) and mouse TANGO 393 (SEQ ID NO:95) demonstrating an identity of 89.2%. The algorithm used to align the sequences was the ALIGN program which calculates a global alignment of two sequences. (Version 2.0u, Myers and Miller, 1989)
Uses of TANGO 393 Nucleic acids. Polypeptides. and Modulators Thereof As both mouse and human TANGO 393 clones were originally identified in a fetal hypothalamus library, TANGO 393 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders and/or modulate the proliferation, differentiation, and/or function of endocrine cells, in particular hypothalamus, cells. TANGO 393 nucleic acids, proteins and modulators thereof can be utilized to modulate processes involved in hypothalamus development, differentiation and activity, including, but not limited to development, and differentiation and activation of hypothalamus tissues and cells as well as any function associated with such cells, and amelioration of one or more symptoms associated with abnormal function of such cell types. Such disorders can include, but are not limited to, malignant or benign hypothalamus cell growth.
Furthermore, as the hypothalamus is the master regulator of the entire endocrine system, as such, TANGO 393 nucleic acids, proteins and modulators thereof can be used as a therapeutic agent to treat mammals with abnormal hypothalamic function wherein the mammal exhibits abnormal whole animal homeostasis, appetite-related disorders, obesity, cachexia, food intake disorders, stress responsiveness disorders, adrenal function disorders, pituitary disorders and adrenal disorders. Further, TANGO 393 proteins, nucleic acids, or modulators thereof, can be used to treat disorders of the adrenal cortex, such as hypoadrenalism (e.g. , primary chronic or acute adrenocortical insufficiency, and secondary adrenocortical insufficiency), hyperadrenalism (Cushing's syndrome, primary hyper-aldosteronism, adrenal virilism, and adrenal hypeφlasia), or neoplasia (e.g., adrenal adenoma and cortical carcinoma). In another example, TANGO 393 polypeptides, nucleic acids, or modulators thereof, can be used to treat disorders of the thyroid gland, which is partially regulated by the hypothalamus, such as hyperthyroidism (e.g., diffuse toxic hypeφlasia, toxic multinodular goiter, toxic adenoma, and acute or subacute thyroiditis), hypothyroidism (e.g., cretinism and myxedema), thyroiditis (e.g., Hashimoto's thyroiditis, subacute granulomatous thyroiditis, subacute lymphocytic thyroiditis, Riedel's thryroiditis), Graves' disease, goiter (e.g., simple diffuse goiter and multinodular goiter), or tumors (e.g., adenoma, papillary carcinoma, follicular carcinoma, medullary carcinoma, undifferentiated malignant carcinoma, Hodgkin's disease, and non-Hodgkin's lymphoma). TANGO 393 exhibits homology to genes which contain sequences refeπed to as
Leucine Rich Repeats (LRR), for example, SLIT-1, leucine-rich α-2-Glycoprotein and Platelet Glycoprotein V precursor. As such, TANGO 393 nucleic acids, proteins and modulators thereof can be used to treat subjects with defects in leucine-rich-repeat genes shown to cause various diseases, including but not limited to Bernard-Soulier disease, a bleeding disorder. Further, as TANGO 393 has homology to Platelet Glycoprotein V
(GPV) precursor, TANGO 393 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate platelet activity, thrombin activity, von Willebrand Factor assembly and activation, or ADP/epinephrine-, cathepsin G-, and TRAP-induced decrease in platelet surface GPV expression.
Furthermore, TANGO 393 proteins, nucleic acids and modulators thereof can be used to modulate the pathogenesis of infectious diseases, for example, diseases that are affected by the expression of leucine-rich-repeat proteins such as the type-1 human immunodeficiency virus (HIV-1) Rev protein, which is the trans-activating region of the virus (Kobe and Deisenhofer, 1994, TEBS, 19:415-421).
LRR containing proteins are tissue organizers, wherein they orient and order collagen fibrils during ontogeny and are involved in pathological processes such as wound healing, tissue repair, and tumor stroma formation. These properties are rooted in their bifunctional character: the protein moiety binding collagen fibrils at strategic loci, the microscopic gaps between staggered fibrils, and the highly charged glycosaminoglycans extending out to regulate interfibrillar distances and thereby establishing the exact topology of fibrillar collagens in tissues. Therefore, TANGO 393 nucleic acids, proteins and modulators thereof can be used to disrupt intercellular and intracellular protein interactions or cellular signaling in tissues or cells, for example in the hypothalamus. More particularly, the TANGO 393 nucleic acids, proteins and modulators thereof can be used to modulate wound healing (e.g., platelet activation), tissue repair and tumor stroma formation as well. Furthermore, TANGO 393 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate the function of the hypothalamus as it relates to control of endocrine function, regulation of whole animal homeostasis and modulation of diurnal requirements, appetite as related to obesity or cachexia, and generally weight control in mammals. Proteins with LRR also interact with soluble growth factors, modulate their functional activity, and bind to cell surface receptors. The latter interaction affects cell cycle progression in a variety of cellular systems and could explain changes in the expression of these gene products around the invasive neoplastic cells and in regenerating tissues. See Generally, Iozzo,1997, Crit. Rev. Biochem. Mol. Biol., 32(2):l4l-14. As such, TANGO 393 nucleic acids, proteins and modulators thereof can be used to modulate disorders associated with abeπant expression of TANGO 393 in cancerous (e.g., tumor) cells that do not normally express TANGO 393. Such disorders can include, for example, ones associated with tumor cell migration and progression to metastasis.
As TANGO 393 exhibits homology to the SLIT-1 proteins, TANGO 393 proteins, nucleic acids and modulators thereof may participate in the formation and maintenance of the nervous and endocrine systems by e.g., protein-protein interactions. Northern blot analysis has revealed that the human SLIT-1, -2, and -3 mRNAs are exclusively expressed in the brain, spinal cord, and thyroid, respectively. In situ hybridization studies indicated that the rat SLIT-1 mRNA is specifically expressed in the neurons of fetal and adult forebrains (Itoh et al., Brain Res Mol Brain Res 1998 Nov 20;62(2): 175-86.) This suggests a role for TANGO 393 nucleic acids, proteins and modulators thereof in brain development and neural function. Therefore, the TANGO 393 nucleic acids, proteins and modulators thereof may be useful to disrupt protein interaction or cellular signaling in brain tissues or cells. En particular, TANGO 393 protein, nucleic acids and modulators thereof could be useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., multiple sclerosis, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementia. TANGO 393 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the hypothalamus) and/or cells (e.g., hypothalamic cells) in which TANGO 393 is expressed. TANGO 393 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human TANGO 402
A cDNA encoding human TANGO 402 was identified by analyzing the sequences of clones present in a human 9 week fetus library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jthga055h07, encoding full-length human TANGO 402. The human TANGO 402 cDNA of this clone is 1348 nucleotides long (Figure 25; SEQ ID NO:l 10). The open reading frame of this cDNA, nucleotides 87 to 707 of SEQ ID NO:110 (SEQ ED NO:l l l), encodes a 207 amino acid transmembrane protein (Figure 25; SEQ ID NO: 112).
Figure 26 depicts a hydropathy plot of human TANGO 402. Relatively hydrophobic regions of the protein are above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) and N- glycosylation sites are (Ngly) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence from the mature protein described below.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human TANGO 402 includes a 50 amino acid signal peptide (amino acid 1 to amino acid 50 of SEQ ID NO:112; SEQ ID NO:114) preceding the mature human TANGO 402 protein (coπesponding to amino acid 51 to amino acid 207 of SEQ ID NO:112; SEQ ED NO:113). The molecular weight of human TANGO 402 protein without post-translational modifications is 24.0 kDa prior to the cleavage of the signal peptide, 18.1 kDa after cleavage of the signal peptide. Human TANGO 402 protein is a transmembrane protein that contains an extracellular domain at amino acids 1 to 133 of SEQ ID NO:l 12 or a mature extracellular domain at amino acid residues 51 to 133 of SEQ ID NO:l 12 (SEQ ID NO:l 15), a transmembrane domain at amino acid residues 134 to 151 of SEQ ED NO: 112 (SEQ ID NO:l 16), and a cytoplasmic domain at amino acid residues 152 to 207 of SEQ ID NO:l 12 (SEQ ID NO: 117).
Alternatively, in another embodiment, a human TANGO 402 protein contains a cytoplasmic domain at amino acids 1 to 133 of SEQ ID NO:l 12 or a mature cytoplasmic domain at amino acid residues 51 to 133 of SEQ ID NO:l 12, a transmembrane domain at amino acid residues 134 to 151 of SEQ ID NO:112 (SEQ ID NO: 116), and an extracellular domain at amino acid residues 152 to 207 of SEQ ED NO: 112.
In one embodiment of a nucleotide sequence of human TANGO 402, the nucleotide at position 28 is cytosine (C)(SEQ ID NO: 111). In this embodiment, the amino acid at position 10 is leucine (L)(SEQ ID NO: 112). In an alternative embodiment, a species variant of human TANGO 402 has a nucleotide at position 28 which is guanine (G)(SEQ ID NO:208). In this embodiment, the amino acid at position 10 is valine (V)(SEQ ID NO:209), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 402, the nucleotide at position 58 is cytosine (C)(SEQ ID NO:l 11). In this embodiment, the amino acid at position 20 is glutamine (A)(SEQ ID NO:l 12). In an alternative embodiment, a species variant of human TANGO 402 has a nucleotide at position 58 which is guanine (G)(SEQ ID NO:210). In this embodiment, the amino acid at position 20 is glutamate (E)(SEQ ID NO:211), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 402, the nucleotide at position 61 is adenine (A)(SEQ ID NO:l 11). In this embodiment, the amino acid at position 21 is lysine (K)(SEQ ID NO:l 12). In an alternative embodiment, a species variant of human TANGO 402 has a nucleotide at position 61 which is guanine (G)(SEQ ED NO:212). In this embodiment, the amino acid at position 21 is arginine (R)(SEQ ID NO:213), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human TANGO 402, the nucleotide at position 64 is thymine (T)(SEQ ID NO: 111). In this embodiment, the amino acid at position 22 is serine (S)(SEQ ID NO:l 12). In an alternative embodiment, a species variant of human TANGO 402 has a nucleotide at position 64 which is adenine (A)(SEQ ID NO:214). In this embodiment, the amino acid at position 22 is threonine (T)(SEQ ID NO:215), i.e., a conservative substitution.
Two N-glycosylation sites are present in human TANGO 402. The first has the sequence NISS (at amino acid residues 67 to 70 of SEQ ID NO: 112) and the second has the sequence NGTS (at amino acid residues 202 to 205 of SEQ ED NO: 112). Six protein kinase C phosphorylation sites are present in human TANGO 402. The first has the sequence SFK (at amino acid residues 11 to 13 of SEQ ID NO: 112), the second has the sequence SFK (at amino acid residues 95 to 97 of SEQ ED NO: 112), the third has the sequence TWK (at amino acid residues 98 to 100 of SEQ ED NO: 112), the fourth has the sequence SQR (at amino acid residues 102 to 104 of SEQ ID NO: 112), the fifth has the sequence SLK (at amino acid residues 128 to 130 of SEQ ID NO:l 12), and the sixth has the sequence TFK (at amino acid residues 188 to 190 of SEQ ID NO:l 12). Three casein kinase II phosphorylation sites are present in human TANGO 402. The first has the sequence TGID (at amino acid residues 49 to 52 of SEQ ID NO: 112), the second has the sequence TWKE (at amino acid residues 98 to 101 of SEQ ID NO:l 12), and the third has the sequence SQRD (at amino acid residues 102 to 105 of SEQ ID NO: 112). Human TANGO 402 has a tyrosine kinase phosphorylation site having the sequence KSKDFSLY at amino acid residues 21 to 28 of SEQ ID NO:l 12). Human TANGO 402 has an N- myristylation site having the sequence GLYVTF at amino acid residues 138 to 143 of SEQ ID NO: 112.
Human TANGO 402 includes a C-type lectin (CTL)-like domain at amino acid residues 104 to 193 of SEQ ID NO:112 (SEQ ID NO: 118). CTL domains have been shown to function as a calcium-dependent carbohydrate-recognition domain. Human TANGO 402 is homologous to human lectin-like oxidized LDL receptor 1
(LOX-1), which is the receptor for oxidized lipoprotein (Sawamura et al., 1997, Science, 386:73-77). LOX-1 is involved in oxidized low-density lipoprotein (Ox-LDL) uptake and subsequent foam cell transformation in macrophages and smooth muscle cells in the atherosclerotic intima (Kume et al., 1998, Cir. Res., 83:322-327; Yamada, et al., 1998, Cell. Mol. Life Sci., 54(D:628-640: Moriwaki et al., 1998, Artherioscler. Thromb. Vαsc. Biol, 1800): 1541-1547; Napase et al., 1998, J. Biol. Chem., 273{50):33702-33707). Figure 27 shows an alignment of the human TANGO 402 amino acid sequence (SEQ ID NO:l 12) with the human LOX-1 amino acid sequence (SEQ ID NO:67; Accession Number ABO 10710). As shown in the figure, the amino acid sequence of LOX-1 is 25.1% identical to the amino acid sequence of human TANGO 402 (SEQ ID NO:l 12). As shown in Figure 28. The coding regions of the human TANGO 402 nucleic acid sequence (SEQ ID NO:56) and LOX-1 nucleic acid sequence (SEQ ID NO:66) are 42.0 % identical. The overall nucleic acid sequence identity between full-length human TANGO 402 (SEQ ID NO: 110) and full-length LOX-1 (SEQ ID NO:65) is 44.0 %.
Clone EpT402, which encodes human TANGO 402, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-294. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Puφoses of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of TANGO 402 Nucleic acids. Polypeptides. and Modulators Thereof
As TANGO 402 was originally found in a human fetal library, TANGO 402 nucleic acids, proteins, and modulators thereof can be used to diagnose disorders associated with cells, tissues, and/or organs in the embryo or fetus, or modulate the proliferation, development, differentiation, and/or function of cells, tissues, and/or organs in the embryo or fetus.
In addition, as TANGO 402 is homologous to LOX-1, TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to diagnose disorders, modulate development, differentiation, proliferation and/or activity of immune cells, such as macrophages and endothelial cells, e.g., the phagocytosis of aged/apoptotic cells by endothelial cells. TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat, inhibit and/or prevent disorders and diseases associated with the abeπant activity of the cells, tissues or organs in which TANGO 402 is expressed, e.g. endothelial activity. TANGO 402 nucleic acids, proteins and modulations thereof can also be used to diagnose disorders and/or modulate symptoms associated with atherosclerosis (e.g., atherosclerotic cardiovascular disease) and Alzheimer's disease. TANGO 402 nucleic acids, proteins and modulators thereof can be used to diagnose disorders associated with host immune defenses and/or modulate host immune defenses, e.g., modulating the activation of macrophages. TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat and/or prevent obesity, diabetes, and inflammatory disorders (e.g. , asthma, arthritis, multiple sclerosis, allergies, hepatitis and infections).
As TANGO 402 has homology to LOX-1 proteins, TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate TANGO 402 biological activities, which include, e.g., (1) the ability to bind proteins, e.g., lipoproteins, e.g., low- density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (2) the ability to modulate internalization of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (3) the ability to modulate degradation, e.g., proteolytic degradation, of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins; (4) the ability to modulate, e.g., increase, uptake of proteins, e.g., lipoproteins, e.g., low-density lipoproteins, e.g., oxidatively modified low-density lipoproteins, by cells, e.g., macrophages and muscle cells, e.g., smooth muscle cells; (5) the ability to modulate the function of a cell expressing LOX-1 or TANGO 402; (6) the ability to modulate the binding of a protein, e.g., oxidized low-density lipoprotein (Ox-LDL), to a cell which expresses LOX-1 or TANGO 402; and (7) the ability to modulate the binding of a protein, e.g. , Ox-LDL, to LOX-1 or TANGO 402.
TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to modulate, e.g., prevent, lipid deposition, e.g., in arteries, and thus modulate, e.g., prevent, intimal thickening; (2) the ability to modulate, e.g., induce or prevent, changes in cells, e.g., transformation of cells (e.g., macrophages and smooth muscle cells) into foam cells and functional alteration of cells (e.g., endothelial cells, e.g., intimal neovascular endothelial cells); (3) the ability to bind and phagocytose cells, e.g., aged and apoptotic cells; and (4) the ability to remove debris, e.g., apoptotic cells, from blood vessel walls. In another example, TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to modulate homeostasis, e.g., vascular homeostasis, e.g., by modulating, e.g., preventing the impairment of, nitric oxide production; (2) the ability to modulate, e.g., inhibit, the expression of molecules, e.g., adhesion molecules (e.g., leukocyte adhesion molecules) and growth factors (e.g., smooth- muscle growth factors); (3) the ability to alter, e.g., increase, expression in response to stimuli, e.g. , TNF, shear stress, and pathophysiological stimuli relevant to disorders (e.g. , atherosclerosis and inflammation).
In yet another example, TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate e.g., (1) the ability to form, e.g., stabilize, promote, facilitate, inhibit, or disrupt, cell-extracellular matrix interactions, e.g., adhesion between cells and extracellular matrix; (2) the ability to form, e.g., stabilize, promote, facilitate, inhibit, or disrupt, cell to cell and cell to blood product interaction, e.g. , between leukocytes and platelets or leukocytes and vascular endothelial cells; and (3) the ability to recognize large molecules, e.g., carbohydrates. In light of the fact that TANGO 402 is homologous to LOX-1 , TANGO 402 nucleic acids, proteins and modulators thereof have biological activities that can also include the ability to perform one or more of the functions of LOX-1 described, for example, in the following: Sawamura et al. (1997) Nature. 386:73-77; Kataoka et al. (1999) Circulation. 99:3110-3117; and Kita (1999) Circulation Research. 84:1113-1115, the contents of each of which is incoφorated herein by reference in its entirety. Moreover, due to TANGO 402's homology to LOX-1, as evidenced by the presence of similar domains and mapping coordinates between the two molecules, TANGO 402 nucleic acids, proteins and modulators thereof can be used to modulate or treat disorders in which LOX-1 plays a role, some of which are described in the following references: Sawamura et al. (1997) Nature. 386:73-77; Kataoka et al. (1999) Circulation. 99:3110-3117; and Kita (1999) Circulation Research. 84:1113-1115, the contents of each of which is incoφorated herein by reference in its entirety.
Furthermore, TANGO 402 nucleic acids, proteins and modulators thereof can modulate or treat atherosclerosis, e.g., by binding to oxidatively modified low density lipoprotein (Ox-LDL) and its lipid constituents, thus preventing lipid deposition and intimal thickening in the arteries, and thus preventing the induction of endothelial expression of leukocyte adhesion molecules and smooth-muscle growth factors (both which are implicated in atherogenesis).
In another example, TANGO 402 nucleic acids, proteins and modulators thereof modulate or treat immune related diseases and disorders. As LOX-1 is implicated in inflammation, and as LOX-1 has highest homology with the NKR-Pl family of proteins, which are involved in target-cell recognition and natural killer cell activation, TANGO 402 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate or treat inflammatory disorders such as bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, multiple sclerosis, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis), and processes. Further, TANGO 402 nucleic acids, proteins and modulators thereof can be used to identify, diagnose and/or modulate or treat immune disorders including, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), and T cell autoimmune disorders (e.g., AIDS)) and inflammatory disorders.
TANGO 402 nucleic acids, proteins and modulators thereof be used to identify, diagnose and/or modulate or treat TNF-related disorders, as LOX-1 expression is induced by tumor necrosis factors. Such disorders include, e.g., acute myocarditis, myocardial infarction, congestive heart failure, T cell disorders (e.g., dermatitis, fibrosis)), differentiative and apoptotic disorders, and disorders related to angiogenesis (e.g., tumor formation and/or metastasis, cancer). As LOX-1 expression is upregulated in hypertensive rats, and as LOX-1 levels are downregulated in patients treated with ACE (angiotensin converting enzyme) inhibitors, TANGO 402 can also play a role in treating hypertension and congestive heart failure.
As both TANGO 402 has C-type lectin domains or C-type lectin-like domains, and is similar in that respect to the selectins, which are implicated in cell-cell recognition (including endothelial-leukocyte adhesion), TANGO 402 nucleic acids, proteins and modulators thereof can be used to identify, diagnose and/or modulate or treat cell adhesion or cell migration/motility related disorders. Such disorders include, e.g., disorders associated with adhesion and migration of cells, e.g., platelet aggregation disorders (e.g., Glanzmarm's thromboasthemia, which is a bleeding disorders characterized by failure of platelet aggregation in response to cell stimuli), inflammatory disorders (e.g., leukocyte adhesion deficiency, which is a disorder associated with impaired migration of neutrophils to sites of extravascular inflammation), disorders associated with abnormal tissue migration during embryo development, and tumor metastasis. As TANGO 402 has a C-type lectin domain or C-type lectin-like domain, TANGO
402 nucleic acids, proteins and modulators thereof can be used to diagnose C-type lectin disorders and/or modulate calcium-dependent carbohydrate recognition. TANGO 402 proteins exhibit homology to lectins. In light of this, TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to modulate cell-cell, cell-extracellular matrix (ECM) interactions, cell adhesion, cell migration and cell signaling. TANGO 402 nucleic acids, proteins and modulators thereof can be utilized to treat and/or prevent disorders and diseases associated with abeπant cell-cell, cell-ECM interactions, cell migration, cell adhesion and cell-signaling, as well as treating and preventing tumor cell metastasis. TANGO 402 nucleic acids, proteins and modulators thereof can also be utilized to treat and/or prevent the migration of cancerous and precancerous cells (e.g., tumor migration). TANGO 402 nucleic acids, proteins and modulators thereof can also be used to modulate cell proliferation, e.g., abnormal cell proliferation. Such modulation may, for example, be via modulation of one or more elements involved in signal transduction cascades. TANGO 402 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., fetal tissues) and/or cells (e.g., fetal cells) in which TANGO 402 is expressed. TANGO 402 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human MANGO 346 A MANGO 346 cDNA was identified from clones present in a human brain library among sequences that encode signal peptides. This analysis led to the identification of a clone, jlhbab575g04, encoding full-length human MANGO 346. The human MANGO 346 cDNA of this clone is 1196 nucleotides long (Figure 29; SEQ ID NO: 123). The open reading frame of this cDNA, nucleotides 319 to 498 of SEQ ID NO: 123 (SEQ ID NO: 124), encodes a 60 amino acid secreted protein (Figure 18; SEQ ID NO: 125).
Figure 30 depicts a hydropathy plot of human MANGO 346. Relatively hydrophobic regions of the protein are shown above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence (amino acids 1 to 19 of SEQ ID NO:125; SEQ ID NO: 126) on the left from the mature protein (amino acids 20 to 60 of SEQ ID NO: 125; SEQ ID NO: 127) on the right.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10:1-6) predicted that human MANGO 346 includes a 19 amino acid signal peptide (amino acid 1 to amino acid 19 of SEQ ID NO:125; SEQ ID NO:126) preceding the mature human protein (coπesponding to amino acid 20 to amino acid 60 of SEQ ED NO: 125; SEQ ID NO: 127). The molecular weight of protein without post-translational modifications is 7.1 kDa prior to the cleavage of the signal peptide, 5.0 kDa after cleavage of the signal peptide.
In one embodiment of a nucleotide sequence of human MANGO 346, the nucleotide at position 13 is cytosine (C)(SEQ ID NO:124). In this embodiment, the amino acid at position 5 is leucine (L)(SEQ ID NO: 125). In an alternative embodiment, a species variant of human MANGO 346 has a nucleotide at position 13 which is adenine (A)(SEQ ID NO:216). In this embodiment, the amino acid at position 5 is isoleucine (I)(SEQ ID NO:217), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 346, the nucleotide at position 59 is adenine (A)(SEQ ID NO: 124). In this embodiment, the amino acid at position 20 is tyrosine (Y)(SEQ ID NO: 125). In an alternative embodiment, a species variant of human MANGO 346 has a nucleotide at position 59 which is thymidine (T)(SEQ ID NO:218). In this embodiment, the amino acid at position 20 is phenylalanine (F)(SEQ ID NO:219), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 346, the nucleotide at position 61 is thymidine (T)(SEQ ID NO: 124). In this embodiment, the amino acid at position 21 is serine (S)(SEQ ID NO: 125). In an alternative embodiment, a species variant of human MANGO 346 has a nucleotide at position 61 which is adenine (A)(SEQ ID NO:220). In this embodiment, the amino acid at position 21 is threonine (T)(SEQ ID NO:221), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 346, the nucleotide at position 80 is guanine (G)(SEQ ID NO: 124). In this embodiment, the amino acid at position 27 is arginine (R)(SEQ ED NO: 125). In an alternative embodiment, a species variant of human MANGO 346 has a nucleotide at position 80 which is adenine (A)(SEQ ID NO:222). In this embodiment, the amino acid at position 27 is lysine (K)(SEQ ID NO:223), i.e., a conservative substitution.
One protein kinase C phosphorylation site is present in human MANGO 346 which has the sequence, TIK (at amino acids 44 to 46 of SEQ ID NO: 125). Human
MANGO 346 has three Casein Kinase II phosphorylation sites. The first has the sequence SFLE (at amino acids 21 to 24 of SEQ ED NO: 125), the second has the sequence TIKE (at amino acids 44 to 47 of SEQ ID NO: 125) and the third has the sequence TYYD (at amino acids 51 to 54 of SEQ ID NO: 125). Human MANGO 346 has one prokaryotic membrane lipoprotein lipid attachment site. The sequence is CILPLLLLASC (at amino acids 6 to 16 of SEQ ID NO:125).
Clone EpM346, which encodes human MANGO 346, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-291. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Puφoses of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of MANGO 346 Nucleic acids. Polypeptides. and Modulators Thereof
As MANGO 346 was originally found in a human brain library, nucleic acids, proteins, and modulators thereof can be used to diagnose or identify disorders and/or modulate the proliferation, development, differentiation, and/or function of neural organs, e.g., neural tissues and cells, e.g., cells of the central nervous system, e.g., cells of the peripheral nervous system. MANGO 346 nucleic acids, proteins, and modulators thereof can also be used to diagnose or identify disorders and/or modulate symptoms associated with abnormal neural signaling and function, e.g., epilepsy, spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias. MANGO 346 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to diagnose disorders, regulate or modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs.
MANGO 346 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to regulate or diagnose disorders, modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs. Furthermore, the TANGO 346 proteins can be used to disrupt protein interaction or cellular signaling in brain tissues or cells. En particular, TANGO 346 proteins are useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g. , spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
As MANGO 346 is a secreted protein and thus likely a signaling molecule, MANGO 346 nucleic acids, proteins or modulators thereof, can be used to modulate MANGO 346 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g. , homophilic and/or heterophilic), and protein-ligand interactions, e.g., in receptor-ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate the proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g., signal transduction cascades).
MANGO 346 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g., the brain) and/or cells (e.g., neurons) in which MANGO 346 is expressed. MANGO 346 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g., in radiation hybrid mapping.
Human MANGO 349
A cDNA encoding human MANGO 349 was identified by analyzing the sequences of clones present in a human brain library for sequences that encode wholly secreted or transmembrane proteins. This analysis led to the identification of a clone, jlhbae318gd08, encoding full-length human MANGO 349. The human cDNA of this clone is 3649 nucleotides long (Figure 31; SEQ ID NO:128). The open reading frame of this cDNA, nucleotides 221 to 721 of SEQ ID NO:128 (SEQ ID NO:129), encodes a 167 amino acid secreted protein (Figure 31; SEQ ID NO: 130).
Figure 32 depicts a hydropathy plot of human MANGO 349. Relatively hydrophobic regions of the protein are above the horizontal line, and relatively hydrophilic regions of the protein are below the horizontal line. The cysteine residues (cys) are indicated by short vertical lines just below the hydropathy trace. The dashed vertical line separates the signal sequence from the mature protein described below.
The signal peptide prediction program SIGNALP (Nielsen et al., 1997, Protein Engineering 10: 1-6) predicted that human MANGO 349 includes a 26 amino acid signal peptide (amino acid 1 to amino acid 26 of SEQ ID NO:130; SEQ ID NO:131) preceding the mature human protein (coπesponding to amino acid 27 to amino acid 167 of SEQ ID NO:130; SEQ ED NO:132). The molecular weight of human protein without post- translational modifications is 17.6 kDa prior to the cleavage of the signal peptide, 15.1 kDa after cleavage of the signal peptide. In one embodiment of a nucleotide sequence of human MANGO 349, the nucleotide at position 4 is adenine (A)(SEQ ID NO: 129). In this embodiment, the amino acid at position 2 is threonine (T)(SEQ ID NO: 130). In an alternative embodiment, a species variant of human MANGO 349 has a nucleotide at position 4 which is thymine (T)(SEQ ID NO:224). In this embodiment, the amino acid at position 2 is serine (S)(SEQ ID NO:225), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 349, the nucleotide at position 61 is adenine (A)(SEQ ED NO: 129). En this embodiment, the amino acid at position 21 is isoleucine (I)(SEQ ED NO: 130). In an alternative embodiment, a species variant of human MANGO 349 has a nucleotide at position 61 which is cytosine (C)(SEQ ID NO:226). In this embodiment, the amino acid at position 21 is leucine (L)(SEQ ID NO:227), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 349, the nucleotide at position 86 is guanine (G)(SEQ ID NO: 129). In this embodiment, the amino acid at position 29 is arginine (R)(SEQ ID NO: 130). In an alternative embodiment, a species variant of human MANGO 349 has a nucleotide at position 86 which is adenine (A)(SEQ ID NO:228). In this embodiment, the amino acid at position 29 is lysine (K)(SEQ ID NO:229), i.e., a conservative substitution.
In one embodiment of a nucleotide sequence of human MANGO 349, the nucleotide at position 123 is guanine (G)(SEQ ID NO: 129). In this embodiment, the amino acid at position 41 is glutamate (E)(SEQ ID NO: 130). In an alternative embodiment, a species variant of human MANGO 349 has a nucleotide at position 123 which is cytosine (C)(SEQ ID NO:230). In this embodiment, the amino acid at position 41 is aspartate (D)(SEQ ID NO:231), i.e., a conservative substitution.
Two Protein C Kinase phosphorylation sites are present in human MANGO 349. The first has the sequence SLK (at amino acids 136 to 139 of SEQ ID NO: 130) and the second has the sequence SGR (at amino acids 152 to 154 of SEQ ID NO: 130). Two casein kinase II phosphorylation sites are present in human MANGO 349. The first has the sequence SGTE (at amino acids 38 to 41 of SEQ ID NO: 130), and the second has the sequence SGRE (at amino acids 152 to 155 of SEQ ID NO: 130). Human MANGO 349 has four N-myristylation sites. The first has the sequence GGILAT (at amino acids 10 to 15 of SEQ ID NO: 130), the second has the sequence GTEVAD (at amino acids 39 to 44 of SEQ ID NO: 130), the third has the sequence GVAASH (at amino acids 89 to 94 of SEQ ID NO: 17), and the fourth has the sequence GGPPSL (at amino acids 132 to 137 of SEQ ID NO: 130).
Clone EpM349, which encodes human MANGO 349, was deposited with the American Type Culture Collection (10801 University Boulevard, Manassas, VA 20110- 2209) on June 29, 1999 and assigned Accession Number PTA-295. This deposit will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Puφoses of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112.
Uses of MANGO 349 Nucleic acids. Polypeptides. and Modulators Thereof
As MANGO 349 was originally found in a human brain library, nucleic acids, proteins, and modulators thereof can be used to diagnose or identify disorders and/or modulate the proliferation, development, differentiation, and/or function of neural organs, e.g., neural tissues and cells, e.g., cells of the central nervous system, e.g., cells of the peripheral nervous system. MANGO 349 nucleic acids, proteins, and modulators thereof can also be used to diagnose or identify disorders and/or modulate symptoms associated with abnormal neural signaling and function, e.g., epilepsy, stroke, traumatic injury, etc. MANGO 349 nucleic acids, proteins and modulators thereof can, in addition to the above, be utilized to diagnose disorders, regulate or modulate development and/or differentiation of processes involved in central or peripheral nervous system formation and activity, as well as in ameliorating any symptom associated with a disorder of such cell types, tissues and organs. Furthermore, the TANGO 349 proteins can be used to disrupt protein interaction or cellular signaling in brain tissues or cells. In particular, TANGO 349 proteins could be useful to treat neural related disorders or neural damage, such as for regenerative neural repair after damage by trauma, degeneration, or inflammation e.g., spinal cord injuries, infarction, infection, malignancy, exposure to toxic agents, nutritional deficiency, paraneoplastic syndromes, and degenerative nerve diseases including but not limited to Alzheimer's disease, Parkinson's disease, Huntington's Chorea, amyotrophic lateral sclerosis, progressive supra-nuclear palsy, and other dementias.
As MANGO 349 is a secreted protein and thus likely a signaling molecular, MANGO 349 nucleic acids, proteins and modulators thereof can be used to diagnose disorders and/or modulate MANGO 349 biological activities, which include, e.g., (1) the ability to modulate, e.g., stabilize, promote, inhibit or disrupt, protein-protein interactions (e.g., homophilic and/or heterophilic), and protein-ligand interactions, e.g. in receptor- ligand recognition; (2) ability to modulate cell-cell interactions; (3) the ability to modulate proliferation, differentiation and/or activity of neural cells; and (4) the ability to modulate intracellular signaling cascades (e.g. signal transduction cascades).
MANGO 349 expression can be utilized as a marker (e.g., an in situ marker) for specific tissues (e.g. , the brain) and/or cells (e.g. , neurons) in which MANGO 349 is expressed. MANGO 349 nucleic acids can also be utilized for chromosomal mapping, or as chromosomal markers, e.g. , in radiation hybrid mapping.
Tables 1 provides a summary of the sequence information for TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, human TANGO 393, mouse TANGO 393, TANGO 402, MANGO 346, and MANGO 349.
Table 2 provides a summary of the domains of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, human TANGO 393, mouse TANGO 393, TANGO 402, MANGO 346, and MANGO 349. It is noted that human and mouse TANGO 393 leucine-rich repeats are not included in Table 2, but are described supra.
TABLE 1 : Summary of Human TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, MANGO 349 and Mouse TANGO 393 Sequence Information
TABLE 2: Summary of Domains of TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 Proteins
Various aspects of the invention are described in further detail in the following subsections: I. Isolated Nucleic Acid Molecules
One aspect of the invention pertains to isolated nucleic acid molecules that encode a polypeptide of the invention or a biologically active portion thereof, as well as nucleic acid molecules sufficient for use as hybridization probes to identify nucleic acid molecules encoding a polypeptide of the invention and fragments of such nucleic acid molecules suitable for use as PCR primers for the amplification or mutation of nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be single-stranded or double-stranded, but preferably is double-stranded DNA.
An "isolated" nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid molecule. Preferably, an "isolated" nucleic acid molecule is free of sequences (preferably protein encoding sequences) which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated nucleic acid molecule can contain less than about 5 kB, 4 kB, 3 kB, 2 kB, 1 kB, 0.5 kB or 0.1 kB of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized. As used herein, the term "isolated"when referring to a nucleic acid molecule does not include an isolated chromosome.
In instances wherein the nucleic acid molecule is a cDNA or RNA, e.g., mRNA, molecule, such molecules can include a poly A "tail", or, alternatively, can lack such a 3' tail. Although cDNA or RNA nucleotide sequences may be depicted herein with such tail sequences, it is to be understood that cDNA nucleic acid molecules of the invention are also intended to include such sequences lacking the depicted poly A tails.
A nucleic acid molecule of the present invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230 or a complement thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion of the nucleic acid sequences of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230 as a hybridization probe, nucleic acid molecules of the invention can be isolated using standard hybridization and cloning techniques (e.g. , as described in Sambrook et al., eds., Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989).
A nucleic acid molecule of the invention can be amplified using cDNA, mRNA or genomic DNA as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to all or a portion of a nucleic acid molecule of the invention can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
In another preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of the nucleotide sequence of SEQ ED NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, or a portion thereof. A nucleic acid molecule which is complementary to a given nucleotide sequence is one which is sufficiently complementary to the given nucleotide sequence that it can hybridize to the given nucleotide sequence under the conditions set forth herein, thereby forming a stable duplex.
Moreover, a nucleic acid molecule of the invention can comprise only a portion of a nucleic acid sequence encoding a full length polypeptide of the invention for example, a fragment which can be used as a probe or primer or a fragment encoding a biologically active portion of a polypeptide of the invention. The nucleotide sequence determined from the cloning one gene allows for the generation of probes and primers designed for use in identifying and/or cloning homologs in other cell types, e.g. , from other tissues, as well as homologs from other mammals. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, preferably about 25, more preferably about 50, 75, 100, 125, 150, 175, 200, 250, 300, 350 or 400 consecutive nucleotides of the sense or anti-sense sequence of SEQ ID NO: 1 , 2, 27, 28,
36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291 , PTA 292, PTA 294 or PTA 295, or of a naturally occurring mutant of SEQ ID NO:l, 2, 27, 28, 36,
37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129. In another embodiment, the oligonucleotide comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least 400, preferably 450, 500, 530, 550, 600, 700,800, 900, 1000 or 1150 consecutive oligonucleotides of the sense or antisense sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, or of a naturally occurring mutant of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, or 129.
Probes based on the sequence of a nucleic acid molecule of the invention can be used to detect transcripts or genomic sequences encoding the same protein molecule encoded by a selected nucleic acid molecule. The probe comprises a label group attached thereto, e.g., a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as part of a diagnostic test kit for identifying cells or tissues which mis-express the protein, such as by measuring levels of a nucleic acid molecule encoding the protein in a sample of cells from a subject, e.g., detecting mRNA levels or determining whether a gene encoding the protein has been mutated or deleted.
A nucleic acid fragment encoding a biologically active portion of a polypeptide of the invention can be prepared by isolating a portion of any of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, expressing the encoded portion of the polypeptide protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of the polypeptide. The invention further encompasses nucleic acid molecules that differ from the nucleotide sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, due to degeneracy of the genetic code and thus encode the same protein as that encoded by the nucleotide sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221 , 223, 225, 227, 229 or 231.
In addition to the nucleotide sequences of SEQ ED NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the nucleotide sequence of the cDNA of a clone deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequence may exist within a population (e.g., the human population). Such genetic polymorphisms may exist among individuals within a population due to natural allelic variation.
An allele is one of a group of genes which occur alternatively at a given genetic locus. For example, TANGO 393 has been mapped to chromosome 22, and therefore TANGO 393 family members can include nucleotide sequence polymorphisms (e.g., nucleotide sequences that vary from SEQ ID NO:73 and SEQ ID NO:74) that map to this chromosome 22 region, and such sequences represent TANGO 393 allelic variants. In another example, TANGO 339 has been mapped to chromosome 10, and therefore TANGO 339, family members can include nucleotide sequence polymorphisms (e.g., nucleotide sequences that vary from SEQ ID NO:l and SEQ ID NO:2) that map to this chromosome 10 region, and such sequences represent allelic variants. As used herein, the phrase "allelic variant" refers to a nucleotide sequence which occurs at a given locus or to a polypeptide encoded by the nucleotide sequence. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame encoding a polypeptide of the invention. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of a given gene. Alternative alleles can be identified by sequencing the gene of interest in a number of different individuals. This can be readily carried out by using hybridization probes to identify the same genetic locus in a variety of individuals. Any and all such nucleotide variations and resulting amino acid polymorphisms or variations that are the result of natural allelic variation and that do not alter the functional activity are intended to be within the scope of the invention. In one embodiment, polymorphisms that are associated with a particular disease and/or disorder are used as markers to diagnose said disease or disorder. In a preferred embodiment, polymorphisms are used as a marker to diagnose abnormal coronary function such as atherosclerosis. Moreover, nucleic acid molecules, encoding proteins of the invention from other species (homologs), which have a nucleotide sequence which differs from that of the human or mouse protein described herein are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologs of a cDNA of the invention can be isolated based on their identity to the human nucleic acid molecule disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. For example, a cDNA encoding a soluble form of a membrane-bound protein of the invention isolated based on its hybridization to a nucleic acid molecule encoding all or part of the membrane-bound form. Likewise, a cDNA encoding a membrane-bound form can be isolated based on its hybridization to a nucleic acid molecule encoding all or part of the soluble form.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900 or 2000 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence, preferably the coding sequence, of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123, or 128 or a complement thereof.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 20, 50, 100, 200, 300, 400, 500, 600, 700, 800 or 900 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence, preferably the coding sequence, of SEQ ID NO:2, 28, 37, 45, 53, 59, 64, 74, 94, 111, 124, or, 129, or a complement thereof.
As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% (65%, 70%, preferably 75%) identical to each other typically remain hybridized to each other. Such stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45° C, followed by one or more washes in 0.2 X SSC, 0.1% SDS at 50-65° C. Preferably, an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or a complement thereof, corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
In addition to naturally-occurring allelic variants of a nucleic acid molecule of the invention sequence that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation thereby leading to changes in the amino acid sequence of the encoded protein, without altering the biological activity of the protein. For example, one can make nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequence without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity. For example, amino acid residues that are not conserved or only semi-conserved among homologs of various species may be non-essential for activity and thus would be likely targets for alteration. Specific examples of conservative amino acid alterations from the original sequence are shown in SEQ ID NOs:137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231. Alternatively, amino acid residues that are conserved among the orthologs of various species (e.g., murine and human) may be essential for activity and thus would not be likely targets for alteration. Accordingly, another aspect of the invention pertains to nucleic acid molecules encoding a polypeptide of the invention that contain changes in amino acid residues that are not essential for activity. Such polypeptides differ in amino acid sequence from SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule includes a nucleotide sequence encoding a protein that includes an amino acid sequence that is at least about 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 75%), 85%o, 95%), or 98% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231. An isolated nucleic acid molecule encoding a variant protein can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ED NO: 1, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or complement thereof, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid, asparagine, glutamine), uncharged polar side chains (e.g., glycine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for biological activity to identify mutants that retain activity. Following mutagenesis, the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
In a preferred embodiment, a mutant polypeptide that is a variant of a polypeptide of the invention can be assayed for: (1) the ability to form protein-protein interactions with proteins in a signaling pathway of the polypeptide of the invention such as in central nervous system cells, lymphoid cells, hypothalamus cells, or prostate cells with the proteins encoded by the genes of the present invention (e.g., leucine-rich repeat interactions and transmembrane 4); (2) the ability to bind a ligand of the polypeptide of the invention (i.e., in transmembrane proteins of the invention or alternatively, secreted proteins which are the ligand for a cellular receptor); or (3) the ability to bind to an intracellular target protein of the polypeptide of the invention. In yet another preferred embodiment, the mutant polypeptide can be assayed for the ability to modulate cellular proliferation, cellular migration or chemotaxis, or cellular differentiation.
The present invention encompasses antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid encoding a polypeptide of the invention, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid. The antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof, e.g., all or part of the protein coding region (or open reading frame). An antisense nucleic acid molecule can be antisense to all or part of a non-coding region of the coding strand of a nucleotide sequence encoding a polypeptide of the invention. The non-coding regions ("51 and 3' untranslated regions") are the 5' and 3' sequences which flank the coding region and are not translated into amino acids. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35,
40, 45 or 50 nucleotides or more in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection). The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a selected polypeptide of the invention to thereby inhibit expression, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g. , by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
An antisense nucleic acid molecule of the invention can be an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids Res. 15:6625-6641). The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1981) FEBS Lett. 215:327-330).
The invention also encompasses ribozymes. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave mRNA transcripts to thereby inhibit translation of the protein encoded by the mRNA. A ribozyme having specificity for a nucleic acid molecule encoding a polypeptide of the invention can be designed based upon the nucleotide sequence of a cDNA disclosed herein. For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a Cech et al. U.S. Patent No. 4,987,071; and Cech et al. U.S. Patent No. 5,116,742. Alternatively, an mRNA encoding a polypeptide of the invention can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel and Szostak (1993) Science 261 :1411-1418.
The invention also encompasses nucleic acid molecules which form triple helical structures. For example, expression of a polypeptide of the invention can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the gene encoding the polypeptide (e.g. , the promoter and/or enhancer) to form triple helical structures that prevent transcription of the gene in target cells. See generally Helene (1991) Anticancer Drug Des. 6(6):569-84; Helene (1992) Ann. N Y. Acad. Sci. 660:27-36; and Maher (1992) Bioassays 14(12):807-15.
In various embodiments, the nucleic acid molecules of the invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g. , the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids (see Hyrup et al. (1996) Bioorganic & Medicinal Chemistry 4(1): 5-23). As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup et al. (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675. PNAs can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs can also be used, e.g., in the analysis of single base pair mutations in a gene by, e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., SI nucleases (Hyrup (1996), supra; or as probes or primers for DNA sequence and hybridization (Hyrup (1996), supra; Perry-O'Keefe et al. (1996) Proc. Natl. Acad. Sci. USA 93: 14670-675).
In another embodiment, PNAs can be modified, e.g., to enhance their stability or cellular uptake, by attaching Hpophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras can be generated which may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes, e.g., RNAse H and DNA polymerases, to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup (1996), supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup (1996), supra, and Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs. Compounds such as 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite can be used as a link between the PNA and the 5' end of DNA (Mag et al. (1989) Nucleic Acids Res. 17:5973-88). PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment (Finn et al. (1996) Nucleic Acids Res. 24(17):3357-63). Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment (Peterser et al. (1975) Bioorganic Med. Chem. Lett. 5:1119-11124).
In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo ), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Acad. Sci. USA 84:648-652; PCT Publication No. W0 88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. W0 89/10134). In addition, oligonucleotides can be modified with hybridization-triggered cleavage agents (see, e.g., Krol et al. (1988) Bio/Techniques 6:958-976) or intercalating agents (see, e.g., Zon (1988) Pharm. Res. 5:539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, etc.
II. Isolated Proteins and Antibodies One aspect of the invention pertains to isolated proteins, and biologically active portions thereof, as well as polypeptide fragments suitable for use as immunogens to raise antibodies directed against a polypeptide of the invention. In one embodiment, the native polypeptide can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, polypeptides of the invention are produced by recombinant DNA techniques. Alternative to recombinant expression, a polypeptide of the invention can be synthesized chemically using standard peptide synthesis techniques.
An "isolated" or "purified" protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, protein that is substantially free of cellular material includes preparations of protein having less than about 30%, 20%, 10%, or 5%> (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein"). When the protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%>, 10%, or 5% of the volume of the protein preparation. When the protein is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly such preparations of the protein have less than about 30%, 20%), 10%), 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest. Biologically active portions of a polypeptide of the invention include polypeptides comprising amino acid sequences sufficiently identical to or derived from the amino acid sequence of the protein (e.g., the amino acid sequence shown in any of SEQ ID NO:6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 32, 33, 34, 41, 42, 43, 49, 50, 51, 68, 69, 70, 71, 72, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 115, 116, 117, 118, 119, 133, 134, 135, or 136, which include fewer amino acids than the full length protein, and exhibit at least one activity of the corresponding full-length protein. Typically, biologically active portions comprise a domain or motif with at least one activity of the coπesponding protein. A biologically active portion of a protein of the invention can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acids in length. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of the native form of a polypeptide of the invention.
Prefeπed polypeptides have the amino acid sequence of SEQ ID NO:6, 7, 8, 9, 10, 19, 20, 21, 22 or 23. Other useful proteins are substantially identical (e.g., at least about 45%, preferably 55%, 65%, 75%, 85%, 95%, or 99%) to any of SEQ ID NO:6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 32, 33, 34, 41, 42, 43, 49, 50, 51, 68, 69, 70, 71, 72, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 115, 116, 117, 118, 119, 133, 134, 135, or 136, and retain the functional activity of the protein of the coπesponding naturally-occurring protein yet differ in amino acid sequence due to natural allelic variation or mutagenesis.
To determine the percent identity of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g. , gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at coπesponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the coπesponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions (e.g., overlapping positions) x 100). In one embodiment, the two sequences are the same length.
The determination of percent identity between two sequences can be accomplished using a mathematical algorithm. A prefeπed, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to a nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to a protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402. Alternatively, PSI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.). When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov.
Another prefeπed, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, (1988) CABIOS 4: 11-17. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM as described in Torellis and Robotti (1994) Comput. Appl Biosci., 10:3-5; and FASTA described in Pearson and Lipman (1988) Proc. Natl. Acad. Sci. 55:2444-8. Within FASTA, ktup is a control option that sets the sensitivity and speed of the search. If ktup=2, similar regions in the two sequences being compared are found by looking at pairs of aligned residues; if ktup=l, single aligned amino acids are examined, ktup can be set to 2 or 1 for protein sequences, or from 1 to 6 for DNA sequences. The default if ktup is not specified is 2 for proteins and 6 for DNA. For a further description of FASTA parameters, see http://bioweb.pasteur.fr/docs/man man/fasta. I.html#sect2, the contents of which are incoφorated herein by reference.
The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, only exact matches are counted.
The invention also provides chimeric or fusion proteins. As used herein, a "chimeric protein" or "fusion protein" comprises all or part (preferably biologically active) of a polypeptide of the invention operably linked to a heterologous polypeptide (i.e., a polypeptide other than the same polypeptide of the invention). Within the fusion protein, the term "operably linked" is intended to indicate that the polypeptide of the invention and the heterologous polypeptide are fused in-frame to each other. The heterologous polypeptide can be fused to the N-terminus or C-terminus of the polypeptide of the invention. One useful fusion protein is a GST fusion protein in which the polypeptide of the invention is fused to the C-terminus of GST sequences. Such fusion proteins can facilitate the purification of a recombinant polypeptide of the invention.
In another embodiment, the fusion protein contains a heterologous signal sequence at its N-terminus. For example, the native signal sequence of a polypeptide of the invention can be removed and replaced with a signal sequence from another protein. For example, the gp67 secretory sequence of the baculovirus envelope protein can be used as a heterologous signal sequence (Current Protocols in Molecular Biology, Ausubel et al., eds., John Wiley & Sons, 1992). Other examples of eukaryotic heterologous signal sequences include the secretory sequences of melittin and human placental alkaline phosphatase (Stratagene; La Jolla, California). In yet another example, useful prokaryotic heterologous signal sequences include the phoA secretory signal (Sambrook et al., supra) and the protein A secretory signal (Pharmacia Biotech; Piscataway, New Jersey).
In yet another embodiment, the fusion protein is an immunoglobulin fusion protein in which all or part of a polypeptide of the invention is fused to sequences derived from a member of the immunoglobulin protein family. The immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject to inhibit an interaction between a ligand (soluble or membrane-bound) and a protein on the surface of a cell (receptor), to thereby suppress signal transduction in vivo. The immunoglobulin fusion protein can be used to affect the bioavailability of a cognate ligand of a polypeptide of the invention. Inhibition of ligand/receptor interaction may be useful therapeutically, both for treating proliferative and differentiative disorders and for modulating (e.g. , promoting or inhibiting) cell survival. Moreover, the immunoglobulin fusion proteins of the invention can be used as immunogens to produce antibodies directed against a polypeptide of the invention in a subject, to purify ligands and in screening assays to identify molecules which inhibit the interaction of receptors with ligands. Chimeric and fusion proteins of the invention can be produced by standard recombinant DNA techniques. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel et al., supra). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). A nucleic acid encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide of the invention. A signal sequence of a polypeptide of the invention (SEQ ED NO:5, 31, 40, 47, 56, 62, 66, 76, 96, 114, 126 or 131) can be used to facilitate secretion and isolation of the secreted protein or other proteins of interest. Signal sequences are typically characterized by a core of hydrophobic amino acids which are generally cleaved from the mature protein during secretion in one or more cleavage events. Such signal peptides contain processing sites that allow cleavage of the signal sequence from the mature proteins as they pass through the secretory pathway. Thus, the invention pertains to the described polypeptides having a signal sequence, as well as to the signal sequence itself and to the polypeptide in the absence of the signal sequence (i.e., the cleavage products). In one embodiment, a nucleic acid sequence encoding a signal sequence of the invention can be operably linked in an expression vector to a protein of interest, such as a protein which is ordinarily not secreted or is otherwise difficult to isolate. The signal sequence directs secretion of the protein, such as from a eukaryotic host into which the expression vector is transformed, and the signal sequence is subsequently or concuπently cleaved. The protein can then be readily purified from the extracellular medium by art recognized methods. Alternatively, the signal sequence can be linked to the protein of interest using a sequence which facilitates purification, such as with a GST domain.
In another embodiment, the signal sequences of the present invention can be used to identify regulatory sequences, e.g., promoters, enhancers, repressors. Since signal sequences are the most amino-terminal sequences of a peptide, it is expected that the nucleic acids which flank the signal sequence on its amino-terminal side will be regulatory sequences which affect transcription. Thus, a nucleotide sequence which encodes all or a portion of a signal sequence can be used as a probe to identify and isolate signal sequences and their flanking regions, and these flanking regions can be studied to identify regulatory elements therein.
The present invention also pertains to variants of the polypeptides of the invention. Such variants have an altered amino acid sequence which can function as either agonists (mimetics) or as antagonists. Variants can be generated by mutagenesis, e.g. , discrete point mutation or truncation. An agonist can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of the protein. An antagonist of a protein can inhibit one or more of the activities of the naturally occurring form of the protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the protein of interest. Thus, specific biological effects can be elicited by treatment with a variant of limited function. Treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein can have fewer side effects in a subject relative to treatment with the naturally occurring form of the protein.
Variants of a protein of the invention which function as either agonists (mimetics) or as antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the protein of the invention for agonist or antagonist activity. In one embodiment, a variegated library of variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential protein sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display). There are a variety of methods which can be used to produce libraries of potential variants of the polypeptides of the invention from a degenerate oligonucleotide sequence. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang (1983) Tetrahedron 39:3; Itakura et al. (1984) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477).
In addition, libraries of fragments of the coding sequence of a polypeptide of the invention can be used to generate a variegated population of polypeptides for screening and subsequent selection of variants. For example, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of the coding sequence of interest with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with SI nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes N-terminal and internal fragments of various sizes of the protein of interest.
Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. The most widely used techniques, which are amenable to high through-put analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify variants of a protein of the invention (Arkin and Yourvan (1992) Proc. Natl Acad. Sci. USA 59:7811-7815; Delgrave et al. (1993) Protein Engineering 6(3):327-331).
The polypeptides of the invention can exhibit post-translational modifications, including, but not limited to glycosylations, (e.g., N-linked or O-linked glycosylations), myristylations, palmitylations, acetylations and phosphorylations (e.g., serine/threonine or tyrosine). In one embodiment, the TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 polypeptides of the invention exhibit reduced levels of O-linked glycosylation and/or N-linked glycosylation relative to endogenously expressed TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, MANGO 349 polypeptides. In another embodiment, the TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 or MANGO 349 polypeptides of the invention do not exhibit O-linked glycosylation or N-linked glycosylation.
The polypeptides of the invention can, for example, include modifications that can increase such attributes as stability, half-life, ability to enter cells and aid in administration, e.g., in vivo administration of the polypeptides of the invention. For example, polypeptides of the invention can comprise a protein transduction domain of the HIV TAT protein as described in Schwarze, et al. (1999 Science 285:1569-1572), thereby facilitating delivery of polypeptides of the invention into cells.
An isolated polypeptide of the invention, or a fragment thereof, can be used as an immunogen to generate antibodies using standard techniques for polyclonal and monoclonal antibody preparation. The full-length polypeptide or protein can be used or, alternatively, the invention provides antigenic peptide fragments for use as immunogens. The antigenic peptide of a protein of the invention comprises at least 8 (preferably 10, 15, 20, or 30) amino acid residues of the amino acid sequence of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, and encompasses an epitope of the protein such that an antibody raised against the peptide forms a specific immune complex with the protein.
Prefeπed epitopes encompassed by the antigenic peptide are regions that are located on the surface of the protein, e.g., hydrophilic regions. Figures 2, 6, 8, 10, 12, 15, 17, 20, 22, 26, 30, and 32 are hydropathy plots of the proteins of the invention. These plots or similar analyses can be used to identify hydrophilic regions. In certain embodiments, the nucleic acid molecules of the invention are present as part of nucleic acid molecules comprising nucleic acid sequences that contain or encode heterologous (e.g. , vector, expression vector, or fusion protein) sequences. These nucleotides can then be used to express proteins which can be used as immunogens to generate an immune response, or more particularly, to generate polyclonal or monoclonal antibodies specific to the expressed protein.
An immunogen typically is used to prepare antibodies by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal). An appropriate immunogenic preparation can contain, for example, recombinantly expressed or chemically synthesized polypeptide. The preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent.
Accordingly, another aspect of the invention pertains to antibodies directed against a polypeptide of the invention. The term "antibody" as used herein refers to immunoglobulin molecules and immuno logically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site which specifically binds an antigen, such as a polypeptide of the invention, e.g., an epitope of a polypeptide of the invention. A molecule which specifically binds to a given polypeptide of the invention is a molecule which binds the polypeptide, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin. The invention provides polyclonal and monoclonal antibodies. The term "monoclonal antibody" or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope.
Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a polypeptide of the invention as an immunogen. Prefeπed polyclonal antibody compositions are ones that have been selected for antibodies directed against a polypeptide or polypeptides of the invention. Particularly prefeπed polyclonal antibody preparations are ones that contain only antibodies directed against a polypeptide or polypeptides of the invention. Particularly prefeπed immunogen compositions are those that contain no other human proteins such as, for example, immunogen compositions made using a non-human host cell for recombinant expression of a polypeptide of the invention. In such a manner, the only human epitope or epitopes recognized by the resulting antibody compositions raised against this immunogen will be present as part of a polypeptide or polypeptides of the invention.
The antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide. If desired, the antibody molecules can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction. Alternatively, antibodies specific for a protein or polypeptide of the invention can be selected for (e.g., partially purified) or purified by, e.g., affinity chromatography. For example, a recombinantly expressed and purified (or partially purified) protein of the invention is produced as described herein, and covalently or non-covalently coupled to a solid support such as, for example, a chromatography column. The column can then be used to affinity purify antibodies specific for the proteins of the invention from a sample containing antibodies directed against a large number of different epitopes, thereby generating a substantially purified antibody composition, i.e., one that is substantially free of contaminating antibodies. By a substantially purified antibody composition is meant, in this context, that the antibody sample contains at most only 30% (by dry weight) of contaminating antibodies directed against epitopes other than those on the desired protein or polypeptide of the invention, and preferably at most 20%>, yet more preferably at most 10%>, and most preferably at most 5% (by dry weight) of the sample is contaminating antibodies. A purified antibody composition means that at least 99% of the antibodies in the composition are directed against the desired protein or polypeptide of the invention.
At an appropriate time after immunization, e.g., when the specific antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96) or trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al. (eds.) John Wiley & Sons, Inc., New York, NY). Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind the polypeptide of interest, e.g., using a standard ELISA assay.
Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody directed against a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide of interest. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271; PCT Publication No. WO 92/20791; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffiths et al. (1993) EMBOJ. 12:725-734.
Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region. (See, e.g., Cabilly et al., U.S. Patent No. 4,816,567; and Boss et al., U.S. Patent No. 4,816,397, which are incoφorated herein by reference in their entirety.) Humanized antibodies are antibody molecules from non- human species having one or more complementarily determining regions (CDRs) from the non-human species and a framework region from a human immunoglobulin molecule. (See, e.g., Queen, U.S. Patent No. 5,585,089, which is incoφorated herein by reference in its entirety.) Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No. WO 86/01533; U.S. Patent No. 4,816,567; European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043; Liu et al. (1987) Proc. Natl Acad. Sci. USA 84:3439-3443; Liu et al. (1987) J Immunol. 139:3521-3526; Sun et al. (1987) Proc. Natl. Acad. Sci. USA 84:214-218; Nishimura et al. (1987) Cane. Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al. (1988) J. Natl. Cancer Inst. 80:1553-1559); Morrison (1985) Science 229:1202-1207; Oi et al. (1986) Bio/Techniques 4:214; U.S. Patent 5,225,539; Jones et al. (1986) Nαtwre 321:552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol. 141:4053-4060. Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Such antibodies can be produced, for example, using transgenic mice which are incapable of expressing endogenous immunoglobulin heavy and light chains genes, but which can express human heavy and light chain genes. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice reaπange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar (1995, Int. Rev. Immunol. 13:65-93). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., U.S. Patent 5,625,126; U.S. Patent 5,633,425; U.S. Patent 5,569,825; U.S. Patent 5,661,016; and U.S. Patent 5,545,806. In addition, companies such as Abgenix, Inc. (Freemont, CA), can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
Completely human antibodies which recognize a selected epitope can be generated using a technique refeπed to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a murine antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al. (1994) Bio/technology 12:899-903).
An antibody directed against a polypeptide of the invention (e.g., monoclonal antibody) can be used to isolate the polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. Moreover, such an antibody can be used to detect the protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the polypeptide. The antibodies can also be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125^ 131j) 35g or 3fj. Further, an antibody (or fragment thereof) can be conjugated to a therapeutic moiety such as a cytotoxin, a therapeutic agent or a radioactive metal ion. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydro testosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g. , mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g. , daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
The conjugates of the invention can be used for modifying a given biological response, the drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, α-interferon, β-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-l"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), interleukin- 10 ("IL- 10"), interleukin- 12 ("IL- 12"), interferon-γ ("EFN-γ"), interferon-α ("EFN-α"), granulocyte macrophase colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other immune or growth factors.
Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243- 56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thoφe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thoφe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol. Rev., 62:119-58 (1982).
Alternatively, an antibody of the invention can be conjugated to a second antibody to form an "antibody heteroconjugate" as described by Segal in U.S. Patent No. 4,676,980 or alternatively, the antibodies can be conjugated to form an "antibody heteropolymer" as described in Taylor et al, in U.S. Patent Nos. 5,470,570 and 5,487,890.
An antibody with or without a therapeutic moiety conjugated to it can be used as a therapeutic that is administered alone or in combination with cytotoxic factor(s) and/or cytokine(s).
In yet a further aspect, the invention provides substantially purified antibodies or fragments thereof, including human or non-human antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide of the invention comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; a fragment of at least 15 amino acid residues of the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, wherein the percent identity is determined using the ALIGN program of the GCG software package with a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4; and an amino acid sequence which is encoded by a nucleic acid molecule which hybridizes to the nucleic acid molecule consisting of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or to the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291 , PTA 292, PTA 294 or PTA 295, under conditions of hybridization of 6X SSC at 45°C and washing in 0.2 X SSC, 0.1% SDS at 65°C. In various embodiments, the substantially purified antibodies of the invention, or fragments thereof, can be human, non-human, chimeric and/or humanized antibodies.
In another aspect, the invention provides human or non-human antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; a fragment of at least 15 amino acid residues of the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, wherein the percent identity is determined using the ALIGN program of the GCG software package with a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4; and an amino acid sequence which is encoded by a nucleic acid molecule which hybridizes to the nucleic acid molecule consisting of SEQ ID NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230 or to the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, under conditions of hybridization of 6X SSC at 45°C and washing in 0.2 X SSC, 0.1% SDS at 65°C. Such non-human antibodies can be goat, mouse, sheep, horse, chicken, rabbit, or rat antibodies. Alternatively, the non-human antibodies of the invention can be chimeric and/or humanized antibodies. In addition, the non-human antibodies of the invention can be polyclonal antibodies or monoclonal antibodies.
In still a further aspect, the invention provides monoclonal antibodies or fragments thereof, which antibodies or fragments specifically bind to a polypeptide of the invention comprising an amino acid sequence selected from the group consisting of: the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291 , PTA 292, PTA 294 or PTA 295; a fragment of at least 15 amino acid residues of the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; an amino acid sequence which is at least 95% identical to the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, wherein the percent identity is determined using the ALIGN program of the GCG software package with a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4; and an amino acid sequence which is encoded by a nucleic acid molecule which hybridizes to the nucleic acid molecule consisting of SEQ FD NO:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230 or the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, under conditions of hybridization of 6X SSC at 45°C and washing in 0.2 X SSC, 0.1% SDS at 65°C. The monoclonal antibodies can be human, humanized, chimeric and/or non-human antibodies. The substantially purified antibodies or fragments thereof specifically bind to a signal peptide, a secreted sequence, an extracellular domain, a transmembrane or a cytoplasmic domain cytoplasmic membrane of a polypeptide of the invention. In a particularly prefeπed embodiment, the substantially purified antibodies or fragments thereof, the non-human antibodies or fragments thereof, and/or the monoclonal antibodies or fragments thereof, of the invention specifically bind to a secreted sequence, or alternatively, to an extracellular domain of the amino acid sequence of the invention. Examples of extracellular domains of the invention are shown in SEQ ID NOs:20, 21, 32, 41, 51, 89, 109, 112, 115, 136 or 233.
Any of the antibodies of the invention can be conjugated to a therapeutic moiety or to a detectable substance. Non-limiting examples of detectable substances that can be conjugated to the antibodies of the invention are an enzyme, a prosthetic group, a fluorescent material, a luminescent material, a bioluminescent material, and a radioactive material.
The invention also provides a kit containing an antibody of the invention conjugated to a detectable substance, and instructions for use. Still another aspect of the invention is a pharmaceutical composition comprising an antibody of the invention and a pharmaceutically acceptable carrier. In prefeπed embodiments, the pharmaceutical composition contains an antibody of the invention, a therapeutic moiety, and a pharmaceutically acceptable carrier. Still another aspect of the invention is a method of making an antibody that specifically recognizes TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349, the method comprising immunizing a mammal with a polypeptide. The polypeptide used as an immunogen comprises an amino acid sequence selected from the group consisting of: the amino acid sequence of any one of SEQ ID NOs:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231 , or an amino acid sequence encoded by the cDNA of a clone deposited as ATCC® Accession Number PTA 291, PTA 292, PTA 294 or PTA 295; a fragment of at least 15 amino acid residues of the amino acid sequence of any one of SEQ ID NOs:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, an amino acid sequence which is at least 95% identical to the amino acid sequence of any one of SEQ ID NOs:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, wherein the percent identity is determined using the ALIGN program of the GCG software package with a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4; and an amino acid sequence which is encoded by a nucleic acid molecule which hybridizes to the nucleic acid molecule consisting of any one of SEQ ID NOs:l, 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, or the cDNA of a clone deposited as ATCC® Accession Number PTA 291, PTA 292, PTA 294 or PTA 295, or a complement thereof, under conditions of hybridization of 6X SSC at 45°C and washing in 0.2 X SSC, 0.1% SDS at 65°C. After immunization, a sample is collected from the mammal that contains an antibody that specifically recognizes the immunogen. Preferably, the polypeptide is recombinantly produced using a non-human host cell. Optionally, the antibodies can be further purified from the sample using techniques well known to those of skill in the art. The method can further comprise producing a monoclonal antibody- producing cell from the cells of the mammal. Optionally, antibodies are collected from the antibody-producing cell.
III. Recombinant Expression Vectors and Host Cells
Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding a polypeptide of the invention (or a portion thereof). As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g. , bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors, expression vectors, are capable of directing the expression of genes to which they are operably linked. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors). However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell. This means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein.
The recombinant expression vectors of the invention can be designed for expression of a polypeptide of the invention in prokaryotic (e.g. , E. coli ) or eukaryotic cells (e.g., insect cells (using baculovirus expression vectors), yeast cells or mammalian cells). Suitable host cells are discussed further in Goeddel, supra. Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three puφoses: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and
Johnson (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, MA) and pRET5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) and pET 1 Id (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 60-89). Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid tφ-lac fusion promoter. Target gene expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident λ prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 119-128). Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al. (1992) Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques. In another embodiment, the expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerivisae include pYepSecl (Baldari et al. (1987) EMBOJ. 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-943), pJRY88 (Schultz et al. (1987) Gene 54:113-123), pYES2 (Invitrogen Coφoration, San Diego, CA), and pPicZ (Invitrogen Coφ, San Diego, CA).
Alternatively, the expression vector is a baculovirus expression vector. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf 9 cells) include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39). In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBOJ. 6:187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook et al., supra.
In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1 :268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J. 8:729-733) and immunoglobulins (Banerji et al. (1983) Cell 33:729-740; Queen and Baltimore (1983) Cell 33:741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle (1989) Proc. Natl. Acad. Sci. USA 86:5473-5477), pancreas-specific promoters (Edlund et al. (1985) Science 230:912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Patent No. 4,873,316 and European Application Publication No. 264, 166).
Developmentally-regulated promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the α-fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operably linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to the mRNA encoding a polypeptide of the invention. Regulatory sequences operably linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see Weintraub et al. (Reviews - Trends in Genetics, Vol. 1(1) 1986).
Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. A host cell can be any prokaryotic (e.g., E. coli) or eukaryotic cell (e.g., insect cells, yeast or mammalian cells).
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Prefeπed selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incoφorated the selectable marker gene will survive, while the other cells die).
In another embodiment, the expression characteristics of an endogenous (e.g., TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346 and MANGO 349) nucleic acid within a cell, cell line or microorganism may be modified by inserting a DNA regulatory element heterologous to the endogenous gene of interest into the genome of a cell, stable cell line or cloned microorganism such that the inserted regulatory element is operatively linked with the endogenous gene (e.g., TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes) and controls, modulates or activates the endogenous gene. For example, endogenous TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes which are normally "transcriptionally silent", i.e., a TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes which are normally not expressed, or are expressed only at very low levels in a cell line or microorganism, may be activated by inserting a regulatory element which is capable of promoting the expression of a normally expressed gene product in that cell line or microorganism. Alternatively, transcriptionally silent, endogenous TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes may be activated by insertion of a promiscuous regulatory element that works across cell types.
A heterologous regulatory element may be inserted into a stable cell line or cloned microorganism, such that it is operatively linked with and activates expression of endogenous TANGO 339, TANGO 353, TANGO 358, TANGO, 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 349 genes, using techniques, such as targeted homologous recombination, which are well known to those of skill in the art, and described e.g., in Chappel, U.S. Patent No. 5,272,071; PCT publication No. WO 91/06667, published May 16, 1991.
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce a polypeptide of the invention. Accordingly, the invention further provides methods for producing a polypeptide of the invention using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the polypeptide is produced. In another embodiment, the method further comprises isolating the polypeptide from the medium or the host cell.
The host cells of the invention can also be used to produce nonhuman transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which sequences encoding a polypeptide of the invention have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous sequences encoding a polypeptide of the invention have been introduced into their genome or homologous recombinant animals in which endogenous encoding a polypeptide of the invention sequences have been altered. Such animals are useful for studying the function and/or activity of the polypeptide and for identifying and/or evaluating modulators of polypeptide activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, an "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
A transgenic animal of the invention can be created by introducing nucleic acid encoding a polypeptide of the invention (or a homologue thereof) into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably linked to the transgene to direct expression of the polypeptide of the invention to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009, U.S. Patent No. 4,873,191 and in Hogan, Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986) and Wakayama et al, (1999), Proc. Natl. Acad. Sci. USA, 96:14984-14989. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the transgene in its genome and/or expression of mRNA encoding the transgene in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying the transgene can further be bred to other transgenic animals carrying other transgenes.
To create an homologous recombinant animal, a vector is prepared which contains at least a portion of a gene encoding a polypeptide of the invention into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the gene. In a prefeπed embodiment, the vector is designed such that, upon homologous recombination, the endogenous gene is functionally disrupted (i.e., no longer encodes a functional protein; also refeπed to as a "knock out" vector). Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous protein). In the homologous recombination vector, the altered portion of the gene is flanked at its 5' and 3' ends by additional nucleic acid of the gene to allow for homologous recombination to occur between the exogenous gene carried by the vector and an endogenous gene in an embryonic stem cell. The additional flanking nucleic acid sequences are of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5' and 3' ends) are included in the vector (see, e.g., Thomas and Capecchi (1987) Cell 51 :503 for a description of homologous recombination vectors). The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced gene has homologously recombined with the endogenous gene are selected (see, e.g., Li et al. (1992) Cell 69:915). The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see, e.g., Bradley in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed. (IRL, Oxford, 1987) pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley (1991) Current Opinion in Bio/Technology 2:823-829 and in PCT Publication Nos. WO 90/11354, WO 91/01140, WO 92/0968, and WO 93/04169. In another embodiment, transgenic non-human animals can be produced which contain selected systems which allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage PI . For a description of the cre/loxP recombinase system, see, e.g., Lakso et al. (1992) Proc. Natl. Acad. Sci. USA 89:6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut et al. (1997) Nature 385:810-813 and PCT Publication NOS. WO 97/07668 and WO 97/07669.
FV. Pharmaceutical Compositions
The nucleic acid molecules, polypeptides, and antibodies (also refeπed to herein as "active compounds") of the invention can be incoφorated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
The invention includes methods for preparing pharmaceutical compositions for modulating the expression or activity of a polypeptide or nucleic acid of the invention. Such methods comprise formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid of the invention. Such compositions can further include additional active agents. Thus, the invention further includes methods for preparing a pharmaceutical composition by formulating a pharmaceutically acceptable carrier with an agent which modulates expression or activity of a polypeptide or nucleic acid of the invention and one or more additional active compounds. A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF; Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the prefeπed methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. For administration by inhalation, the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
The compounds can also be prepared in the form of suppositories (e.g. , with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Coφoration and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
For antibodies, the prefeπed dosage is 0.1 mg/kg to 100 mg/kg of body weight (generally 10 mg/kg to 20 mg/kg). If the antibody is to act in the brain, a dosage of 50 mg/kg to 100 mg/kg is usually appropriate. Generally, partially human antibodies and fully human antibodies have a longer half-life within the human body than other antibodies. Accordingly, lower dosages and less frequent administration is often possible. Modifications such as lipidation can be used to stabilize antibodies and to enhance uptake and tissue penetration (e.g., into the brain). A method for lipidation of antibodies is described by Cruikshank et al. ((1997) J. Acquired Immune Deficiency Syndromes and Human Retrovirology 14:193).
Antibodies or antibodies conjugated to therapeutic moieties can be administered to an individual alone or in combination with cytotoxic factor(s), chemotherapeutic drug(s), and/or cytokine(s). If the latter, preferably, the antibodies are administered first and the cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) are administered thereafter within 24 hours. The antibodies and cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) can be administered by multiple cycles depending upon the clinical response of the patient. Further, the antibodies and cytotoxic factor(s), chemotherapeutic drug(s) and/or cytokine(s) can be administered by the same or separate routes, for example, by intravenous, intranasal or intramuscular administration. Cytotoxic factors include, but are not limited to, TNF-α, TNF-β, IL-1, IFN-γ and IL-2. Chemotherapeutic drugs include, but are not limited to, 5-fluorouracil (5FU), vinblastine, actinomycin D, etoposide, cisplatin, methotrexate and doxorubicin. Cytokines include, but are not limited to, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10 and IL-12. As defined herein, a therapeutically effective amount of protein or polypeptide (i.e., an effective dosage) ranges from about 0.001 to 30 mg/kg body weight, preferably about 0.01 to 25 mg/kg body weight, more preferably about 0.1 to 20 mg/kg body weight, and even more preferably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a protein, polypeptide, or antibody can include a single treatment or, preferably, can include a series of treatments. In a prefeπed example, a subject is treated with antibody, protein, or polypeptide in the range of between about 0.1 to 20 mg/kg body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks. It will also be appreciated that the effective dosage of antibody, protein, or polypeptide used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of diagnostic assays as described herein.
The present invention encompasses agents which modulate expression or activity. An agent may, for example, be a small molecule. For example, such small molecules include, but are not limited to, peptides, peptidomimetics, amino acids, amino acid analogs, polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic or inorganic compounds (i.e,. including heteroorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds. It is understood that appropriate doses of small molecule agents depends upon a number of factors within the ken of the ordinarily skilled physician, veterinarian, or researcher. The dose(s) of the small molecule will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the small molecule to have upon the nucleic acid or polypeptide of the invention. Exemplary doses include milligram or microgram amounts of the small molecule per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram. It is furthermore understood that appropriate doses of a small molecule depend upon the potency of the small molecule with respect to the expression or activity to be modulated. Such appropriate doses may be determined using the assays described herein. When one or more of these small molecules is to be administered to an animal (e.g. , a human) in order to modulate expression or activity of a polypeptide or nucleic acid of the invention, a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (U.S. Patent 5,328,470) or by stereotactic injection (see, e.g., Chen et al. (1994) Proc. Nαtl. Acαd. Sci. USA 91 :3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g. , retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
V. Uses and Methods of the Invention
The nucleic acid molecules, proteins, protein homologs, and antibodies described herein can be used in one or more of the following methods: a) screening assays; b) detection assays (e.g., chromosomal mapping, tissue typing, forensic biology); c) predictive medicine (e.g., diagnostic assays, prognostic assays, monitoring clinical trials, and pharmacogenomics); and d) methods of treatment (e.g., therapeutic and prophylactic). For example, polypeptides of the invention can to used to (i) modulate cellular proliferation; (ii) modulate cellular differentiation; and/or (iii) modulate cellular adhesion. The isolated nucleic acid molecules of the invention can be used to express proteins (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect mRNA (e.g., in a biological sample) or a genetic lesion, and to modulate activity of a polypeptide of the invention. In addition, the polypeptides of the invention can be used to screen drugs or compounds which modulate activity or expression of a polypeptide of the invention as well as to treat disorders characterized by insufficient or excessive production of a protein of the invention or production of a form of a protein of the invention which has decreased or abeπant activity compared to the wild type protein. In addition, the antibodies of the invention can be used to detect and isolate a protein of the and modulate activity of a protein of the invention.
This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
A. Screening Assays
The invention provides a method (also refeπed to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) which bind to polypeptide of the invention or have a stimulatory or inhibitory effect on, for example, expression or activity of a polypeptide of the invention.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of a polypeptide of the invention or biologically active portion thereof. The test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam (1997) Anticancer DrugDes. 12:145).
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al. (1993) Proc. Natl. Acad. Sci. USA 90:6909; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91 :11422; Zuckermann et al. (1994). J. Med. Chem. 37:2678; Cho et al. (1993) Science 261:1303; Caπell et al. (1994) Angew. Chem. Int. Ed. Engl 33:2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl 33:2061; and Gallop et al. (1994) J. Med. Chem. 37:1233.
Libraries of compounds may be presented in solution (e.g., Houghten (1992) Bio/Techniques 13:412-421), or on beads (Lam (1991) Nature 354:82-84), chips (Fodor (1993) Nature 364:555-556), bacteria (U.S. Patent No. 5,223,409), spores (Patent NOS. 5,571,698; 5,403,484; and 5,223,409), plasmids (Cull et al. (1992) Proc. Natl. Acad. Sci. USA 89:1865-1869) or phage (Scott and Smith (1990) Science 249:386-390; Devlin (1990) Science 249:404-406; Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87:6378-6382; and Felici (1991) J. Mol Biol. 222:301-310). In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to the polypeptide determined. The cell, for example, can be a yeast cell or a cell of mammalian origin. Determining the ability of the test compound to bind to the polypeptide can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the polypeptide or biologically active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 125^ 3 s, 14 or -1H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting. Alternatively, test compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. In a prefeπed embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the test compound to preferentially bind to the polypeptide or a biologically active portion thereof as compared to the known compound.
In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of a polypeptide of the invention, or a biologically active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the polypeptide or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of the polypeptide or a biologically active portion thereof can be accomplished, for example, by determining the ability of the polypeptide protein to bind to or interact with a target molecule.
Determining the ability of a polypeptide of the invention to bind to or interact with a target molecule can be accomplished by one of the methods described above for determining direct binding. As used herein, a "target molecule" is a molecule with which a selected polypeptide (e.g., a polypeptide of the invention) binds or interacts with in nature, for example, a molecule on the surface of a cell which expresses the selected protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule. A target molecule can be a polypeptide of the invention or some other polypeptide or protein. For example, a target molecule can be a component of a signal transduction pathway which facilitates transduction of an extracellular signal (e.g., a signal generated by binding of a compound to a polypeptide of the invention) through the cell membrane and into the cell or a second intercellular protein which has catalytic activity or a protein which facilitates the association of downstream signaling molecules with a polypeptide of the invention. Determining the ability of a polypeptide of the invention to bind to or interact with a target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (e.g. , intracellular
Ca +, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target on an appropriate substrate, detecting the induction of a reporter gene (e.g., a regulatory element that is responsive to a polypeptide of the invention operably linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cellular differentiation, or cell proliferation.
In yet another embodiment, an assay of the present invention is a cell- free assay comprising contacting a polypeptide of the invention or biologically active portion thereof with a test compound and determining the ability of the test compound to bind to the polypeptide or biologically active portion thereof. Binding of the test compound to the polypeptide can be determined either directly or indirectly as described above. In a prefeπed embodiment, the assay includes contacting the polypeptide of the invention or biologically active portion thereof with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the test compound to preferentially bind to the polypeptide or biologically active portion thereof as compared to the known compound.
In another embodiment, an assay is a cell-free assay comprising contacting a polypeptide of the invention or biologically active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the polypeptide or biologically active portion thereof. Determining the ability of the test compound to modulate the activity of the polypeptide can be accomplished, for example, by determining the ability of the polypeptide to bind to a target molecule by one of the methods described above for determining direct binding. En an alternative embodiment, determining the ability of the test compound to modulate the activity of the polypeptide can be accomplished by determining the ability of the polypeptide of the invention to further modulate the target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as previously described. In yet another embodiment, the cell-free assay comprises contacting a polypeptide of the invention or biologically active portion thereof with a known compound which binds the polypeptide to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the polypeptide, wherein determining the ability of the test compound to interact with the polypeptide comprises determining the ability of the polypeptide to preferentially bind to or modulate the activity of a target molecule.
The cell- free assays of the present invention are amenable to use of both a soluble form or the membrane-bound form of a polypeptide of the invention. In the case of cell- free assays comprising the membrane-bound form of the polypeptide, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of the polypeptide is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-octylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton X-100, Triton X-l 14, Thesit, Isotridecypoly(ethylene glycol ether)n, 3-[(3-cholamidopropyl)dimethylamminio]-l-propane sulfonate (CHAPS),
3-[(3-cholamidopropyl)dimethylamminio]-2-hydroxy-l-propane sulfonate (CHAPSO), or N-dodecyl=N,N-dimethyl-3-ammonio-l -propane sulfonate.
In more than one embodiment of the above assay methods of the present invention, it may be desirable to immobilize either the polypeptide of the invention or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to the polypeptide, or interaction of the polypeptide with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix. For example, glutathione-S-transferase fusion proteins or glutathione-S-transferase fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical; St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or A polypeptide of the invention, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtitre plate wells are washed to remove any unbound components and complex formation is measured either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of binding or activity of the polypeptide of the invention can be determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either the polypeptide of the invention or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated polypeptide of the invention or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g. , biotinylation kit, Pierce Chemicals; Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with the polypeptide of the invention or target molecules but which do not interfere with binding of the polypeptide of the invention to its target molecule can be derivatized to the wells of the plate, and unbound target or polypeptide of the invention trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the polypeptide of the invention or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the polypeptide of the invention or target molecule.
In another embodiment, modulators of expression of a polypeptide of the invention are identified in a method in which a cell is contacted with a candidate compound and the expression of the selected mRNA or protein (i.e., the mRNA or protein coπesponding to a polypeptide or nucleic acid of the invention) in the cell is determined. The level of expression of the selected mRNA or protein in the presence of the candidate compound is compared to the level of expression of the selected mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of expression of the polypeptide of the invention based on this comparison. For example, when expression of the selected mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of the selected mRNA or protein expression. Alternatively, when expression of the selected mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of the selected mRNA or protein expression. The level of the selected mRNA or protein expression in the cells can be determined by methods described herein.
In yet another aspect of the invention, a polypeptide of the inventions can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol Chem. 268:12046-12054; Bartel et al. (1993) Bio/Techniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and PCT Publication No. WO 94/10300), to identify other proteins, which bind to or interact with the polypeptide of the invention and modulate activity of the polypeptide of the invention. Such binding proteins are also likely to be involved in the propagation of signals by the polypeptide of the inventions as, for example, upstream or downstream elements of a signaling pathway involving the polypeptide of the invention.
This invention further pertains to novel agents identified by the above-described screening assays and uses thereof for treatments as described herein.
B. Detection Assays
Portions or fragments of the cDNA sequences identified herein (and the coπesponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. These applications are described in the subsections below.
1. Chromosome Mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. Accordingly, nucleic acid molecules described herein or fragments thereof, can be used to map the location of the coπesponding genes on a chromosome. The mapping of the sequences to chromosomes is an important first step in coπelating these sequences with genes associated with disease. Briefly, genes can be mapped to chromosomes by preparing PCR primers
(preferably 15-25 bp in length) from the sequence of a gene of the invention. Computer analysis of the sequence of a gene of the invention can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene coπesponding to the gene sequences will yield an amplified fragment. For a review of this technique, see D'Eustachio et al. ((1983) Science 220:919-924).
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the nucleic acid sequences of the invention to design oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes. Other mapping strategies which can similarly be used to map a gene to its chromosome include in situ hybridization (described in Fan et al. (1990) Proc. Natl. Acad. Sci. USA 87:6223-27), pre-screening with labeled flow-sorted chromosomes (CITE), and pre-selection by hybridization to chromosome specific cDNA libraries. Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. For a review of this technique, see Verma et al., (Human Chromosomes: A Manual of Basic Techniques (Pergamon Press, New York, 1988)). Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents coπesponding to noncoding regions of the genes actually are prefeπed for mapping puφoses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be coπelated with genetic map data. (Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland et al. (1987) Nαtwre 325:783-787.
Moreover, differences in the DΝA sequences between individuals affected and unaffected with a disease associated with a gene of the invention can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DΝA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymoφhisms.
Furthermore, the nucleic acid sequences disclosed herein can be used to perform searches against "mapping databases", e.g., BLAST-type search, such that the chromosome position of the gene is identified by sequence homology or identity with known sequence fragments which have been mapped to chromosomes.
A polypeptide and fragments and sequences thereof and antibodies specific thereto can be used to map the location of the gene encoding the polypeptide on a chromosome. This mapping can be carried out by specifically detecting the presence of the polypeptide in members of a panel of somatic cell hybrids between cells of a first species of animal from which the protein originates and cells from a second species of animal and then determining which somatic cell hybrid(s) expresses the polypeptide and noting the chromosome(s) from the first species of animal that it contains. For examples of this technique, see Pajunen et al. (1988) Cytogenet. Cell Genet. 47:31-41 and Van Keuren et al. (1986) Hum. Genet. 74:34-40. Alternatively, the presence of the polypeptide in the somatic cell hybrids can be determined by assaying an activity or property of the polypeptide, for example, enzymatic activity, as described in Bordelon-Riser et al. ( 919) Somatic Cell Genetics 5:597-613 and Owerbach et al. (1978; Proc. Natl. Acad. Sci. USA 75:5640-5644.
2. Tissue Typing
The nucleic acid sequences of the present invention can also be used to identify individuals from minute biological samples. The United States military, for example, is considering the use of restriction fragment length polymoφhism (RFLP) for identification of its personnel. In this technique, an individual's genomic DΝA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. This method does not suffer from the cuπent limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult. The sequences of the present invention are useful as additional DNA markers for RFLP (described in U.S. Patent 5,272,057).
Furthermore, the sequences of the present invention can be used to provide an alternative technique which determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the nucleic acid sequences described herein can be used to prepare two PCR primers from the 5' and 3' ends of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it. Panels of coπesponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the present invention can be used to obtain such identification sequences from individuals and from tissue. The nucleic acid sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification puφoses. Because greater numbers of polymoφhisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128 can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers which each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128 are used, a more appropriate number of primers for positive individual identification would be 500-2,000.
If a panel of reagents from the nucleic acid sequences described herein is used to generate a unique identification database for an individual, those same reagents can later be used to identify tissue from that individual. Using the unique identification database, positive identification of the individual, living or dead, can be made from extremely small tissue samples.
3. Use of Partial Gene Sequences in Forensic Biology
DNA-based identification techniques can also be used in forensic biology. Forensic biology is a scientific field employing genetic typing of biological evidence found at a crime scene as a means for positively identifying, for example, a peφetrator of a crime. To make such an identification, PCR technology can be used to amplify DNA sequences taken from very small biological samples such as tissues, e.g. , hair or skin, or body fluids, e.g. , blood, saliva, or semen found at a crime scene. The amplified sequence can then be compared to a standard, thereby allowing identification of the origin of the biological sample. The sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another "identification marker" (i.e. another DNA sequence that is unique to a particular individual). As mentioned above, actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments. Sequences targeted to noncoding regions are particularly appropriate for this use as greater numbers of polymoφhisms occur in the noncoding regions, making it easier to differentiate individuals using this technique. Examples of polynucleotide reagents include the nucleic acid sequences of the invention or portions thereof, e.g., fragments derived from noncoding regions having a length of at least 20 or 30 bases.
The nucleic acid sequences described herein can further be used to provide polynucleotide reagents, e.g., labeled or labelable probes which can be used in, for example, an in situ hybridization technique, to identify a specific tissue, e.g., brain tissue. This can be very useful in cases where a forensic pathologist is presented with a tissue of unknown origin. Panels of such probes can be used to identify tissue by species and/or by organ type.
C. Predictive Medicine
The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trails are used for prognostic (predictive) puφoses to thereby treat an individual prophylactically. Accordingly, one aspect of the present invention relates to diagnostic assays for determining expression of a polypeptide or nucleic acid of the invention and/or activity of a polypeptide of the invention, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with abeπant expression or activity of a polypeptide of the invention. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with abeπant expression or activity of a polypeptide of the invention. For example, mutations in a gene of the invention can be assayed in a biological sample. Such assays can be used for prognostic or predictive puφose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with abeπant expression or activity of a polypeptide of the invention.
Another aspect of the invention provides methods for expression of a nucleic acid or polypeptide of the invention or activity of a polypeptide of the invention in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (refeπed to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent). Yet another aspect of the invention pertains to monitoring the influence of agents
(e.g., drugs or other compounds) on the expression or activity of a polypeptide of the invention in clinical trials. These and other agents are described in further detail in the following sections.
1. Diagnostic Assays
An exemplary method for detecting the presence or absence of a polypeptide or nucleic acid of the invention in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting a polypeptide or nucleic acid (e.g., mRNA, genomic DNA) of the invention such that the presence of a polypeptide or nucleic acid of the invention is detected in the biological sample. A prefeπed agent for detecting mRNA or genomic DNA encoding a polypeptide of the invention is a labeled nucleic acid probe capable of hybridizing to mRNA or genomic DNA encoding a polypeptide of the invention. The nucleic acid probe can be, for example, a full-length cDNA, such as the nucleic acid of SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123 or 128, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to a mRNA or genomic DNA encoding a polypeptide of the invention. Other suitable probes for use in the diagnostic assays of the invention are described herein. A prefeπed agent for detecting a polypeptide of the invention is an antibody capable of binding to a polypeptide of the invention, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of a polypeptide of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence. In vitro techniques for detection of genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of a polypeptide of the invention include introducing into a subject a labeled antibody directed against the polypeptide. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A prefeπed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting a polypeptide of the invention or mRNA or genomic DNA encoding a polypeptide of the invention, such that the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide is detected in the biological sample, and comparing the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide in the control sample with the presence of the polypeptide or mRNA or genomic DNA encoding the polypeptide in the test sample.
The invention also encompasses kits for detecting the presence of a polypeptide or nucleic acid of the invention in a biological sample (a test sample). Such kits can be used to determine if a subject is suffering from or is at increased risk of developing a disorder associated with abeπant expression of a polypeptide of the invention as discussed, for example, in sections above relating to uses of the sequences of the invention.
For example, kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, neurological disorders, eye disorders and embryonic disorders, which are associated with abeπant TANGO 339 expression. En another example, kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g. , rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)). Disorders associated with TANGO 339 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g. , B cell cancers stimulated by TNF), which are associated with abeπant TANGO 353 expression. In another example, kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders (e.g., thymic disorders) and embryonic disorders, which are associated with abeπant TANGO 358 expression. In another example, kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders, e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)). Disorders associated with decreased [x] activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF), which are associated with abeπant TANGO 368 or TANGO 369 expression. In another example, kits can be used to determine if a subject is suffering from or is at increased risk of disorders such as immunological disorders (e.g., platelet disorders) endothelial disorders and embryonic disorders, which are associated with aberrant TANGO 402 expression. In another example, kits can be used to determine if a subject is suffering from or is at risk for brain-related disorders such as cerebral edema, hydrocephalus, brain herniations, iatrogenic disease (due to, e.g., infection, toxins, or drugs), inflammations (e.g., bacterial and viral meningitis, encephalitis, and cerebral toxoplasmosis), cerebrovascular diseases (e.g., hypoxia, ischemia, and infarction, intracranial hemoπhage and vascular malformations, and hypertensive encephalopathy), and tumors (e.g., neuroglial tumors, neuronal tumors, tumors of pineal cells, meningeal tumors, primary and secondary lymphomas, intracranial tumors, and medulloblastoma), and to treat injury or trauma to the brain, which are associated with abeπant MANGO 346 or MANGO 349 expression. In still another example, kits can be used to determine if a subject is suffering from or is at risk for prostate-related disorders, (e.g. prostate cancer, prostatitis, benign prostatic hypertrophy, benign prostatic hypeφlasia and atypical prostatic stromal lesions) which can be associated with abeπant TANGO 365 or TANGO 383 expression. In another example, kits can be used to determine if a subject is suffering from or is at risk for endocrine-related disorders, e.g., whole animal homeostasis, appetite-related disorders, which are associated with abeπant TANGO 393 expression. The kit, for example, may comprise a labeled compound or agent capable of detecting the polypeptide or mRNA encoding the polypeptide in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample (e.g. , an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide). Kits may also include instructions for observing that the tested subject is suffering from or is at risk of developing a disorder associated with abeπant expression of the polypeptide if the amount of the polypeptide or mRNA encoding the polypeptide is above or below a normal level.
For antibody-based kits, the kit may comprise, for example: (1) a first antibody (e.g., attached to a solid support) which binds to a polypeptide of the invention; and, optionally, (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable agent.
For oligonucleotide-based kits, the kit may comprise, for example: (1) an oligonucleotide, e.g., a detectably labeled oligonucleotide, which hybridizes to a nucleic acid sequence encoding a polypeptide of the invention or (2) a pair of primers useful for amplifying a nucleic acid molecule encoding a polypeptide of the invention. The kit may also comprise, e.g., a buffering agent, a preservative, or a protein stabilizing agent. The kit may also comprise components necessary for detecting the detectable agent (e.g., an enzyme or a substrate). The kit may also contain a control sample or a series of control samples which can be assayed and compared to the test sample contained. Each component of the kit is usually enclosed within an individual container and all of the various containers are within a single package along with instructions for observing whether the tested subject is suffering from or is at risk of developing a disorder associated with abeπant expression of the polypeptide.
2. Prognostic Assays
The methods described herein can furthermore be utilized as diagnostic or prognostic assays to identify subjects having or at risk of developing a disease or disorder associated with abeπant expression or activity of a polypeptide of the invention. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with abeπant expression or activity of a polypeptide of the invention, such as a proliferative disorder, e.g., psoriasis or cancer, or an angiogenic disorder. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing such a disease or disorder. Thus, the present invention provides a method in which a test sample is obtained from a subject and a polypeptide or nucleic acid (e.g., mRNA, genomic DNA) of the invention is detected, wherein the presence of the polypeptide or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with abeπant expression or activity of the polypeptide. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
The prognostic assays described herein, for example, can be used to identify a subject having or at risk of developing disorders such as disorders discussed, for example, in sections above relating to uses of the sequences of the invention. For example, prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders, neurological disorders and embryonic disorders, which are associated with abeπant TANGO 339 expression. En another example, prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AEDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)). Disorders associated with TANGO 339 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g. , B cell cancers stimulated by TNF), which are associated with abeπant TANGO 353 expression. In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders (e.g., thymic disorders) and embryonic disorders, which are associated with abeπant TANGO 358 expression. In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders e.g., autoimmune disorders (e.g., arthritis, graft rejection (e.g., allograft rejection), T cell disorders (e.g., AIDS)) and inflammatory disorders (e.g., bacterial infection, psoriasis, septicemia, cerebral malaria, inflammatory bowel disease, arthritis (e.g., rheumatoid arthritis, osteoarthritis), and allergic inflammatory disorders (e.g., asthma, psoriasis)). Disorders associated with TANGO 358 activity also include apoptotic disorders (e.g., rheumatoid arthritis, systemic lupus erythematosus, insulin- dependent diabetes mellitus), cytotoxic disorders, septic shock, cachexia, and proliferative disorders (e.g., B cell cancers stimulated by TNF), which are associated with abeπant TANGO 368 or TANGO 369 expression. In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing immunological disorders (e.g., platelet disorders), endothelial disorders and embryonic disorders, which are associated with abeπant TANGO 402 expression. In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing brain-related disorders such as cerebral edema, hydrocephalus, brain herniations, iatrogenic disease (due to, e.g., infection, toxins, or drugs), inflammations (e.g., bacterial and viral meningitis, encephalitis, and cerebral toxoplasmosis), cerebrovascular diseases (e.g. , hypoxia, ischemia, and infarction, intracranial hemoπhage and vascular malformations, and hypertensive encephalopathy), and tumors (e.g., neuroglial tumors, neuronal tumors, tumors of pineal cells, meningeal tumors, primary and secondary lymphomas, intracranial tumors, and meduUoblastoma), and to treat injury or trauma to the brain, which are associated with abeπant MANGO 346 or MANGO 349 expression. In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing prostate-related disorders (e.g, prostate cancer, prostatis, benign prostatic hypertrophy, benign prostatic hypeφlasmia and atypical prostatic stromal lesions). In another example, prognostic assays described herein can be used to identify a subject having or at risk of developing endocrine-related disorders (e.g., animal homeostasis and appetite-related disorders), which are associated with abeπant TANGO 393 expression.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with abeπant expression or activity of a polypeptide of the invention. For example, such methods can be used to determine whether a subject can be effectively treated with a specific agent or class of agents (e.g., agents of a type which decrease activity of the polypeptide). Thus, the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abeπant expression or activity of a polypeptide of the invention in which a test sample is obtained and the polypeptide or nucleic acid encoding the polypeptide is detected (e.g., wherein the presence of the polypeptide or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with abeπant expression or activity of the polypeptide). The methods of the invention can also be used to detect genetic lesions or mutations in a gene of the invention, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized abeπant expression or activity of a polypeptide of the invention. In prefeπed embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion or mutation characterized by at least one of an alteration affecting the integrity of a gene encoding the polypeptide of the invention, or the mis-expression of the gene encoding the polypeptide of the invention. For example, such genetic lesions or mutations can be detected by ascertaining the existence of at least one of: 1) a deletion of one or more nucleotides from the gene; 2) an addition of one or more nucleotides to the gene; 3) a substitution of one or more nucleotides of the gene; 4) a chromosomal reaπangement of the gene; 5) an alteration in the level of a messenger RNA transcript of the gene; 6) an abeπant modification of the gene, such as of the methylation pattern of the genomic DNA; 7) the presence of a non-wild type splicing pattern of a messenger RNA transcript of the gene; 8) a non-wild type level of a the protein encoded by the gene; 9) an allelic loss of the gene; and 10) an inappropriate post-translational modification of the protein encoded by the gene. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in a gene.
En certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and
4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1994) Proc. Natl. Acad. Sci. USA 91:360-364), the latter of which can be particularly useful for detecting point mutations in a gene (see, e.g., Abravaya et al. (1995) Nucleic Acids Res. 23:675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to the selected gene under conditions such that hybridization and amplification of the gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (Guatelli et al. (1990) Proc. Natl Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh, et al. (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al. (1988) Bio/Technology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
In an alternative embodiment, mutations in a selected gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,498,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In other embodiments, genetic mutations can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density aπays containing hundreds or thousands of oligonucleotides probes (Cronin et al. (1996) Human Mutation 7:244-255; Kozal et al. (1996) Nature Medicine 2:753-759). For example, genetic mutations can be identified in two-dimensional arrays containing light-generated DNA probes as described in Cronin et al., supra. Briefly, a first hybridization aπay of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear aπays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization aπay that allows the characterization of specific mutations by using smaller, specialized probe aπays complementary to all variants or mutations detected. Each mutation aπay is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the selected gene and detect mutations by comparing the sequence of the sample nucleic acids with the coπesponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995) Bio/Techniques 19:448), including sequencing by mass spectrometry ( see, e.g., PCT Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36:127-162; and Griffin et al. (1993) Appl Biochem. Biotechnol. 38:147-159). Other methods for detecting mutations in a selected gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242). In general, the technique of "mismatch cleavage" entails providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild-type sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. RNA/DNA duplexes can be treated with RNase to digest mismatched regions, and DNA/DNA hybrids can be treated with S 1 nuclease to digest mismatched regions.
In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton et al. (1988) Proc. Natl. Acad. Sci. USA 85:4397; Saleeba et al. (1992) Methods Enzymol. 217:286-295. In a prefeπed embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662). According to an exemplary embodiment, a probe based on a selected sequence, e.g. , a wild-type sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in genes. For example, single strand conformation polymoφhism
(SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (Orita et al. (1989) Proc. Natl. Acad. Sci. USA 86:2766; see also Cotton (1993) Mutat. Res. 285:125-144; Hayashi (1992) Genet. Anal. Tech. Appl. 9:73-79). Single-stranded DNA fragments of sample and control nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, and the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a prefeπed embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet. 7:5).
In yet another embodiment, the movement of mutant or wild- type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. (1985) Nature 313:495). When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a 'GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys. Chem. 265:12753). Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al. (1989) Proc. Natl. Acad. Sci. USA 86:6230). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
Alternatively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention.
Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent or reduce polymerase extension (Prossner (1993) Tibtech 11 :238). In addition, it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al. (1992) Mol. Cell Probes 6:1). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci. USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving a gene encoding a polypeptide of the invention. Furthermore, any cell type or tissue, e.g., preferably peripheral blood leukocytes, in which the polypeptide of the invention is expressed may be utilized in the prognostic assays described herein.
3. Pharmaco genomics
Agents, or modulators which have a stimulatory or inhibitory effect on activity or expression of a polypeptide of the invention as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders associated with abeπant activity of the polypeptide. In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of a polypeptide of the invention, expression of a nucleic acid of the invention, or mutation content of a gene of the invention in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See, e.g., Linder (1997) Clin. Chem. 43(2):254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body are refeπed to as "altered drug action." Genetic conditions transmitted as single factors altering the way the body acts on drugs are refeπed to as "altered drug metabolism". These pharmacogenetic conditions can occur either as rare defects or as polymoφhisms. For example, glucose-6-phosphate dehydrogenase deficiency (G6PD) is a common inherited enzymopathy in which the main clinical complication is haemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitro urans) and consumption of fava beans.
As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymoφhisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymoφhisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymoφhic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, a PM will show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite moφhine. The other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification. Thus, the activity of a polypeptide of the invention, expression of a nucleic acid encoding the polypeptide, or mutation content of a gene encoding the polypeptide in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymoφhic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with a modulator of activity or expression of the polypeptide, such as a modulator identified by one of the exemplary screening assays described herein.
4. Monitoring of Effects During Clinical Trials
Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of a polypeptide of the invention (e.g., the ability to modulate abeπant cell proliferation chemotaxis, and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent, as determined by a screening assay as described herein, to increase gene expression, protein levels or protein activity, can be monitored in clinical trials of subjects exhibiting decreased gene expression, protein levels, or protein activity. Alternatively, the effectiveness of an agent, as determined by a screening assay, to decrease gene expression, protein levels or protein activity, can be monitored in clinical trials of subjects exhibiting increased gene expression, protein levels, or protein activity. In such clinical trials, expression or activity of a polypeptide of the invention and preferably, that of other polypeptide that have been implicated in for example, a cellular proliferation disorder, can be used as a marker of the immune responsiveness of a particular cell.
For example, and not by way of limitation, genes, including those of the invention, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) which modulates activity or expression of a polypeptide of the invention (e.g., as identified in a screening assay described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of a gene of the invention and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of a gene of the invention or other genes. In this way, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
En a preferred embodiment, the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of the polypeptide or nucleic acid of the invention in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level the of the polypeptide or nucleic acid of the invention in the post-administration samples; (v) comparing the level of the polypeptide or nucleic acid of the invention in the pre-administration sample with the level of the polypeptide or nucleic acid of the invention in the post-administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of the polypeptide to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of the polypeptide to lower levels than detected, i.e., to decrease the effectiveness of the agent.
C. Methods of Treatment
The present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with abeπant expression or activity of a polypeptide of the invention. For example, disorders characterized by abeπant expression or activity of the polypeptides of the invention include proliferative disorders such as cancer.
1. Prophylactic Methods
In one aspect, the invention provides a method for preventing in a subject, a disease or condition associated with an abeπant expression or activity of a polypeptide of the invention, by administering to the subject an agent which modulates expression or at least one activity of the polypeptide. Subjects at risk for a disease which is caused or contributed to by abeπant expression or activity of a polypeptide of the invention can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the abeπancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending on the type of abeπancy, for example, an agonist or antagonist agent can be used for treating the subject. For example, an antagonist of a TANGO 339 protein may be used to treat an immunological disorder. The appropriate agent can be determined based on screening assays described herein.
2. Therapeutic Methods
Another aspect of the invention pertains to methods of modulating expression or activity of a polypeptide of the invention for therapeutic puφoses. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of the polypeptide. An agent that modulates activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of the polypeptide, a peptide, a peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more of the biological activities of the polypeptide. Examples of such stimulatory agents include the active polypeptide of the invention and a nucleic acid molecule encoding the polypeptide of the invention that has been introduced into the cell. In another embodiment, the agent inhibits one or more of the biological activities of the polypeptide of the invention. Examples of such inhibitory agents include antisense nucleic acid molecules and antibodies. These modulatory methods can be performed in vitro (e.g. , by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by abeπant expression or activity of a polypeptide of the invention. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g. , upregulates or downregulates) expression or activity. In another embodiment, the method involves administering a polypeptide of the invention or a nucleic acid molecule of the invention as therapy to compensate for reduced or abeπant expression or activity of the polypeptide.
Stimulation of activity is desirable in situations in which activity or expression is abnormally low or downregulated and/or in which increased activity is likely to have a beneficial effect. Conversely, inhibition of activity is desirable in situations in which activity or expression is abnormally high or upregulated and/or in which decreased activity is likely to have a beneficial effect.
This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are hereby incoφorated by reference.
Deposit of Clones
Clones containing cDNA molecules encoding TANGO 339, TANGO 353, and TANGO 358 (clones EpT339, EpT353, and EpT358, respectively), were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-292, as part of a composite deposit representing a mixture of three strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
To distinguish the strains and isolate a strain harboring a particular cDNA clone, an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOOμg/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure. Next, a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8%o agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows: TANGO 339 (EpT339) 2.7 kb TANGO 353 (EpT353) 1.3 kb TANGO 358 (EpT358) 1.6 kb
The identity of the strains can be infeπed from the fragments liberated.
Clones containing cDNA molecules encoding MANGO 346, TANGO 365, and TANGO 368 (clones EpM346, EpT365, and EpT368, respectively), were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-291, as part of a composite deposit representing a mixture of three strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
To distinguish the strains and isolate a strain harboring a particular cDNA clone, an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOOμg/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure. Next, a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows:
MANGO 346 (EpM346): 1.2 kb
TANGO 365 (EpT365): 1.4 kb TANGO 368 (EpT368): 1.0 kb
The identity of the strains can be infeπed from the fragments liberated.
Clones containing cDNA molecules encoding MANGO 349, TANGO 369, TANGO 383, and TANGO 393 (clones EpM349, EpT369, EpT383, and EpT393, respectively), were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-295, as part of a composite deposit representing a mixture of four strains, each carrying one recombinant plasmid harboring a particular cDNA clone.
To distinguish the strains and isolate a strain harboring a particular cDNA clone, an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with lOOμg/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure. Next, a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows:
MANGO 349 (EpM349): 3.7 kb TANGO 369 (EpT369): 1.1 kb TANGO 383 (EpT383): 1.4 kb TANGO 393 (EpT393): 1.8 kb
The identity of the strains can be infeπed from the fragments liberated.
Clones containing cDNA molecules encoding TANGO 402 (clone EpT402), were deposited with the American Type Culture Collection (Manassas, VA) on June 29, 1999 as Accession Number PTA-294, as part of a composite deposit representing a mixture of two strains, each carrying one recombinant plasmid harboring a particular cDNA clone. To distinguish the strains and isolate a strain harboring a particular cDNA clone, an aliquot of the mixture can be streaked out to single colonies on nutrient medium (e.g., LB plates) supplemented with 100 μg/ml ampicillin, single colonies grown, and then plasmid DNA extracted using a standard minipreparation procedure. Next, a sample of the DNA minipreparation can be digested with a combination of the restriction enzymes Sal I and Not I and the resultant products resolved on a 0.8% agarose gel using standard DNA electrophoresis conditions. The digest liberates fragments as follows:
TANGO 402 (EpT402): 1.4 kb
The identity of the strain containing TANGO 402 can be infeπed from the liberation of a fragment of the above-identified size.
All publications, patents and patent applications mentioned in this specification are herein incoφorated by reference into the specification to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incoφorated herein by reference.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. International Application No: PCT/ /
MICROORGANISMS
Optional Sheet in connection with the microorganism referred to on pages , lines of the description
A. IDENTIFICATION OF DEPOSIT '
Further deposits are identified on an additional sheet
Name of depositary institution ' American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Blvd Manassas, VA 201 10-2209 US
Date of deposit ' June 29, 1999 Accession Number ' PTA-291
B. ADDITIONAL INDICATIONS ' (leave blank if not applicable) This information is continued on a separate attached sheet
C DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE ' of<ι».d~∞«...i]<up».su
D. SEPARATE FURNISHING OF INDICATIONS ' (leave blank if not applicable)
The indications listed below will be submitted to the International Bureau later ' (Specify the general nature of the indications e g , "Accession Number of Deposit")
E. with the International application when filed (to be checked by the receiving Office)
D The date of receipt (from the applicant) by the International Bureau '°
(Authorized Officer) Form PCT/RO/134 (January 1981 )
- 166.1 International Application No: PCT/ /
Form PCT/RO/134 (cont.)
American Type Culture Collection
10801 University Blvd. Manassas, VA 201 10-2209 US
Accession No. Date of Deposit
PTA-292 June 29, 1999
PTA-294 June 29, 1999
PTA-295 June 29, 1999
166.2

Claims

What is claimed is:
1. An isolated nucleic acid molecule selected from the group consisting of: a) a nucleic acid molecule comprising a nucleotide sequence which is at least 30% identical to the nucleotide sequence of SEQ ID NO:l, 2, 27 or 28, the cDNA insert of the plasmid deposited with the
ATCC® as Accession Number PTA-292, or a complement thereof; b) a nucleic acid molecule comprising a fragment of at least 480 nucleotides of the nucleotide sequence of SEQ ID NO:l or 2, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292, or a complement thereof; c) a nucleic acid molecule comprising a fragment of at least 575 nucleotides of the nucleotide sequence of SEQ ID NO:27 or 28, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292, or a complement thereof; d) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:3 or 29, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292; e) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:3, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292, or a complement thereof, wherein the fragment comprises at least 10 contiguous amino acids of SEQ ID NO:3, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as
Accession Number PTA-292; f) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:29, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292, or a complement thereof, wherein the fragment comprises at least 45 contiguous amino acids of SEQ ID NO:29, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292;
167 g) nucleic acid molecule comprising a fragment of at least 50 nucleotides of nucleic acids 1 to 2102 of SEQ ID NO:l, or a complement thereof; h) a nucleic acid molecule comprising a fragment of at least 50 nucleotides of nucleic acids 1 to 634 of SEQ ID NO:27, or a complement thereof; i) a nucleic acid molecule comprising a fragment of at least 150 nucleotides of SEQ ID NO:28, or a complement thereof; j) a nucleic acid molecule comprising a fragment of at least 50 nucleotides of nucleotide sequence of SEQ ID NO:2, or complement thereof; k) a nucleic acid molecule comprising a nucleotide sequence which is at least 30% identical to the nucleotide sequence of SEQ ID NO: 36, 37, 44, 45, 58 or 59, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292 or Accession Number
PTA-291, or a complement thereof; 1) a nucleic acid molecule comprising a nucleotide sequence which is at least 45% identical to the nucleotide sequence of SEQ ID NO: 110 or 111, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-294, or a complement thereof; m) a nucleic acid molecule comprising a fragment of at least 50 nucleotides of the nucleotide sequence of SEQ ID NO: 36, 37, 44, 45, 58, 59, 110 or 111, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA- 292, Accession Number PTA-291 or Accession Number PTA- 294, or a complement thereof; n) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:38, 46, 60, or 112, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292,
Accession Number PTA-291 or Accession Number PTA-294; and o) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:38, 46, 60 or 112, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-
292, Accession Number PTA-291 or Accession Number PTA-294,
168 wherein the fragment comprises at least 10 contiguous amino acids of SEQ ED NO:38, 46, 60 or 112, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-292, Accession Number PTA- 291 or Accession Number PTA-295; p) a nucleic acid molecule comprising a nucleotide sequence which is at least 98% identical to the nucleotide sequence of SEQ ID NO:52 or 53, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291, or a complement thereof; q) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:54, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291; r) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:54, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291, wherein the fragment comprises at least 10 contiguous amino acids of SEQ ED NO: 54, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the Accession Number
PTA-291; s) a nucleic acid molecule comprising a nucleotide sequence which is at least 30% identical to the nucleotide sequence of SEQ ID NO:73, 74, 93, 94, 123, 124, 128 OR 129, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 or
Accession Number PTA-295, or a complement thereof; t) a nucleic acid molecule comprising a fragment of at least 450 nucleotides of the nucleotide sequence of SEQ ID NO: 123, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 , or a complement thereof; u) a nucleic acid molecule comprising a fragment of at least 50 nucleotides of the nucleotide sequence of SEQ ID NO:74, 124, 128 or 129, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, Accession Number PTA-291, or a complement thereof;
169 v) a nucleic acid molecule comprising a fragment of at least 50 nucleotides of nucleic acids 1-1250 of SEQ ID NO:73, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, Accession Number PTA-291, or a complement thereof; w) a nucleic acid molecule comprising a fragment of at least 250 nucleotides of the nucleotide sequence of SEQ ID NO:93, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, Accession Number PTA-291, or a complement thereof; x) a nucleic acid molecule comprising a fragment of at least 200 nucleotides of the nucleotide sequence of SEQ ID NO:94, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, Accession Number PTA-291, or a complement thereof; y) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:75, 95, 125 or 130, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 or Accession Number PTA-295, or a complement thereof; z) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO: 95, 125 or 130, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA- 291 or Accession Number PTA-295, wherein the fragment comprises at least 10 contiguous amino acids of SEQ ID NO:95, 125, or 130, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 or Accession Number PTA-295, or a complement thereof; aa) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO: 75, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 or Accession Number PTA-295, wherein the fragment comprises at least 60 contiguous amino acids of SEQ ID NO:75, or the amino
170 acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 or Accession Number PTA-295, or a complement thereof; ab) a nucleic acid molecule comprising a nucleotide sequence which is at least 40% identical to the nucleotide sequence of SEQ ID NO: 63, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, or a complement thereof; ac) a nucleic acid molecule comprising a nucleotide sequence which is at least 65% identical to the nucleotide sequence of SEQ ID NO:64, the cDNA insert of the plasmid deposited with the ATCC® as
Accession Number PTA-295, or a complement thereof; ad) a nucleic acid molecule comprising a fragment of at least 510 nucleotides of the nucleotide sequence of SEQ ID NO:63, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, or a complement thereof; ae) a nucleic acid molecule comprising a fragment of at least 270 nucleotides of the nucleotide sequence of SEQ ID NO:64, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, or a complement thereof; af) a nucleic acid molecule comprising a fragment of at least 20 least nucleotides of nucleic acids 1-255 of SEQ ID NO:64, or a complement thereof; ag) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:65, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295; ah) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:65, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295, wherein the fragment comprises at least 90 contiguous amino acids of SEQ ED NO:65, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-295;
171 ai) a nucleic acid molecule comprising a fragment of at least 20 nucleotides of nucleic acids 775 to 1386 of SEQ ID NO:63, or a complement thereof; and aj) a nucleic acid molecule comprising a fragment of at least 20 nucleotides of nucleic acids 1 to 984 of SEQ ID NO:93, or a complement thereof; and ak) a nucleic acid molecule comprising a fragment of at least 20 nucleotides of nucleic acids 1666 to 1946 of SEQ ID NO:93, or a complement thereof.
2. The isolated nucleic acid molecule of claim 1, which is selected from the group consisting of: a) a nucleic acid comprising the nucleotide sequence of SEQ ID NO: 1 , 2, 27, 28, 36, 37, 44, 45, 52, 53, 58, 59, 63, 64, 73, 74, 93, 94, 110, 111, 123, 124, 128, 129, 136, 138, 140, 142, 144, 146, 148, 150,
152, 154, 156, 158, 160, 162, 164, 166, 168, 170, 172, 174, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228 or 230, the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 , Accession Number PTA-292,
Accession Number PTA-294, Accession Number PTA-295, or a complement thereof; and b) a nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153,
155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-
291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA-295;
3. The nucleic acid molecule of claim 1 further comprising vector nucleic acid sequences.
172
4. The nucleic acid molecule of claim 1 further comprising nucleic acid sequences encoding a heterologous polypeptide.
5. A host cell which contains the nucleic acid molecule of claim 1.
6. The host cell of claim 5 which is a mammalian host cell.
7. A non-human mammalian host cell containing the nucleic acid molecule of claim 1.
8. An isolated polypeptide selected from the group consisting of: a) a fragment of a polypeptide comprising the amino acid sequence of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189,
191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, wherein the fragment comprises at least 10 contiguous amino acids of SEQ ED NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171,
173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231; b) an allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125,
130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of plasmids deposited with the ATCC® as Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA-295, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule comprising SEQ ID NO:2, 28, 37, 45, 53, 59, 64, 74, 94, 111, 124, or 129, or a complement thereof under stringent conditions; and
173 c) a polypeptide which is encoded by a nucleic acid molecule comprising a nucleotide sequence which is at least 30% identical to a nucleic acid comprising the nucleotide sequence of SEQ ID NO:2, 28, 37, 53, 59, 64, 74, 94, 111, 124, or 129, or at least 98% to a nucleic acid comprising the nucleotide sequence of SEQ ID NO:2,
28, 37, 53, 59, 64, 74, 94, 111, 124, or 129, or a complement thereof. d) a polypeptide which is encoded by a nucleic acid molecule comprising a nucleotide sequence which is at least 98% to a nucleic acid comprising the nucleotide sequence of SEQ ID NO:45, or a complement thereof.
9. The isolated polypeptide of claim 8 comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231.
10. The polypeptide of claim 8 further comprising heterologous amino acid sequences.
11. An antibody which selectively binds to a polypeptide of claim 8.
12. The antibody of claim 11, wherein the antibody is a monoclonal antibody.
13. A method for producing a polypeptide selected from the group consisting of: a) a polypeptide comprising the amino acid sequence of SEQ ID
NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167,
169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291 , Accession Number PTA-292,
Accession Number PTA-294, Accession Number PTA-295;
174 b) a polypeptide comprising a fragment of the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215,
217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA- 295, wherein the fragment comprises at least 10 contiguous amino acids of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA- 295; and c) an allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:3, 29, 38, 46, 54, 60, 65, 75, 95, 112, 125, 130, 137, 139, 141, 143, 145, 147, 149, 151, 153, 155, 157, 159, 161, 163, 165, 167, 169, 171, 173, 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 229 or 231, or the amino acid sequence encoded by the cDNA insert of the plasmid deposited with the ATCC® as Accession Number PTA-291, Accession Number PTA-292, Accession Number PTA-294, Accession Number PTA-295, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule comprising SEQ ID NO:l, 27, 36, 44, 52, 58, 63, 73, 93, 110, 123, or 128, or a complement thereof under stringent conditions; comprising culturing the host cell of claim 5 under conditions in which the nucleic acid molecule is expressed.
175
14. A method for detecting the presence of a polypeptide of claim 8 in a sample, comprising: a) contacting the sample with a compound which selectively binds to a polypeptide of claim 8; and b) determining whether the compound binds to the polypeptide in the sample.
15. The method of claim 14, wherein the compound which binds to the polypeptide is an antibody.
16. A kit comprising a compound which selectively binds to a polypeptide of claim 8 and instructions for use.
17. A method for detecting the presence of a nucleic acid molecule of claim 1 in a sample, comprising the steps of: a) contacting the sample with a nucleic acid probe or primer which selectively hybridizes to the nucleic acid molecule; and b) determining whether the nucleic acid probe or primer binds to a nucleic acid molecule in the sample.
18. The method of claim 17, wherein the sample comprises mRNA molecules and is contacted with a nucleic acid probe.
19. A kit comprising a compound which selectively hybridizes to a nucleic acid molecule of claim 1 and instructions for use.
20. A method for identifying a compound which binds to a polypeptide of claim 8 comprising the steps of: a) contacting a polypeptide, or a cell expressing a polypeptide of claim 8 with a test compound; and b) determining whether the polypeptide binds to the test compound.
21. The method of claim 20, wherein the binding of the test compound to the polypeptide is detected by a method selected from the group consisting of: a) detection of binding by direct detecting of test compounαVpolypeptide binding;
176 b) detection of binding using a competition binding assay; c) detection of binding using an assay for TANGO 339, TANGO 353, TANGO 358, TANGO 365, TANGO 368, TANGO 369, TANGO 383, TANGO 393, TANGO 402, MANGO 346, and MANGO 365 - mediated signal transduction.
22. A method for modulating the activity of a polypeptide of claim 8 comprising contacting a polypeptide or a cell expressing a polypeptide of claim 8 with a compound which binds to the polypeptide in a sufficient concentration to modulate the activity of the polypeptide.
23. A method for identifying a compound which modulates the activity of a polypeptide of claim 8, comprising: a) contacting a polypeptide of claim 8 with a test compound; and b) determining the effect of the test compound on the activity of the polypeptide to thereby identify a compound which modulates the activity of the polypeptide.
177
EP00950916A 1999-07-30 2000-07-31 Secreted proteins and uses thereof Ceased EP1268506A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36516499A 1999-07-30 1999-07-30
US365164 1999-07-30
PCT/US2000/020935 WO2001009162A2 (en) 1999-07-30 2000-07-31 Secreted proteins and uses thereof

Publications (2)

Publication Number Publication Date
EP1268506A1 EP1268506A1 (en) 2003-01-02
EP1268506A4 true EP1268506A4 (en) 2004-07-28

Family

ID=23437730

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00950916A Ceased EP1268506A4 (en) 1999-07-30 2000-07-31 Secreted proteins and uses thereof

Country Status (3)

Country Link
EP (1) EP1268506A4 (en)
AU (1) AU6394400A (en)
WO (1) WO2001009162A2 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7371836B2 (en) 1998-03-27 2008-05-13 Genentech, Inc. PRO526 nucleic acids
US7723488B2 (en) 1998-03-27 2010-05-25 Genentech, Inc. Monoclonal antibodies to secreted and transmembrane polypeptides
NZ520065A (en) * 2000-01-12 2006-02-24 Univ Yale Nogo receptor-mediated blockade of axonal growth
US7119165B2 (en) 2000-01-12 2006-10-10 Yale University Nogo receptor-mediated blockade of axonal growth
US20030039647A1 (en) * 2000-07-14 2003-02-27 Christoph Reinhard Tetraspan protein and uses thereof
PT1325130E (en) 2000-10-06 2010-05-31 Biogen Idec Inc Nogo receptor homologs
EP1463818A2 (en) * 2001-12-14 2004-10-06 Incyte Genomics, Inc. Transporters and ion channels
US20080248011A1 (en) * 2004-01-27 2008-10-09 Medical And Biological Laboratories Co., Ltd. Methods for Isolating Monocytes
JP2008540339A (en) * 2005-04-29 2008-11-20 ワイス Nogo receptor functional motif and peptidomimetics related thereto, and methods of using them
US7893032B2 (en) 2005-07-07 2011-02-22 Yale University NgR variants and compositions thereof for suppressing axonal growth inhibition
CN102245640B (en) 2008-12-09 2014-12-31 霍夫曼-拉罗奇有限公司 Anti-PD-L1 antibodies and their use to enhance T-cell function
EP3971211A1 (en) * 2015-07-13 2022-03-23 Compugen Ltd. Hide1 compositions and methods

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998038209A2 (en) * 1997-02-26 1998-09-03 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO1999028462A2 (en) * 1997-12-03 1999-06-10 Genentech, Inc. Polypeptides and nucleic acids encoding the same
WO1999058660A1 (en) * 1998-05-12 1999-11-18 Human Genome Sciences, Inc. 97 human secreted proteins
WO2000029448A2 (en) * 1998-11-17 2000-05-25 Sagami Chemical Research Center Human proteins having hydrophobic domains and dnas encoding these proteins
WO2001075067A2 (en) * 2000-03-31 2001-10-11 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2002006340A2 (en) * 2000-07-14 2002-01-24 Chiron Corporation Tetraspan protein and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998038209A2 (en) * 1997-02-26 1998-09-03 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO1999028462A2 (en) * 1997-12-03 1999-06-10 Genentech, Inc. Polypeptides and nucleic acids encoding the same
WO1999058660A1 (en) * 1998-05-12 1999-11-18 Human Genome Sciences, Inc. 97 human secreted proteins
WO2000029448A2 (en) * 1998-11-17 2000-05-25 Sagami Chemical Research Center Human proteins having hydrophobic domains and dnas encoding these proteins
WO2001075067A2 (en) * 2000-03-31 2001-10-11 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2002006340A2 (en) * 2000-07-14 2002-01-24 Chiron Corporation Tetraspan protein and uses thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL [online] 11 December 1996 (1996-12-11), HILLIER ET AL.: "zl02b08.s1 Soares_pregnant_uterus_NbHPU Homo sapiens cDNA clone IMAGE:491127 3', mRNA sequence", XP002282678, Database accession no. AA137033 *
DATABASE EMBL [online] 12 March 1999 (1999-03-12), BLOECKER ET AL.: "Homo sapiens mRNA; EST DKFZp434C2328_r1 (from clone DKFZp434C2328)", XP002282677, Database accession no. AL043880 *
DATABASE EMBL [online] 21 December 1998 (1998-12-21), JACOBS ET AL.: "Secreted protein BD380_1", XP002282675, Database accession no. AAW70319 *
DATABASE EMBL [online] 31 December 1998 (1998-12-31), STRAUSBERG: "qk68a03.x1 NCI_GAP_Co8 Homo sapiens cDNA clone IMAGE1874092 3', mRNA sequence", XP002282676, Database accession no. AI339682 *

Also Published As

Publication number Publication date
AU6394400A (en) 2001-02-19
WO2001009162A2 (en) 2001-02-08
EP1268506A1 (en) 2003-01-02

Similar Documents

Publication Publication Date Title
US7385036B2 (en) Human tango 509 polypeptides
US6383779B1 (en) Glycoprotein VI and uses thereof
EP1218411A2 (en) Secreted proteins and uses thereof
US8163503B2 (en) Methods of identifying compounds that bind TANGO509
WO2000052022A1 (en) Secreted proteins and nucleic acids encoding them
WO2000039161A1 (en) Class ii cytokine receptor-like proteins and nucleic acids encoding them
WO2001009162A2 (en) Secreted proteins and uses thereof
US20060246065A1 (en) Fail molecules and uses thereof
EP1140976A2 (en) Secreted proteins and uses thereof
US6406884B1 (en) Secreted proteins and uses thereof
US20140072968A1 (en) Novel Genes Encoding Proteins Having Prognostic, Diagnostic, Preventive, Therapeutic, and Other Uses
EP1223218A1 (en) CD2000 and CD2001 molecules and uses thereof
WO2001023523A2 (en) Secreted proteins and uses thereof
WO2000050443A9 (en) Secreted proteins and uses thereof
US20050260702A1 (en) Novel integrin alpha subunit and uses thereof
EP1444260A2 (en) Secreted proteins and uses thereof
WO2001000672A1 (en) Secreted proteins and uses thereof
EP1141269A2 (en) Secreted proteins and uses thereof
WO2001029088A1 (en) Novel genes encoding proteins having prognostic, diagnostic, preventive, therapeutic, and other uses
WO2000050442A2 (en) Secreted proteins and uses thereof
WO2000032746A2 (en) Netrin-like and ependymin-like nucleic acids and polypeptides and uses thereof
WO2001030831A1 (en) Secreted proteins and uses thereof
WO2001081414A2 (en) Integrin alpha subunit and uses thereof

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020206

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 07K 16/18 B

Ipc: 7G 01N 33/50 B

Ipc: 7C 12N 15/12 B

Ipc: 7G 01N 33/53 B

Ipc: 7C 07K 14/435 A

A4 Supplementary search report drawn up and despatched

Effective date: 20040614

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20061102