Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
RECEPTORS AND MEMBRANE-ASSOCIATED PROTEINS
Document Type and Number:
WIPO Patent Application WO/2002/094990
Kind Code:
A2
Abstract:
The invention provides human receptors and membrane-associated proteins (REMAP) and polynucleotides which identify and encode REMAP. The invention also provides expression vectors, host cells, antibodies, agonists, and antagonists. The invention also provides methods for diagnosing, treating, or preventing disorders associated with aberrant expression of REMAP.

Inventors:
LAL PREETI G (US)
WARREN BRIDGET A (US)
XU YUMING (US)
DUGGAN BRENDAN M (US)
HONCHELL CYNTHIA D (US)
KALLICK DEBORAH A (US)
BAUGHN MARIAH R (US)
TANG Y TOM (US)
YUE HENRY (US)
BANDMAN OLGA
Application Number:
PCT/US2002/015899
Publication Date:
November 28, 2002
Filing Date:
May 16, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INCYTE GENOMICS INC (US)
LAL PREETI G (US)
WARREN BRIDGET A (US)
XU YUMING (US)
DUGGAN BRENDAN M (US)
HONCHELL CYNTHIA D (US)
KALLICK DEBORAH A (US)
BAUGHN MARIAH R (US)
TANG Y TOM (US)
YUE HENRY (US)
BANDMAN OLGA
International Classes:
G01N33/50; A61K45/00; A61K49/00; A61P1/00; A61P3/00; A61P25/00; A61P29/00; A61P31/12; A61P35/00; A61P37/06; C07K14/47; C07K14/705; C07K16/28; C07K16/46; C12N1/15; C12N1/19; C12N1/21; C12N5/10; C12N15/02; C12N15/09; C12P21/02; C12P21/08; C12Q1/02; C12Q1/68; G01N33/15; G01N33/53; G01N33/566; (IPC1-7): C12N/
Other References:
NAGASE T ET AL: 'Prediction of the coding sequences of unidentified human genes. XVI. The complete sequences of 150 new cDNA clones from brain which code for large proteins in vitro.' DNA RES. vol. 7, no. 1, 28 February 2000, pages 65 - 73, XP000923011
DATABASE GENCORE [Online] REZGAOUI ET AL: 'Identification of SorCS2, a novel member of the VPS10 domain containing receptor family, prominently expressed in the developing mouse brain.', XP002993637 Retrieved from stn Database accession no. (Q9EPR5) & MECH DEV. vol. 100, 2001, pages 335 - 338
Attorney, Agent or Firm:
Hamlet-cox, Diana (Inc. 3160 Porter Driv, Palo Alto CA, US)
Download PDF:
Claims:
What is claimed is:
1. An isolated polypeptide selected from the group consisting of : a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 126, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 16, SEQ ID N0 : 812, SEQ ID NO : 15, and SEQ ID N0 : 1725, c) a polypeptide comprising a naturally occurring amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID N0 : 7, d) a polypeptide comprising a naturally occurring amino acid sequence at least 94% identical to the amino acid sequence of SEQ ID NO: 13, e) a polypeptide comprising a naturally occurring amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 14, f) a polypeptide comprising a naturally occurring amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 16, g) a polypeptide consisting essentially of a naturally occurring amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID N0 : 26, h) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 126, and i) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 126.
2. An isolated polypeptide of claim 1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO : 126.
3. An isolated polynucleotide encoding a polypeptide of claim 1.
4. An isolated polynucleotide encoding a polypeptide of claim 2.
5. An isolated polynucleotide of claim 4 comprising a polynucleotide sequence selected from the group consisting of SEQ ID N0 : 2752.
6. A recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide of claim 3.
7. A cell transformed with a recombinant polynucleotide of claim 6.
8. A transgenic organism comprising a recombinant polynucleotide of claim 6.
9. A method of producing a polypeptide of claim 1, the method comprising: a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide, and said recombinant polynucleotide comprises a promoter sequence operably linked to a polynucleotide encoding the polypeptide of claim 1, and b) recovering the polypeptide so expressed.
10. A method of claim 9, wherein the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 126.
11. An isolated antibody which specifically binds to a polypeptide of claim 1.
12. An isolated polynucleotide selected from the group consisting of : a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 2752, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 2732 and SEQ ID NO : 3452, c) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 95% identical to the polynucleotide sequence of SEQ ID NO : 33, d) a polynucleotide complementary to a polynucleotide of a), e) a polynucleotide complementary to a polynucleotide of b), f) a polynucleotide complementary to a polynucleotide of c), and g) an RNA equivalent of a)f).
13. An isolated polynucleotide comprising at least 60 contiguous nucleotides of a polynucleotide of claim 12.
14. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising: a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and, optionally, if present, the amount thereof.
15. A method of claim 14, wherein the probe comprises at least 60 contiguous nucleotides.
16. A method of detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide of claim 12, the method comprising: a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.
17. A composition comprising a polypeptide of claim 1 and a pharmaceutically acceptable excipient.
18. A composition of claim 17, wherein the polypeptide comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 126.
19. A method for treating a disease or condition associated with decreased expression of functional REMAP, comprising administering to a patient in need of such treatment the composition of claim 17.
20. A method of screening a compound for effectiveness as an agonist of a polypeptide of claim 1, the method comprising: a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting agonist activity in the sample.
21. A composition comprising an agonist compound identified by a method of claim 20 and a pharmaceutically acceptable excipient.
22. A method for treating a disease or condition associated with decreased expression of functional REMAP, comprising administering to a patient in need of such treatment a composition of claim 21.
23. A method of screening a compound for effectiveness as an antagonist of a polypeptide of claim 1, the method comprising: a) exposing a sample comprising a polypeptide of claim 1 to a compound, and b) detecting antagonist activity in the sample.
24. A composition comprising an antagonist compound identified by a method of claim 23 and a pharmaceutically acceptable excipient.
25. A method for treating a disease or condition associated with overexpression of functional REMAP, comprising administering to a patient in need of such treatment a composition of claim 24.
26. A method of screening for a compound that specifically binds to the polypeptide of claim 1, the method comprising: a) combining the polypeptide of claim 1 with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide of claim 1 to the test compound, thereby identifying a compound that specifically binds to the polypeptide of claim 1.
27. A method of screening for a compound that modulates the activity of the polypeptide of claim 1, the method comprising: a) combining the polypeptide of claim 1 with at least one test compound under conditions permissive for the activity of the polypeptide of claim 1, b) assessing the activity of the polypeptide of claim 1 in the presence of the test compound, and c) comparing the activity of the polypeptide of claim 1 in the presence of the test compound with the activity of the polypeptide of claim 1 in the absence of the test compound, wherein a change in the activity of the polypeptide of claim 1 in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide of claim 1.
28. A method of screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a sequence of claim 5, the method comprising : a) exposing a sample comprising the target polynucleotide to a compound, under conditions suitable for the expression of the target polynucleotide, b) detecting altered expression of the target polynucleotide, and c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.
29. A method of assessing toxicity of a test compound, the method comprising: a) treating a biological sample containing nucleic acids with the test compound, b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide of claim 12 under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide comprising a polynucleotide sequence of a polynucleotide of claim 12 or fragment thereof, c) quantifying the amount of hybridization complex, and d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.
30. A diagnostic test for a condition or disease associated with the expression of REMAP in a biological sample, the method comprising: a) combining the biological sample with an antibody of claim 11, under conditions suitable for the antibody to bind the polypeptide and form an antibody: polypeptide complex, and b) detecting the complex, wherein the presence of the complex correlates with the presence of the polypeptide in the biological sample.
31. The antibody of claim 11, wherein the antibody is: a) a chimeric antibody, b) a single chain antibody, c) a Fab fragment, d) a F (ab') 2 fragment, or e) a humanized antibody.
32. A composition comprising an antibody of claim 11 and an acceptable excipient.
33. A method of diagnosing a condition or disease associated with the expression of REMAP in a subject, comprising administering to said subject an effective amount of the composition of claim 32.
34. A composition of claim 32, wherein the antibody is labeled.
35. A method of diagnosing a condition or disease associated with the expression of REMAP in a subject, comprising administering to said subject an effective amount of the composition of claim 34.
36. A method of preparing a polyclonal antibody with the specificity of the antibody of claim 11, the method comprising: a) immunizing an animal with a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NO: 126, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibodies from said animal, and c) screening the isolated antibodies with the polypeptide, thereby identifying a polyclonal antibody which specifically binds to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 126.
37. A polyclonal antibody produced by a method of claim 36.
38. A composition comprising the polyclonal antibody of claim 37 and a suitable carrier.
39. A method of making a monoclonal antibody with the specificity of the antibody of claim 11, the method comprising: a) immunizing an animal with a polypeptide consisting of an amino acid sequence selected from the group consisting of SEQ ID NO : 126, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibody producing cells from the animal, c) fusing the antibody producing cells with immortalized cells to form monoclonal antibodyproducing hybridoma cells, d) culturing the hybridoma cells, and e) isolating from the culture monoclonal antibody which specifically binds to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO : 126.
40. A monoclonal antibody produced by a method of claim 39.
41. A composition comprising the monoclonal antibody of claim 40 and a suitable carrier.
42. The antibody of claim 11, wherein the antibody is produced by screening a Fab expression library.
43. The antibody of claim 11, wherein the antibody is produced by screening a recombinant immunoglobulin library.
44. A method of detecting a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO : 126 in a sample, the method comprising: a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and b) detecting specific binding, wherein specific binding indicates the presence of a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 126 in the sample.
45. A method of purifying a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 126 from a sample, the method comprising: a) incubating the antibody of claim 11 with a sample under conditions to allow specific binding of the antibody and the polypeptide, and b) separating the antibody from the sample and obtaining the purified polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO : 126.
46. A microarray wherein at least one element of the microarray is a polynucleotide of claim 13.
47. A method of generating an expression profile of a sample which contains polynucleotides, the method comprising: a) labeling the polynucleotides of the sample, b) contacting the elements of the microarray of claim 46 with the labeled polynucleotides of the sample under conditions suitable for the formation of a hybridization complex, and c) quantifying the expression of the polynucleotides in the sample.
48. An array comprising different nucleotide molecules affixed in distinct physical locations on a solid substrate, wherein at least one of said nucleotide molecules comprises a first oligonucleotide or polynucleotide sequence specifically hybridizable with at least 30 contiguous nucleotides of a target polynucleotide, and wherein said target polynucleotide is a polynucleotide of claim 12.
49. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 30 contiguous nucleotides of said target polynucleotide.
50. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to at least 60 contiguous nucleotides of said target polynucleotide.
51. An array of claim 48, wherein said first oligonucleotide or polynucleotide sequence is completely complementary to said target polynucleotide.
52. An array of claim 48, which is a microarray.
53. An array of claim 48, further comprising said target polynucleotide hybridized to a nucleotide molecule comprising said first oligonucleotide or polynucleotide sequence.
54. An array of claim 48, wherein a linker joins at least one of said nucleotide molecules to said solid substrate.
55. An array of claim 48, wherein each distinct physical location on the substrate contains multiple nucleotide molecules, and the multiple nucleotide molecules at any single distinct physical location have the same sequence, and each distinct physical location on the substrate contains nucleotide molecules having a sequence which differs from the sequence of nucleotide molecules at another distinct physical location on the substrate.
56. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 1.
57. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 2.
58. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 3.
59. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 4.
60. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 5.
61. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 6.
62. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 7.
63. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 8.
64. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 9.
65. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 10.
66. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 11.
67. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 12.
68. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 13.
69. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 14.
70. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 15.
71. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 16.
72. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 17.
73. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 18.
74. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 19.
75. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 20.
76. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 21.
77. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 22.
78. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 23.
79. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 24.
80. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 25.
81. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 26.
82. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 27.
83. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 28.
84. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 29.
85. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ U) NO : 30.
86. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 31.
87. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 32.
88. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 33.
89. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 34.
90. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 35.
91. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 36.
92. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 37.
93. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 38.
94. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 39.
95. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 40.
96. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 41.
97. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 42.
98. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 43.
99. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 44.
100. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 45.
101. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 46.
102. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 47.
103. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 48.
104. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 49.
105. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 50.
106. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 51.
107. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 52.
Description:
RECEPTORS AND MEMBRANE-ASSOCIATED PROTEINS TECHNICAL FIELD This invention relates to nucleic acid and amino acid sequences of receptors and membrane- associated proteins and to the use of these sequences in the diagnosis, treatment, and prevention of cell proliferative, autoimmune/inflammatory, neurological, metabolic, developmental, endocrine, cardiovascular, reproductive, gastrointestinal, metabolic, genetic, and lipid metabolism disorders, cancer, and viral infections, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of receptors and membrane-associated proteins.

BACKGROUND OF THE INVENTION Eukaryotic organisms are distinct from prokaryotes in possessing many intracellular membrane-bound compartments such as organelles and vesicles. Many of the metabolic reactions which distinguish eukaryotic biochemistry from prokaryotic biochemistry take place within these compartments. In particular, many cellular functions require very stringent reaction conditions, and the organelles and vesicles enable compartmentalization and isolation of reactions which might otherwise disrupt cytosolic metabolic processes. The organelles include mitochondria, smooth and rough endoplasmic reticula, sarcoplasmic reticulum, and the Golgi body. The vesicles include phagosomes, lysosomes, endosomes, peroxisomes, and secretory vesicles. Organelles and vesicles are bounded by single or double membranes.

Signal transduction is the general process by which cells respond to extracellular signals.

Signal transduction across the plasma membrane begins with the binding of a signal molecule, e. g., a hormone, neurotransmitter, or growth factor, to a cell membrane receptor. The receptor, thus activated, triggers an intracellular biochemical cascade that ends with the activation of an intracellular target molecule, such as a transcription factor. This process of signal transduction regulates all types of cell functions including cell proliferation, differentiation, and gene transcription.

Biological membranes surround organelles, vesicles, and the cell itself. Membranes are highly selective permeability barriers made up of lipid bilayer sheets composed of phosphoglycerides, fatty acids, cholesterol, phospholipids, glycolipids, proteoglycans, and proteins. Membranes contain ion pumps, ion channels, and specific receptors for external stimuli which transmit biochemical signals across the membranes. These membranes also contain second messenger proteins which interact with these pumps, channels, and receptors to amplify and regulate transmission of these signals.

Plasma Membrane Proteins Plasma membrane proteins (MPs) are divided into two groups based upon methods of protein

extraction from the membrane. Extrinsic or peripheral membrane proteins can be released using extremes of ionic strength or pH, urea, or other disruptors of protein interactions. Intrinsic or integral membrane proteins are released only when the lipid bilayer of the membrane is dissolved by detergent.

The majority of known integral membrane proteins are transmembrane proteins (TM) which are characterized by an extracellular, a transmembrane, and an intracellular domain. TM domains are typically comprised of 15 to 25 hydrophobic amino acids which are predicted to adopt an a-helical conformation. TM proteins are classified as bitopic (Types I and II) and polytopic (Types III and IV) (Singer, S. J. (1990) Annu. Rev. Cell Biol. 6: 247-96). Bitopic proteins span the membrane once while polytopic proteins contain multiple membrane-spanning segments. TM proteins carry out a variety of important cellular functions, including acting as cell-surface receptor proteins involved in signal transduction. These functions are represented by growth and differentiation factor receptors, and receptor-interacting proteins such as Drosophila pecanex and frizzled proteins, LIV-1 protein, NF2 protein, and GNS1/SUR4 eukaryotic integral membrane proteins. TM proteins also act as transporters of ions or metabolites, such as gap junction channels (connexins), and ion channels, and as cell anchoring proteins, such as lectins, integrins, and fibronectins. TM proteins are found in vesicle organelle-forming molecules, such as caveolins; or cell recognition molecules, such as cluster of differentiation (CD) antigens, glycoproteins, and mucins.

Many MPs contain amino acid sequence motifs that serve to localize proteins to specific subcellular sites. Examples of these motifs include PDZ domains, KDEL, RGD, NGR, and GSL sequence motifs, von Willebrand factor A (vWFA) domains, and EGF-like domains. RGD, NGR, and GSL motif-containing peptides have been used as drug delivery agents in targeted cancer treatment of tumor vasculature (Arap, W. et al. (1998) Science, 279: 377-380). Furthermore, MPs may also contain amino acid sequence motifs that serve to interact with extracellular or intracellular molecules, such as carbohydrate recognition domains (CRD).

Chemical modification of amino acid residue side chains alters the manner in which MPs interact with other molecules, for example, phospholipid membranes. Examples of such chemical modifications to amino acid residue side chains are covalent bond formation with glycosaminoglycans, oligosaccharides, phospholipids, acetyl and palmitoyl moieties, ADP-ribose, phosphate, and sulphate groups.

RNA encoding membrane proteins may have alternative splice sites which give rise to proteins encoded by the same gene but with different messenger RNA and amino acid sequences.

Splice variant membrane proteins may interact with other ligand and protein isoforms.

Receptors The term receptor describes proteins that specifically recognize other molecules. The

category is broad and includes proteins with a variety of functions. The bulk of receptors are cell surface proteins which bind extracellular ligands and produce cellular responses in the areas of growth, differentiation, endocytosis, and immune response. Other receptors facilitate the selective transport of proteins out of the endoplasmic reticulum and localize enzymes to particular locations in the cell. The term may also be applied to proteins which act as receptors for ligands with known or unknown chemical composition and which interact with other cellular components. For example, the steroid hormone receptors bind to and regulate transcription of DNA.

Cell surface receptors are typically integral plasma membrane proteins. These receptors recognize hormones such as catecholamines ; peptide hormones; growth and differentiation factors; small peptide factors such as thyrotropin-releasing hormone; galanin, somatostatin, and tachykinins; and circulatory system-borne signaling molecules. Cell surface receptors on immune system cells recognize antigens, antibodies, and major histocompatibility complex (MHC)-bound peptides. Other cell surface receptors bind ligands to be internalized by the cell. This receptor-mediated endocytosis functions in the uptake of low density lipoproteins (LDL), transferrin, glucose-or mannose-terminal glycoproteins, galactose-terminal glycoproteins, immunoglobulins, phosphovitellogenins, fibrin, proteinase-inhibitor complexes, plasminogen activators, and thrombospondin (Lodish, H. et al. (1995) Molecular Cell Biology, Scientific American Books, New York NY, p. 723; Mikhailenko, I. et al.

(1997) J. Biol. Chem. 272: 6784-6791).

Receptor Protein Kinases Many growth factor receptors, including receptors for epidermal growth factor, platelet-derived growth factor, fibroblast growth factor, as well as the growth modulator a-thrombin, contain intrinsic protein kinase activities. When growth factor binds to the receptor, it triggers the autophosphorylation of a serine, threonine, or tyrosine residue on the receptor. These phosphorylated sites are recognition sites for the binding of other cytoplasmic signaling proteins. These proteins participate in signaling pathways that eventually link the initial receptor activation at the cell surface to the activation of a specific intracellular target molecule. In the case of tyrosine residue autophosphorylation, these signaling proteins contain a common domain referred to as a Src homology (SH) domain. SH2 domains and SH3 domains are found in phospholipase C-y, PI-3-K p85 regulatory subunit, Ras-GTPase activating protein, and pp60C-srC (Lowenstein, E. J. et al. (1992) Cell 70: 431-442). The cytokine family of receptors share a different common binding domain and include transmembrane receptors for growth hormone (GH), interleukins, erythropoietin, and prolactin.

Other receptors and second messenger-binding proteins have intrinsic serine/threonine protein kinase activity. These include activin/TGF-p/BMP-superfamily receptors, calcium-and diacylglycerol-activated/phospholipid-dependant protein kinase (PK-C), and RNA-dependant protein kinase (PK-R). In addition, other serine/threonine protein kinases, including nematode Twitchin,

have fibronectin-like, immunoglobulin C2-like domains.

G-protein coupled receptors The G-protein coupled receptors (GPCRs), encoded by one of the largest families of genes yet identified, play a central role in the transduction of extracellular signals across the plasma membrane. GPCRs have a proven history of being successful therapeutic targets.

GPCRs are integral membrane proteins characterized by the presence of seven hydrophobic transmembrane domains which together form a bundle of antiparallel alpha (a) helices. GPCRs range in size from under 400 to over 1000 amino acids (Strosberg, A. D. (1991) Eur. J. Biochem. 196: 1-10; Coughlin, S. R. (1994) Curr. Opin. Cell Biol. 6: 191-197). The amino-terminus of a GPCR is extracellular, is of variable length, and is often glycosylated. The carboxy-terminus is cytoplasmic and generally phosphorylated. Extracellular loops alternate with intracellular loops and link the transmembrane domains. Cysteine disulfide bridges linking the second and third extracellular loops may interact with agonists and antagonists. The most conserved domains of GPCRs are the transmembrane domains and the first two cytoplasmic loops. The transmembrane domains account, in part, for structural and functional features of the receptor. In most cases, the bundle of a helices forms a ligand-binding pocket. The extracellular N-terminal segment, or one or more of the three extracellular loops, may also participate in ligand binding. Ligand binding activates the receptor by inducing a conformational change in intracellular portions of the receptor. In turn, the large, third intracellular loop of the activated receptor interacts with a heterotrimeric guanine nucleotide binding (G) protein complex which mediates further intracellular signaling activities, including the activation of second messengers such as cyclic AMP (cAMP), phospholipase C, and inositol triphosphate, and the interaction of the activated GPCR with ion channel proteins. (See, e. g., Watson, S. and S.

Arkinstall (1994) The G-protein Linked Receptor Facts Book, Academic Press, San Diego CA, pp. 2- 6; Bolander, F. F. (1994) Molecular Endocrinology, Academic Press, San Diego CA, pp. 162-176; Baldwin, J. M. (1994) Curr. Opin. Cell Biol. 6: 180-190.) GPCRs include receptors for sensory signal mediators (e. g., light and olfactory stimulatory molecules); adenosine, y-aminobutyric acid (GABA), hepatocyte growth factor, melanocortins, neuropeptide Y, opioid peptides, opsins, somatostatin, tachykinins, vasoactive intestinal polypeptide family, and vasopressin ; biogenic amines (e. g., dopamine, epinephrine and norepinephrine, histamine, glutamate (metabotropic effect), acetylcholine (muscarinic effect), and serotonin); chemokines; lipid mediators of inflammation (e. g., prostaglandins and prostanoids, platelet activating factor, and leukotrienes); and peptide hormones (e. g., bombesin, bradykinin, calcitonin, C5a anaphylatoxin, endothelin, follicle-stimulating hormone (FSH), gonadotropic-releasing hormone (GnRH), neurokinin, and thyrotropin-releasing hormone (TRH), and oxytocin). GPCRs which act as receptors for stimuli that have yet to be identified are known as orphan receptors.

The diversity of the GPCR family is further increased by alternative splicing. Many GPCR genes contain introns, and there are currently over 30 such receptors for which splice variants have been identified. The largest number of variations are at the protein C-terminus. N-terminal and cytoplasmic loop variants are also frequent, while variants in the extracellular loops or transmembrane domains are less common. Some receptors have more than one site at which variance can occur. The splicing variants appear to be functionally distinct, based upon observed differences in distribution, signaling, coupling, regulation, and ligand binding profiles (Kilpatrick, G. J. et al.

(1999) Trends Pharmacol. Sci. 20: 294-301).

GPCRs can be divided into three major subfamilies: the rhodopsin-like, secretin-like, and metabotropic glutamate receptor subfamilies. Members of these GPCR subfamilies share similar functions and the characteristic seven transmembrane structure, but have divergent amino acid sequences. The largest family consists of the rhodopsin-like GPCRs, which transmit diverse extracellular signals including hormones, neurotransmitters, and light. Rhodopsin is a photosensitive GPCR found in animal retinas. In vertebrates, rhodopsin molecules are embedded in membranous stacks found in photoreceptor (rod) cells. Each rhodopsin molecule responds to a photon of light by triggering a decrease in cGMP levels which leads to the closure of plasma membrane sodium channels. In this manner, a visual signal is converted to a neural impulse. Other rhodopsin-like GPCRs are directly involved in responding to neurotransmitters. These GPCRs include the receptors for adrenaline (adrenergic receptors), acetylcholine (muscarinic receptors), adenosine, galanin, and glutamate (N-methyl-D-aspartate/NMDA receptors). (Reviewed in Watson, S. and S. Arkinstall (1994) The G-Protein Linked Receptor Facts Book, Academic Press, San Diego CA, pp. 7-9,19-22, 32-35,130-131,214-216,221-222; Habert-Ortoli, E. et al. (1994) Proc. Natl. Acad. Sci. USA 91: 9780-9783.) The galanin receptors mediate the activity of the neuroendocrine peptide galanin, which inhibits secretion of insulin, acetylcholine, serotonin and noradrenaline, and stimulates prolactin and growth hormone release. Galanin receptors are involved in feeding disorders, pain, depression, and Alzheimer's disease (Kask, K. et al. (1997) Life Sci. 60: 1523-1533). Other nervous system rhodopsin-like GPCRs include a growing family of receptors for lysophosphatidic acid and other lysophospholipids, which appear to have roles in development and neuropathology (Chun, J. et al.

(1999) Cell Biochem. Biophys. 30: 213-242).

The largest subfamily of GPCRs, the olfactory receptors, are also members of the rhodopsin- like GPCR family. These receptors function by transducing odorant signals. Numerous distinct olfactory receptors are required to distinguish different odors. Each olfactory sensory neuron expresses only one type of olfactory receptor, and distinct spatial zones of neurons expressing distinct receptors are found in nasal passages. For example, the RAlc receptor which was isolated from a rat

brain library, has been shown to be limited in expression to very distinct regions of the brain and a defined zone of the olfactory epithelium (Raming, K. et al. (1998) Receptors Channels 6: 141-151).

However, the expression of olfactory-like receptors is not confined to olfactory tissues. For example, three rat genes encoding olfactory-like receptors having typical GPCR characteristics showed expression patterns not only in taste and olfactory tissue, but also in male reproductive tissue (Thomas, M. B. et al. (1996) Gene 178: 1-5).

GPCR mutations, which may cause loss of function or constitutive activation, have been associated with numerous human diseases (Coughlin, supra). For instance, retinitis pigmentosa may arise from mutations in the rhodopsin gene. Furthermore, somatic activating mutations in the thyrotropin receptor have been reported to cause hyperfunctioning thyroid adenomas, suggesting that certain GPCRs susceptible to constitutive activation may behave as protooncogenes (Parma, J. et al.

(1993) Nature 365: 649-651). GPCR receptors for the following ligands also contain mutations associated with human disease: luteinizing hormone (precocious puberty); vasopressin V2 (X-linked nephrogenic diabetes); glucagon (diabetes and hypertension); calcium (hyperparathyroidism, hypocalcuria, hypercalcemia) ; parathyroid hormone (short limbed dwarfism); P3-adrenoceptor (obesity, non-insulin-dependent diabetes mellitus); growth hormone releasing hormone (dwarfism); and adrenocorticotropin (glucocorticoid deficiency) (Wilson, S. et al. (1998) Br. J. Pharmocol.

125: 1387-1392; Stadel, J. M. et al. (1997) Trends Pharmacol. Sci. 18 : 430-437). GPCRs are also involved in depression, schizophrenia, sleeplessness, hypertension, anxiety, stress, renal failure, and several cardiovascular disorders (Horn, F. and G. Vriend (1998) J. Mol. Med. 76: 464-468).

Mutations and changes in transcriptional activation of GPCR-encoding genes have been associated with neurological disorders such as schizophrenia, Parkinson's disease, Alzheimer's disease, drug addiction, and feeding disorders. The juvenile development and fertility-2 (jdf-2) locus, also called runtyjerky-sterile (rjs), is associated with deletions and point mutations in HERC2, a gene encoding a guanine nucleotide exchange factor protein involved in vesicular trafficking (Walkowicz, M. et al.

(1999) Mamm. Genome 10: 870-878).

In addition, within the past 20 years several hundred new drugs have been recognized that are directed towards activating or inhibiting GPCRs. The therapeutic targets of these drugs span a wide range of diseases and disorders, including cardiovascular, gastrointestinal, and central nervous system disorders as well as cancer, osteoporosis and endometriosis (Wilson, supra ; Stadel, supra). For example, the dopamine agonist L-dopa is used to treat Parkinson's disease, while a dopamine antagonist is used to treat schizophrenia and the early stages of Huntington's disease. Agonists and antagonists of adrenoceptors have been used for the treatment of asthma, high blood pressure, other cardiovascular disorders, and anxiety; muscarinic agonists are used in the treatment of glaucoma and tachycardia; serotonin 5HT1D antagonists are used against migraine; and histamine HI antagonists

are used against allergic and anaphylactic reactions, hay fever, itching, and motion sickness (Horn, supra).

Members of the secretin-like GPCR subfamily have as their ligands peptide hormones such as secretin, calcitonin, glucagon, growth hormone-releasing hormone, parathyroid hormone, and vasoactive intestinal peptide. For example, the secretin receptor responds to secretin, a peptide hormone that stimulates the secretion of enzymes and ions in the pancreas and small intestine (Watson, supra, pp. 278-283). Secretin receptors are about 450 amino acids in length and are found in the plasma membrane of gastrointestinal cells. Binding of secretin to its receptor stimulates the production of cAMP.

Examples of secretin-like GPCRs implicated in inflammation and the immune response include the EGF module-containing, mucin-like hormone receptor (Emrl) and CD97 receptor proteins. CD97 is predominantly expressed in leukocytes and is markedly upregulated on activated B and T cells (McKnight, A. J. and S. Gordon (1998) J. Leukoc. Biol. 63: 271-280). These GPCRs are members of the recently characterized EGF-TM7 receptors subfamily. These seven transmembrane hormone receptors exist as heterodimers in vivo and contain between three and seven potential calcium-binding EGF-like motifs. The EGF motif is about forty amino acid residues in length and includes six conserved cysteine residues, and a calcium-binding site near the N-terminus of the signature sequence. Post-translational hydroxylation of aspartic acid or asparagine residues has been associated with EGF-like domains in several proteins (Prosite PDOC00010 Aspartic acid and asparagine hydroxylation site). The EGF-TM7 family also includes the recently isolated EGF-TM7- latrophilin-related protein (ETL), which is expressed in cardiac myocytes and smooth muscle and is developmentally regulated in the heart. ETL may play a role in effecting the terminal differentiation of cardiac muscle. ETL may also be involved in coronary angiogenesis. (Nechiporuk, T. et al. (2001) J. Biol. Chem. 276: 4150-4157).

Another GPCR subfamily is the metabotropic glutamate receptor family. Glutamate is the major excitatory neurotransmitter in the central nervous system. The metabotropic glutamate receptors modulate the activity of intracellular effectors, and are involved in long-term potentiation (Watson, supra, p. 130). The Ca2+-sensing receptor, which senses changes in the extracellular concentration of calcium ions, has a large extracellular domain including clusters of acidic amino acids which may be involved in calcium binding. The metabotropic glutamate receptor family also includes pheromone receptors, the GABAB receptors, and the taste receptors.

Other subfamilies of GPCRs include two groups of chemoreceptor genes found in the nematodes Caenorhabditis elegans and Caenorhabditis briggsae, which are distantly related to the mammalian olfactory receptor genes. The yeast pheromone receptors STE2 and STE3, involved in the response to mating factors on the cell membrane, have their own seven-transmembrane signature,

as do the cAMP receptors from the slime mold Dictyostelium discoideum, which are thought to regulate the aggregation of individual cells and control the expression of numerous developmentally- regulated genes.

Recent research suggests potential future therapeutic uses for GPCRs in the treatment of metabolic disorders including diabetes, obesity, and osteoporosis. For example, mutant V2 vasopressin receptors causing nephrogenic diabetes could be functionally rescued in vitro by co- expression of a C-terminal V2 receptor peptide spanning the region containing the mutations. This result suggests a possible novel strategy for disease treatment (Schoneberg, T. et al. (1996) EMBO J.

15: 1283-1291). Mutations in melanocortin-4 receptor (MC4R) are implicated in human weight regulation and obesity. As with the vasopressin V2 receptor mutants, these MC4R mutants are defective in trafficking to the plasma membrane (Ho, G. and R. G. MacKenzie (1999) J. Biol. Chem.

274: 35816-35822), and thus might be treated with a similar strategy. The type 1 receptor for parathyroid hormone (PTH) is a GPCR that mediates the PTH-dependent regulation of calcium homeostasis in the bloodstream. Study of PTH/receptor interactions may enable the development of novel PTH receptor ligands for the treatment of osteoporosis (Mannstadt, M. et al. (1999) Am. J.

Physiol. 277: F665-F675).

The chemokine receptor group of GPCRs have potential therapeutic utility in inflammation and infectious disease. (For review, see Locati, M. and P. M. Murphy (1999) Annu. Rev. Med.

50: 425-440.) Chemokines are small polypeptides that act as intracellular signals in the regulation of leukocyte trafficking, hematopoiesis, and angiogenesis. Targeted disruption of various chemokine receptors in mice indicates that these receptors play roles in pathologic inflammation and in autoimmune disorders such as multiple sclerosis. Chemokine receptors are also exploited by infectious agents, including herpesviruses and the human immunodeficiency virus (HIV-1) to facilitate infection. A truncated version of chemokine receptor CCR5, which acts as a coreceptor for infection of T-cells by HIV-1, results in resistance to AIDS, suggesting that CCR5 antagonists could be useful in preventing the development of AIDS.

Interleukins (IL) mediate the interactions between immune and inflammatory cells. Several interleukins have been described; each has unique biological activities as well as some that overlap with the others. Macrophages produce IL-1 and IL-6, whereas T cells produce IL-2, IL-3, IL-4, IL-5 and IL-6 and bone marrow stromal cells produce IL 7. IL 1 and IL 6 not only play important roles in immune cell function, but also stimulate a spectrum of inflammatory cell types. The growth and differentiation of eosinophils is markedly enhanced by IL 5. IL 2 is a potent proliferative signal for T cells, natural killer cells, and lymphokine-activated killer cells. IL 1, IL 3, IL 4, and IL 7 enhance the development of a variety of hematopoietic precursors. IL 4-IL 6 also serve to enhance B cell proliferation and antibody production (Mizel, S. B. (1989) FASEB J. 3: 2379-2388).

Melatonin scavenges free radicals including the hydroxyl radical (. OH), peroxynitrite anion (ONOO-), and hypochlorous acid (HOC1), as well as preventing the translocation of nuclear factor-kappa B (NF-kappa B) to the nucleus and its binding to DNA, thereby reducing the upregulation of proinflammatory cytokines such as interleukins and tumor neurosis factor-alpha.

Melatonin attenuates transendothelial cell migration and edema, which contribute to tissue damage (Reiter, R. J. et al. (2000) Ann. N. Y. Acad. Sci. 917: 376-386). Activation of melatonin receptors enhances the release of T-helper cell cytokines, such as gamma-interferon and interleukin-2 (IL-2), as well as activation of opioid cytokines which crossreact immunologically with both interleukin-4 and dynorphin B. Hematopoiesis is influenced by melatonin-induced-opioids acting on kappa 1-opioid receptors present on bone marrow macrophages (Maestroni, G. J. (1999) Adv. Exp. Med. Biol.

467: 217-226).

Ligand-Gated Receptor Ion Channels Ligand-gated receptor ion channels fall into two categories. The first category, extracellular ligand-gated receptor ion channels (ELGs), rapidly transduce neurotransmitter-binding events into electrical signals, such as fast synaptic neurotransmission. ELG function is regulated by post- translational modification. The second category, intracellular ligand-gated receptor ion channels (ILGs), are activated by many intracellular second messengers and do not require post-translational modification (s) to effect a channel-opening response.

ELGs depolarize excitable cells to the threshold of action potential generation. In non- excitable cells, ELGs permit a limited calcium ion-influx during the presence of agonist. ELGs include channels directly gated by neurotransmitters such as acetylcholine, L-glutamate, glycine, ATP, serotonin, GABA, and histamine. ELG genes encode proteins having strong structural and functional similarities. ILGs are encoded by distinct and unrelated gene families and include receptors for cAMP, cGMP, calcium ions, ATP, and metabolites of arachidonic acid.

Macrophage Scavenger Receptors Macrophage scavenger receptors with broad ligand specificity may participate in the binding of low density lipoproteins (LDL) and foreign antigens. Scavenger receptors types I and II are trimeric membrane proteins with each subunit containing a small N-terminal intracellular domain, a transmembrane domain, a large extracellular domain, and a C-terminal cysteine-rich domain. The extracellular domain contains a short spacer domain, an a-helical coiled-coil domain, and a triple helical collagenous domain. These receptors have been shown to bind a spectrum of ligands, including chemically modified lipoproteins and albumin, polyribonucleotides, polysaccharides, phospholipids, and asbestos (Matsumoto, A. et al. (1990) Proc. Natl. Acad. Sci. USA 87: 9133-9137; Elomaa, O. et al. (1995) Cell 80: 603-609). The scavenger receptors are thought to play a key role in atherogenesis by mediating uptake of modified LDL in arterial walls, and in host defense by binding

bacterial endotoxins, bacteria, and protozoa.

T-Cell Receptors T cells play a dual role in the immune system as effectors and regulators, coupling antigen recognition with the transmission of signals that induce cell death in infected cells and stimulate proliferation of other immune cells. Although a population of T cells can recognize a wide range of different antigens, an individual T cell can only recognize a single antigen and only when it is presented to the T cell receptor (TCR) as a peptide complexed with a major histocompatibility molecule (MHC) on the surface of an antigen presenting cell. The TCR on most T cells consists of immunoglobulin-like integral membrane glycoproteins containing two polypeptide subunits, a and p, of similar molecular weight. Both TCR subunits have an extracellular domain containing both variable and constant regions, a transmembrane domain that traverses the membrane once, and a short intracellular domain (Saito, H. et al. (1984) Nature 309: 757-762). The genes for the TCR subunits are constructed through somatic rearrangement of different gene segments. Interaction of antigen in the proper MHC context with the TCR initiates signaling cascades that induce the proliferation, maturation, and function of cellular components of the immune system (Weiss, A. (1991) Annu. Rev.

Genet. 25: 487-510). Rearrangements in TCR genes and alterations in TCR expression have been noted in lymphomas, leukemias, autoimmune disorders, and immunodeficiency disorders (Aisenberg, A. C. et al. (1985) N. Engl. J. Med. 313: 529-533; Weiss, supra).

Netrin Receptors: The netrins are a family of molecules that function as diffusible attractants and repellants to guide migrating cells and axons to their targets within the developing nervous system. The netrin receptors include the C. elegans protein UNC-5, as well as homologues recently identified in vertebrates (Leonardo, E. D. et al. (1997) Nature 386: 833-838). These receptors are members of the immunoglobulin superfamily, and also contain a characteristic domain called the ZU5 domain.

Mutations in the mouse member of the netrin receptor family, Rcm (rostral cerebellar malformation) result in cerebellar and midbrain defects as an apparent result of abnormal neuronal migration (Ackerman, S. L. et al. (1997) Nature 386: 838-842).

VPS 10 Domain Containing Receptors The members of the VPS 10 domain containing receptor family all contain a domain with homology to the yeast vacuolar sorting protein 10 (VPS 10) receptor. This family includes the mosaic receptor SorLA, the neurotensin receptor sortilin, and SorCS, which is expressed during mouse embryonal and early postnatal nervous system development (Hermey, G. et al. (1999) Biochem.

Biophys. Res. Commun. 266: 347-351 ; Hermey, G. et al. (2001) Neuroreport 12: 29-32). A recently identified member of this family, SorCS2, is highly expressed in the developing and mature mouse central nervous system. Its main site of expression is the floor plate, and high levels are also detected

transiently in brain regions including the dopaminergic brain nuclei and the dorsal thalamus (Rezgaoui, M. (2001) Mech. Dev. 100: 335-338).

Membrane-Associated Proteins Tetraspan Family Proteins The transmembrane 4 superfamily (TM4SF) or tetraspan family is a multigene family encoding type III integral membrane proteins (Wright, M. D. and Tomlinson, M. G. (1994) Immunol.

Today 15: 588-594). The TM4SF is comprised of membrane proteins which traverse the cell membrane four times. Members of the TM4SF include platelet and endothelial cell membrane proteins, melanoma-associated antigens, leukocyte surface glycoproteins, colonal carcinoma antigens, tumor-associated antigens, and surface proteins of the schistosome parasites (Jankowski, S. A. (1994) Oncogene 9: 1205-1211). Members of the TM4SF share about 25-30% amino acid sequence identity with one another. A number of TM4SF members have been implicated in signal transduction, control of cell adhesion, regulation of cell growth and proliferation, including development and oncogenesis, and cell motility, including tumor cell metastasis. Expression of TM4SF proteins is associated with a variety of tumors and the level of expression may be altered when cells are growing or activated.

Tumor Antigens Tumor antigens are surface molecules that are differentially expressed in tumor cells relative to normal cells. Tumor antigens distinguish tumor cells immunologically from normal cells and provide diagnostic and therapeutic targets for human cancers (Takagi, S. et al. (1995) Int. J. Cancer 61: 706-715; Liu, E. et al. (1992) Oncogene 7: 1027-1032).

Ion Channels Ion channels are found in the plasma membranes of virtually every cell in the body. For example, chloride channels mediate a variety of cellular functions including regulation of membrane potentials and absorption and secretion of ions across epithelial membranes. When present in intracellular membranes of the Golgi apparatus and endocytic vesicles, chloride channels also regulate organelle pH. (See, e. g., Greger, R. (1988) Annu. Rev. Physiol. 50: 111-122.) Electrophysiological and pharmacological properties of chloride channels, including ion conductance, current-voltage relationships, and sensitivity to modulators, suggest that different chloride channels exist in muscles, neurons, fibroblasts, epithelial cells, and lymphocytes. Many channels have sites for phosphorylation by one or more protein kinases including protein kinase A, protein kinase C, tyrosine kinase, and casein kinase II, all of which regulate ion channel activity in cells. Inappropriate phosphorylation of proteins in cells has been linked to changes in cell cycle progression and cell differentiation. Changes in the cell cycle have been linked to induction of apoptosis or cancer.

Changes in cell differentiation have been linked to diseases and disorders of the reproductive system,

immune system, and skeletal muscle.

Cerebellar granule neurons possess a non-inactivating potassium current which modulates firing frequency upon receptor stimulation by neurotransmitters and controls the resting membrane potential. Potassium channels that exhibit non-inactivating currents include the ether a go-go (EAG) channel. A membrane protein designated KCR1 specifically binds to rat EAG by means of its C- terminal region and regulates the cerebellar non-inactivating potassium current. KCR1 is predicted to contain 12 transmembrane domains, with intracellular amino and carboxyl termini. Structural characteristics of these transmembrane regions appear to be similar to those of the transporter superfamily, but no homology between KCR1 and known transporters was found, suggesting that KCR1 belongs to a novel class of transporters. KCR1 appears to be the regulatory component of non-inactivating potassium channels (Hoshi, N. et al. (1998) J. Biol. Chem. 273: 23080-23085).

Proton pumps Proton ATPases are a large class of membrane proteins that use the energy of ATP hydrolysis to generate an electrochemical proton gradient across a membrane. The resultant gradient may be used to transport other ions across the membrane (Na+, K+, or Cl') or to maintain organelle pH.

Proton ATPases are further subdivided into the mitochondrial F-ATPases, the plasma membrane ATPases, and the vacuolar ATPases. The vacuolar ATPases establish and maintain an acidic pH within various vesicles involved in the processes of endocytosis and exocytosis (Mellman, I. et al.

(1986) Ann. Rev. Biochem. 55: 663-700).

Proton-coupled, 12 membrane-spanning domain transporters such as PEPT 1 and PEPT 2 are responsible for gastrointestinal absorption and for renal reabsorption of peptides using an electrochemical H gradient as the driving force. Another type of peptide transporter, the TAP transporter, is a heterodimer consisting of TAP 1 and TAP 2 and is associated with antigen processing. Peptide antigens are transported across the membrane of the endoplasmic reticulum by TAP so they can be expressed on the cell surface in association with MHC molecules. Each TAP protein consists of multiple hydrophobic membrane spanning segments and a highly conserved ATP-binding cassette (Boll, M. et al. (1996) Proc. Natl. Acad. Sci. 93: 284-289). Pathogenic microorganisms, such as herpes simplex virus, may encode inhibitors of TAP-mediated peptide transport in order to evade immune surveillance (Marusina, K. and Manaco, J. J. (1996) Curr. Opin.

Hematol. 3: 19-26).

ABC Transporters ATP-binding cassette (ABC) transporters, also called the"traffic ATPases", are a superfamily of membrane proteins that mediate transport and channel functions in prokaryotes and eukaryotes (Higgins, C. F. (1992) Annu. Rev. Cell Biol. 8: 67-113). ABC proteins share a similar overall structure and significant sequence homology. All ABC proteins contain a conserved domain of

approximately two hundred amino acid residues which includes one or more nucleotide binding domains. Mutations in ABC transporter genes are associated with various disorders, such as hyperbilirubinemia II/Dubin-Johnson syndrome, recessive Stargardt's disease, X-linked adrenoleukodystrophy, multidrug resistance, celiac disease, and cystic fibrosis.

Semaphorins and Neuropilins Semaphorins are a large group of axonal guidance molecules consisting of at least 30 different members and are found in vertebrates, invertebrates, and even certain viruses. All semaphorins contain the sema domain which is approximately 500 amino acids in length. Neuropilin, a semaphorin receptor, has been shown to promote neurite outgrowth in vitro. The extracellular region of neuropilins consists of three different domains: CUB, discoidin, and MAM domains. The CUB and the MAM motifs of neuropilin have been suggested to have roles in protein-protein interactions and are thought to be involved in the binding of semaphorins through the sema and the C-terminal domains (reviewed in Raper, J. A. (2000) Curr. Opin. Neurobiol. 10: 88-94).

Membrane Proteins Associated with Intercellular Communication Intercellular communication is essential for the development and survival of multicellular organisms. Cells communicate with one another through the secretion and uptake of protein signaling molecules. The uptake of proteins into the cell is achieved by endocytosis, in which the interaction of signaling molecules with the plasma membrane surface, often via binding to specific receptors, results in the formation of plasma membrane-derived vesicles that enclose and transport the molecules into the cytosol. The secretion of proteins from the cell is achieved by exocytosis, in which molecules inside of the cell are packaged into membrane-bound transport vesicles derived from the trans Golgi network. These vesicles fuse with the plasma membrane and release their contents into the surrounding extracellular space. Endocytosis and exocytosis result in the removal and addition of plasma membrane components, and the recycling of these components is essential to maintain the integrity, identity, and functionality of both the plasma membrane and internal membrane-bound compartments.

Synaptobrevins are synaptic vesicle-associated membrane proteins (VAMPs) which were first discovered in rat brain. These proteins were initially thought to be limited to neuronal cells and to function in the movement of vesicles from the plasmalemma of one cell, across the synapse, to the plasmalemma of another cell. Synaptobrevins are now known to occur and function in constitutive vesicle trafficking pathways involving receptor-mediated endocytotic and exocytotic pathways of many non-neuronal cell types. This regulated vesicle trafficking pathway may be blocked by the highly specific action of clostridial neurotoxins which cleave the synaptobrevin molecule.

In vitro studies of various cellular membranes (Galli et al. (1994) J. Cell. Biol. 125: 1015-24; Link et al. (1993) J. Biol. Chem. 268: 18423-6) have shown that VAMPS are widely distributed.

These important membrane trafficking proteins appear to participate in axon extension via exocytosis during development, in the release of neurotransmitters and modulator peptides, and in endocytosis.

Endocytotic vesicular transport includes such intracellular events as the fusions and fissions of the nuclear membrane, endoplasmic reticulum, Golgi apparatus, and various inclusion bodies such as peroxisomes or lysosomes. Endocytotic processes appear to be universal in eukaryotic cells as diverse as yeast, Caenorhabditis elegans, Drosophila, and mammals.

VAMP-1B is involved in subcellular targeting and is an isoform of VAMP-lA (Isenmann, S. et al. (1998) Mol. Biol. Cell 9: 1649-1660). Four additional splice variants (VAMP-1C to F) have recently been identified. Each variant has variable sequences only at the extreme C-terminus, suggesting that the C-terminus is important in vesicle targeting (Berglund, L. et al. (1999) Biochem.

Biophys. Res. Commun. 264: 777-780).

Nogo has been identified as a component of the central nervous system myelin that prevents axonal regeneration in adult vertebrates. Cleavage of the Nogo-66 receptor and other glycophosphatidylinositol-linked proteins from axonal surfaces renders neurons insensitive to Nogo- 66, facilitating potential recovery from CNS damage (Fournier, A. E. et al. (2001) Nature 409: 341- 346).

The slit proteins are extracellular matrix proteins expressed by cells at the ventral midline of the nervous system. Slit proteins are ligands for the repulsive guidance receptor Roundabout (Robo) and thus play a role in repulsive axon guidance (Brose, K. et al. (1999) Cell 96: 795-806).

Lysosomes are the site of degradation of intracellular material during autophagy and of extracellular molecules following endocytosis. Lysosomal enzymes are packaged into vesicles which bud from the trans-Golgi network. These vesicles fuse with endosomes to form the mature lysosome in which hydrolytic digestion of endocytosed material occurs. Lysosomes can fuse with autophagosomes to form a unique compartment in which the degradation of organelles and other intracellular components occurs.

Protein sorting by transport vesicles, such as the endosome, has important consequences for a variety of physiological processes including cell surface growth, the biogenesis of distinct intracellular organelles, endocytosis, and the controlled secretion of hormones and neurotransmitters (Rothman, J. E. and Wieland, F. T. (1996) Science 272: 227-234). In particular, neurodegenerative disorders and other neuronal pathologies are associated with biochemical flaws during endosomal protein sorting or endosomal biogenesis (Mayer R. J. et al. (1996) Adv. Exp. Med. Biol. 389: 261- 269).

Peroxisomes are organelles independent from the secretory pathway. They are the site of many peroxide-generating oxidative reactions in the cell. Peroxisomes are unique among eukaryotic organelles in that their size, number, and enzyme content vary depending upon organism, cell type,

and metabolic needs (Waterham, H. R. and Cregg, J. M. (1997) BioEssays 19: 57-66). Genetic defects in peroxisome proteins which result in peroxisomal deficiencies have been linked to a number of human pathologies, including Zellweger syndrome, rhizomelic chonrodysplasia punctata, X-linked adrenoleukodystrophy, acyl-CoA oxidase deficiency, bifunctional enzyme deficiency, classical Refsum's disease, DHAP alkyl transferase deficiency, and acatalasemia (Moser, H. W. and Moser, A. B. (1996) Ann. NY Acad. Sci. 804: 427-441). In addition, Gartner, J. et al. (1991; Pediatr. Res.

29: 141-146) found a 22 kDa integral membrane protein associated with lower density peroxisome- like subcellular fractions in patients with Zellweger syndrome.

Normal embryonic development and control of germ cell maturation is modulated by a number of secretory proteins which interact with their respective membrane-bound receptors. Cell fate during embryonic development is determined by members of the activin/TGF-P superfamily, cadherins, IGF-2, and other morphogens. In addition, proliferation, maturation, and redifferentiation of germ cell and reproductive tissues are regulated, for example, by IGF-2, inhibins, activins, and follistatins (Petraglia, F. (1997) Placenta 18: 3-8; Mather, J. P. et al. (1997) Proc. Soc. Exp. Biol. Med.

215: 209-222). Transforming growth factor beta (TGFbeta) signal transduction is mediated by two receptor Ser/Thr kinases acting in series, type n TGFbeta receptor and (TbetaR-II) phosphorylating type I TGFbeta receptor (TbetaR-I). TbetaR-I-associated protein-I (TRECAP-1), which distinguishes between quiescent and activated forms of the type I transforming growth factor beta receptor, has been associated with TGFbeta signaling (Charng, M. J et al. (1998) J. Biol. Chem. 273: 9365-9368).

Retinoic acid receptor alpha (RAR alpha) mediates retinoic-acid induced maturation and has been implicated in myeloid development. Genes induced by retinoic acid during granulocytic differentiation include E3, a hematopoietic-specific gene that is an immediate target for the activated RAR alpha during myelopoiesis (Scott, L. M. et al. (1996) Blood 88 : 2517-2530).

The st-opioid receptor (MOR) mediates the actions of analgesic agents including morphine, codeine, methadone, and fentanyl as well as heroin. MOR is functionally coupled to a G-protein- activated potassium channel (Mestek A. et al. (1995) J. Neurosci. 15: 2396-2406). A variety of MOR subtypes exist. Alternative splicing has been observed with MOR-1 as with a number of G protein-coupled receptors including somatostatin 2, dopamine D2, prostaglandin EP3, and serotonin receptor subtypes 5-hydroxytryptamine4 and 5-hydroxytryptamine7 (Pan, Y. X. et al. (1999) Mol.

Pharm. 56: 396-403).

Peripheral and Anchored Membrane Proteins Some membrane proteins are not membrane-spanning but are attached to the plasma membrane via membrane anchors or interactions with integral membrane proteins. Membrane anchors are covalently joined to a protein post-translationally and include such moieties as prenyl, myristyl, and glycosylphosphatidyl inositol groups. Membrane localization of peripheral and

anchored proteins is important for their function in processes such as receptor-mediated signal transduction. For example, prenylation of Ras is required for its localization to the plasma membrane and for its normal and oncogenic functions in signal transduction.

Endoplasmic Reticulum Membrane Proteins The normal functioning of the eukaryotic cell requires that all newly synthesized proteins be correctly folded, modified, and delivered to specific intra-and extracellular sites. Newly synthesized membrane and secretory proteins enter a cellular sorting and distribution network during or immediately after synthesis and are routed to specific locations inside and outside of the cell. The initial compartment in this process is the endoplasmic reticulum (ER) where proteins undergo modifications such as glycosylation, disulfide bond formation, and oligomerization. The modified proteins are then transported through a series of membrane-bound compartments which include the various cisternae of the Golgi complex, where further carbohydrate modifications occur. Transport between compartments occurs by means of vesicle budding and fusion. Once within the secretory pathway, proteins do not have to cross a membrane to reach the cell surface.

Although the majority of proteins processed through the ER are transported out of the organelle, some are retained. The signal for retention in the ER in mammalian cells consists of the tetrapeptide sequence, KDEL, located at the carboxyl terminus of resident ER membrane proteins (Munro, S. (1986) Cell 46: 291-300). Proteins containing this sequence leave the ER but are quickly retrieved from the early Golgi cisternae and returned to the ER, while proteins lacking this signal continue through the secretory pathway.

Disruptions in the cellular secretory pathway have been implicated in several human diseases. In familial hypercholesterolemia the low density lipoprotein receptors remain in the ER, rather than moving to the cell surface (Pathak, R. K. (1988) J. Cell Biol. 106: 1831-1841). Altered transport and processing of the P-amyloid precursor protein (TAP) involves the putative vesicle transport protein presenilin and may play a role in early-onset Alzheimer's disease (Levy-Lahad, E. et al. (1995) Science 269: 973-977). Changes in ER-derived calcium homeostasis have been associated with diseases such as cardiomyopathy, cardiac hypertrophy, myotonic dystrophy, Brody disease, Smith-McCort dysplasia, and diabetes mellitus.

Mitochondrial Membrane Proteins The mitochondrial electron transport (or respiratory) chain is a series of three enzyme complexes in the mitochondrial membrane that is responsible for the transport of electrons from NADH to oxygen and the coupling of this oxidation to the synthesis of ATP (oxidative phosphorylation). ATP then provides the primary source of energy for driving the many energy-requiring reactions of a cell.

Most of the protein components of the mitochondrial respiratory chain are the products of

nuclear encoded genes that are imported into the mitochondria, and the remainder are products of mitochondrial genes. Defects and altered expression of enzymes in the respiratory chain are associated with a variety of disease conditions in man, including, for example, neurodegenerative diseases, myopathies, and cancer.

Lymphocyte and Leukocyte Membrane Proteins The B-cell response to antigens is an essential component of the normal immune system.

Mature B cells recognize foreign antigens through B cell receptors (BCR) which are membrane- bound, specific antibodies that bind foreign antigens. The antigen/receptor complex is internalized, and the antigen is proteolytically processed. To generate an efficient response to complex antigens, the BCR, BCR-associated proteins, and T cell response are all required. Proteolytic fragments of the antigen are complexed with major histocompatability complex-11 (MHCII) molecules on the surface of the B cells where the complex can be recognized by T cells. In contrast, macrophages and other lymphoid cells present antigens in association with MHCI molecules to T cells. T cells recognize and are activated by the MHCI-antigen complex through interactions with the T cell receptor/CD3 complex, a T cell-surface multimeric protein located in the plasma membrane. T cells activated by antigen presentation secrete a variety of lymphokines that induce B cell maturation and T cell proliferation, and activate macrophages, which kill target cells.

Leukocytes have a fundamental role in the inflammatory and immune response, and include monocytes/macrophages, mast cells, polymorphonucleoleukocytes, natural killer cells, neutrophils, eosinophils, basophils, and myeloid precursors. Leukocyte membrane proteins include members of the CD antigens, N-CAM, I-CAM, human leukocyte antigen (HLA) class I and HLA class II gene products, immunoglobulins, immunoglobulin receptors, complement, complement receptors, interferons, interferon receptors, interleukin receptors, and chemokine receptors.

Abnormal lymphocyte and leukocyte activity has been associated with acute disorders such as AIDS, immune hypersensitivity, leukemias, leukopenia, systemic lupus, granulomatous disease, and eosinophilia.

Apoptosis-Associated Membrane Proteins A variety of ligands, receptors, enzymes, tumor suppressors, viral gene products, pharmacological agents, and inorganic ions have important positive or negative roles in regulating and implementing the apoptotic destruction of a cell. Although some specific components of the apoptotic pathway have been identified and characterized, many interactions between the proteins involved are undefined, leaving major aspects of the pathway unknown.

A requirement for calcium in apoptosis was previously suggested by studies showing the involvement of calcium levels in DNA cleavage and Fas-mediated cell death (Hewish, D. R. and L. A.

Burgoyne (1973) Biochem. Biophys. Res. Comm. 52: 504-510 ; Vignaux, F. et al. (1995) J. Exp. Med.

181: 781-786; Oshimi, Y. and S. Miyazaki (1995) J. Immunol. 154: 599-609). Other studies show that intracellular calcium concentrations increase when apoptosis is triggered in thymocytes by either T cell receptor cross-linking or by glucocorticoids, and cell death can be prevented by blocking this increase (McConkey, D. J. et al. (1989) J. Immunol. 143: 1801-1806; McConkey, D. J. et al. (1989) Arch. Biochem. Biophys. 269: 365-370). Therefore, membrane proteins such as calcium channels and the Fas receptor are important for the apoptotic response.

Nuclear Hormone Receptors The nuclear hormone receptors, also known as the nuclear receptors or the intracellular receptors, constitute a protein superfamily whose members are both receptors and transcriptional regulators. Nuclear hormone receptors rely on both their receptor function and their transcriptional regulatory function to affect a broad array of biological processes, including development, homeostasis, cell proliferation, and cell differentiation. (Reviewed in Mangelsdorf, D. J. et al. (1995) Cell 83: 835-840; Wen, D. X. and D. P. McDonnell (1995) Curr. Opin. Biotechnol. 6: 582-589; Perlmann, T. and R. M. Evans (1997) Cell 90: 391-397; Tenbaum, S. and A. Baniahmad (1997) Int. J.

Biochem. Cell Biol. 29: 1325-1341; Moras, D. and H. Gronemeyer (1998) Curr. Opin. Cell Biol.

10: 384-391; Willy, P. J. and D. J. Mangelsdorf (1998) in: Hormones and Signaling (ed: B. W.

O'Malley) vol. 1, Academic Press, San Diego CA, pp. 307-358 ; Weatherman, R. V. et al. (1999) Annu. Rev. Biochem. 68: 559-581.) Nuclear hormone receptors as receptors Generally, the term receptor describes a protein that specifically recognizes other molecules.

As receptors, nuclear hormone receptors specifically recognize and bind to their cognate ligands.

Although nuclear hormone receptors are located intracellularly, many receptors are extracellular cell surface proteins which bind extracellular ligands. Such extracellular receptors produce cellular responses affecting growth, differentiation, endocytosis, and the immune response. Other receptors facilitate the selective transport of proteins out of the endoplasmic reticulum and localize enzymes to particular regions of the cell. Transcriptional regulation by nuclear hormone receptors, propagation of cellular signals by extracellular receptors, and transport and localization of proteins by other receptors, all rely upon specific interactions between the receptors and a variety of cellular components. In many cases, the identity of the cognate ligand to which a receptor binds is unknown.

Such receptors are termed orphan receptors. This term also applies to those nuclear hormone receptors which carry out their transcriptional regulatory functions without binding any ligands.

Nuclear hormone receptors as transcriptional regulators Multicellular organisms are comprised of diverse cell types that differ dramatically both in structure and function. The identity of a cell is determined by its characteristic pattern of gene expression, and different cell types express overlapping but distinctive sets of genes throughout

development. Spatial and temporal regulation of gene expression is critical for the control of cell proliferation, cell differentiation, apoptosis, and other processes that contribute to organismal development. As transcriptional regulators, nuclear hormone receptors play key roles in controlling these fundamental biological processes. Other transcriptional regulators affect gene expression in response to extracellular signals that mediate cell-cell communication and that coordinate the activities of different cell types.

In general, transcriptional regulators such as nuclear hormone receptors initiate, activate, repress, or terminate gene transcription by binding to the promoter, enhancer, and upstream regulatory regions of a gene in a sequence-specific manner. However, some transcriptional regulators bind regulatory elements within or downstream of a gene's coding region. Transcriptional regulatory proteins may bind to a specific region of DNA singly, or in a complex with other accessory factors. (Reviewed in Lewin, B. (1990) in: Genes IV, Oxford University Press, New York NY, and Cell Press, Cambridge MA, pp. 554-570.) Mechanism of nuclear hormone receptor function In the unliganded state, a nuclear hormone receptor exists in association with a multiprotein complex of chaperones, including heat shock proteins such as hsp90 and immunophilins such as hsp56. These chaperones maintain the ligand-free receptor in an inactive state which is amenable to binding of free ligand, and prevent the ligand-free receptor from translocating to the nucleus. Upon activation by its cognate ligand, the receptor may form a homodimer or heterodimer which translocates to the nucleus, binds to specific DNA sequences, and exerts its transcriptional regulatory function. In order to effectively carry out its regulatory roles, an activated nuclear hormone receptor dissociates from a histone deacetylase-containing corepressor complex and associates with a histone acetyltransferase-containing coactivator complex (Xu, L. et al. (1999) Curr. Opin. Genet. Dev. 9: 140- 147). The association of the activated receptor with coactivator proteins results in remodeling of chromatin so that it adopts an open transcriptionally active state, providing access to the transcriptional regulatory elements of the activated nuclear receptor (Lemon, B. D. and L. P. Freedman (1999) Curr. Opin. Genet. Dev. 9: 499-504).

Structure of nuclear hormone receptors Nuclear hormone receptors function as signal transducers by converting hormonal signals into transcriptional responses. In general, nuclear hormone receptors consist of a variable amino- terminal domain, a highly conserved DNA-binding domain, and a conserved C-terminal ligand- binding domain. In the steroid-binding nuclear hormone receptors, the amino-terminal domain harbors a trans-activation element termed AF-1. Some nuclear hormone receptors also contain a trans-activation element in the ligand-binding domain termed AF-2. The DNA-binding and ligand- binding domains of nuclear hormone receptors may contain dimerization elements, and the DNA-

binding domain may contain a nuclear localization signal (Weatherman, R. V. et al. (1999) Annu.

Rev. Biochem. 68: 559-581).

The DNA-binding domain of nuclear hormone receptors is composed of two zinc finger motifs which mediate recognition of specific DNA sequences. A zinc finger motif contains periodically spaced cysteine and histidine residues which coordinate Zon". Examples of this sequence pattern include the C2H2-type, C4-type, and C3HC4-type ("RING"finger) zinc fingers, and the PHD domain (Lewin, supra ; Aasland, R. et al. (1995) Trends Biochem. Sci. 20: 56-59). A zinc finger motif contains an oc helix and an antiparallel B sheet whose proximity and conformation are maintained by the zinc ion. Contact with DNA is made by the arginine preceding the a helix and by the second, third, and sixth residues of the a helix. Zinc finger motifs may be repeated in a tandem array within a protein such that the a helix of each zinc finger in the protein makes contact with the major groove of the DNA double helix. This repeated contact between the protein and the DNA produces a strong and specific DNA-protein interaction. The strength and specificity of the interaction can be regulated by the number of zinc finger motifs within the protein. Although zinc fingers were originally identified in DNA-binding proteins as regions that interact directly with DNA, they have since been found in proteins that do not bind to DNA. (See, e. g., Lodish, H. et al. (1995) Molecular Cell Biology, Scientific American Books, New York NY, pp. 447-451.) The ligand-binding domain of nuclear hormone receptors is responsible for binding to ligands, coactivator proteins, and corepressor proteins. This domain is composed of three layers of a helices, with the central layer consisting of two helices containing many hydrophobic side chains (Moras, D. and H. Gronemeyer (1998) Curr. Opin. Cell Biol. 10: 384-391). These two central a helices thus create a hydrophobic pocket which is the site of ligand binding. A ligand bound in this hydrophobic ligand-binding site is completely buried inside the receptor protein and is not exposed to solvent. This suggests that large conformational changes in the ligand-binding domain would accompany binding of a ligand. One of the a helices of the ligand-binding domain provides many of the inter-subunit contacts in dimers of nuclear receptors. This a helix contacts the ligand when it is bound in the ligand-binding pocket, suggesting that ligand binding can affect formation of receptor dimers (Weatherman, R. V. et al. (1999) Annu. Rev. Biochem. 68: 559-581).

Classes of nuclear hormone receptors and associated proteins Nuclear hormone receptors can be grouped into three broad classes: the steroid receptors, the RXR-heterodimeric receptors, and the orphan nuclear hormone receptors. The steroid receptors bind to steroid hormones, and this class includes the androgen receptor, mineralocorticoid receptor, estrogen receptor, glucocorticoid receptor, and progesterone receptor. The RXR-heterodimeric receptors bind to nonsteroid ligands, and this class includes the thyroid hormone receptor, retinoic acid receptor, vitamin D receptor, ecdysone receptor, and peroxisome proliferator activated receptor.

The orphan nuclear hormone receptors include steroidogenic factor 1, nerve growth factor-induced receptor, and X-linked orphan receptor DAX-1.

The steroid hormone receptors are activated upon binding to specific steroid hormones. The conformational change induced by ligand binding leads to dissociation of the receptor from heat shock proteins and formation of receptor homodimers which recognize specific palindromic DNA sequences called hormone response elements (HREs). Upon binding to an BE, a steroid hormone receptor homodimer can regulate the transcription of target genes.

For example, the progesterone receptor (PR) is a steroid hormone receptor which is activated by progesterone, a 4-pregnene-3,20-dione derived from cholesterol which is a critical oscillating component of the female reproductive cycle. These oscillations correlate with anatomical and morphological changes including menstruation and pregnancy. The activities of progesterone are mediated through PR. In the cytoplasm, PR associates with several other proteins and factors known as the PR heterocomplex. This heterocomplex includes heat shock proteins and immunophilins such as hsp70, hsp90, hsp27, p59 (hsp56), p48, and p23 (Johnson, J. L. et al. (1994) Mol. Cell. Biol.

14: 1956-1963). Upon binding progesterone, activated PR translocates to the nucleus, binds to canonical DNA transcriptional elements, and regulates progesterone-regulated genes implicated in differentiation and the cell cycle (Moutsatsou, P and C. E. Sekeris (1997) Ann. N. Y. Acad. Sci.

816: 99-115). The PR antagonist RU 486, which can be used to terminate a pregnancy, is an example of a commercial therapeutic targeted toward a steroid hormone receptor.

The RXR-heterodimeric nuclear receptors are distinguished from the steroid hormone receptors in that members of the former group bind to their target DNA sequences upon formation of heterodimers with retinoid X receptors (RXRs) (Mangelsdorf, D. J. and R. M. Evans (1995) Cell 83: 841-850). Three different isoforms of RXR have been identified (Minucci, S. and K. Ozato (1996) Curr. Opin. Genet. Dev. 6: 567-574). The retinoic acid receptors (RARs) are examples of RXR-heterodimeric nuclear receptors. Retinoic acid (RA) is a biologically active metabolite of vitamin A (retinol), a fat-soluble vitamin found mainly in fish liver oils, liver, egg yolk, butter, and cream. While 9-cis-RA binds to RARs and RXRs, all-trans-RA binds only to RXRs. RAR/RXR heterodimers bind with high affinity to specific DNA sequences known as retinoic acid response elements (RAREs), thus acting as regulators of RA-dependent transcription.

Peroxisome proliferator activated receptors (PPARs) are therapeutically important RXR- heterodimeric nuclear receptors which are induced by fatty acids and eicosanoids. There are three known isotypes of PPAR, each with specific expression patterns, and these PPARs are involved in the regulation of genes involved in systemic homeostatis of glucose and lipids (Kliewer, S. A. and T. M.

Willson (1998) Curr. Opin. Genet. Dev. 8: 576-581; Michalik, L. and W. Wahli (1999) Curr. Opin.

Biotechnol. 10: 564-570). As such, PPARs are therapeutic targets for disorders such as diabetes,

dyslipidemia, and obesity (Smith, S. A. (1996) Pharmacol. Rev. Commun. 8 : 57-64; Willson, T. M. and W. Wahli (1997) Curr. Opin. Chem. Biol. 1: 235-241; Barroso, I. et al. (1999) Nature 402: 880-883).

The orphan nuclear receptors either have no known activating ligand, or can exert their transcriptional regulatory activities without benefit of ligand binding. For example, in Caenorhabditis elegans, the X-chromosome encoded nuclear hormone receptor homologue SEX-1 regulates transcription of the sex determination gene xol-l (Carmi, I. et al. (1998) Nature 396: 168- 173). Rather than relying on ligand binding, SEX-1 acts as a transcriptional regulator in a dose- dependent manner, in effect. controlling sexual differentiation through an X-chromosome-counting mechanism.

Retinoid-related orphan receptor alpha (ROR alpha) is another member of the nuclear receptor superfamily. Mice carrying deletions in the ROR alpha gene demonstrate immune system abnormalities. ROR alphalexpression negatively affects the NF-kappaB signaling pathway apparently through the induction of IkappaB alpha, the major inhibitory protein of the NF-kappaB signaling pathway. These observations have suggested that ROR alphal is a target for treatment of chronic inflammatory diseases, including atherosclerosis and rheumatoid arthritis (Delerive, P. et al.

(2001) EMBO 2: 42-48).

NSD1 is a murine nuclear protein that interacts with the ligand-binding domains (LBDs) of several nuclear receptors. NSD1 contains a SET domain of the subtype represented by the proteins encoded by the Drosophila gene Ashl and the S. cerevisiae gene YJQ8. SET domains are involved in chromatin organization and function and are found in a number of eukaryotic proteins. NSD1 also contains multiple zinc finger-like motifs known as PHD fingers or C4HC3 motifs. NSD1 contains two distinct nuclear receptor-interacting domains, designated NID-''and NID+L. NID-L interacts with the unliganded LBDs of retinoic acid receptors (RAR) and thyroid hormone receptors (TR). NID interacts with the liganded LBDs of RAR, TR, retinoid X receptor (RXR), and estrogen receptor (ER). It is therefore likely that NSD1 plays different roles with respect to transcriptional regulation depending on the presence of bound ligand in the LBDs of target nuclear receptors (Ningwu Huangl, N. et al. (1998) EMBO 17: 3398-3412 and references within).

Some nuclear hormone receptors lack the conventional DNA-binding domain typically associated with the nuclear hormone receptor family. DAX-1 is one such nuclear hormone receptor lacking the conventional DNA-binding domain, and mutations in DAX-1 have been shown to cause X-linked adrenal hypoplasia congenita (Zanaria, E. F. et al. (1994) Nature 372: 635-641). DAX-1 is an orphan nuclear receptor which interacts directly with steroidogenic factor 1 (SF-1) (Ito, M. et al.

(1997) Mol. Cell. Biol. 17: 1476-1483), and DAX-1 is capable of modulating the action of SF-1 in sex-specific gene expression (Nachtigal, M. W. et al. (1998) Cell 445-454). SF-1 is an orphan nuclear receptor which acts as a transcription factor for several steroidogenic enzyme genes in the adrenal

gland and gonads (Lala, D. S. et al. (1992) Mol. Endocrinol. 6: 1249-1258; Lynch, J. P. et al. (1993) Mol. Endocrinol. 7: 776-786; Clemens, J. W. et al. (1994) Endocrinology 134: 1499-1508), and can also regulate several genes expressed in pituitary gonadotrope cells (Barnhart, K. M. and P. L. Mellon (1994) Mol. Endocrinol. 8: 878-885; Ingraham, H. A. et al. (1994) Genes Dev. 8: 2302-2312; Halvorson, L. M. et al. (1996) J. Biol. Chem. 271: 6645-6650; Keri, R. A. and J. H. Nilson (1996) J.

Biol. Chem. 271: 10782-10785).

SF-1 also acts as a potent transactivator of small heterodimer partner (SHP ; short heterodimer partner) (Lee, Y. K. et al. (1999) J. Biol. Chem. 274: 20869-20873). SHP is another example of a nuclear hormone receptor lacking the conventional DNA-binding domain (Seol, W. et al. (1996) Science 272: 1336-1339; Lee, H.-K. et al. (1998) J. Biol. Chem. 273: 14398-14402). SHP interacts with many members of the nuclear hormone receptor family, including retinoid receptors, estrogen receptor, thyroid hormone receptor, and the orphan receptor CAR. SHP acts as an inhibitor of estrogen receptor-mediated transcriptional activation by competing with coactivators for binding to estrogen receptor (Johansson, L. et al. (1999) J. Biol. Chem. 274: 345-353). SHP also inhibits transactivation by the orphan receptor hepatocyte nuclear factor 4, and by retinoid X receptor (Lee, Y. K. et al. (2000) Mol. Cell. Biol. 20: 187-195).

The human thyroid hormone receptor-associated protein (TRAP) complex is a coactivator for nuclear receptors. TRAP appears to be the equivalent of the yeast SRB-and MED-containing cofactor complex (SRB/MED, SMCC) capable of mediating activated transcription in vitro in the absence of TATA-binding (TBP)-associated factors (TAFs). TAFs comprise the subunits of the TFIID that are distinct from TBP. SRB/MED comprises polypeptides identified genetically as suppressors of truncations of the carboxy-tenziaal repeat donaain of the of largest subuhit of RNA polymerase 11 (SRP) that mediate (MED) activated transcription in the absence of TAFs (Ito, M. et al. (1999) Mol. Cell 3: 361-370; reviewed in Wei-Hua Wu, W-H. and Hampsey, M. (1999) Current Biology 9: R606-R609).

Consequences of defective transcription regulation Many neoplastic disorders in humans can be attributed to inappropriate gene expression.

Malignant cell growth may result from either excessive expression of tumor promoting genes or insufficient expression of tumor suppressor genes (Cleary, M. L. (1992) Cancer Surv. 15: 89-104).

Chromosomal translocations may also produce chimeric loci which fuse the coding sequence of one gene with the regulatory regions of a second unrelated gene. Such an arrangement likely results in inappropriate gene transcription, potentially contributing to malignancy.

In addition, the immune system responds to infection or trauma by activating a cascade of events that coordinate the progressive selection, amplification, and mobilization of cellular defense mechanisms. A complex and balanced program of gene activation and repression is involved in this

process. However, hyperactivity of the immune system as a result of improper or insufficient regulation of gene expression may result in considerable tissue or organ damage. This damage is well documented in immunological responses associated with arthritis, allergens, heart attack, stroke, and infections. (See, e. g., Isselbacher et al. (1996) Harrison's Principles of Internal Medicine, 13/e, McGraw Hill, Inc. and Teton Data Systems Software.) Furthermore, the growth of multicellular organisms is based upon the induction and coordination of cell differentiation at the appropriate stages of development. Central to this process is differential gene expression, which confers the distinct identities of cells and tissues throughout the body. Failure to regulate gene expression during development could result in developmental disorders.

T cell Activation Human T cells can be specifically activated by Staphyloccocal exotoxins, resulting in cytokine production and cell proliferation which can lead to septic shock (Muraille, E. et al. (1999) Int. Immunol. 11: 1403-1410). Activation of T cells by Staphyloccocal exotoxins requires the presence of antigen presenting cells (APC) to present the exotoxin molecules to the T cells and to deliver the costimulatory signals required for optimum T cell activation. Although Staphyloccocal exotoxins must be presented to T cells by APC, these molecules do not require processing by APC.

Instead, Staphyloccocal exotoxins directly bind to a non-polymorphic portion of the human major histocompatibility complex (MHC) class II molecules, thus bypassing the need for capture, cleavage, and binding of the peptides to the polymorphic antigenic groove of the MHC class IN molecules.

Expression profiling Array technology can provide a simple way to explore the expression of a single polymorphic gene or the expression profile of a large number of related or unrelated genes. When the expression of a single gene is examined, arrays are employed to detect the expression of a specific gene or its variants. When an expression profile is examined, arrays provide a platform for identifying genes that are tissue specific, are affected by a substance being tested in a toxicology assay, are part of a signaling cascade, carry out housekeeping functions, or are specifically related to a particular genetic predisposition, condition, disease, or disorder.

The discovery of new receptors and membrane-associated proteins and the polynucleotides encoding them satisfies a need in the art by providing new compositions which are useful in the diagnosis, prevention, and treatment of cell proliferative, autoimmune/inflammatory, neurological, metabolic, developmental, endocrine, cardiovascular, reproductive, gastrointestinal, metabolic, genetic, and lipid metabolism disorders, cancer, and viral infections, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of receptors and membrane-associated proteins.

SUMMARY OF THE INVENTION The invention features purified polypeptides, receptors and membrane-associated proteins, referred to collectively as"REMAP"and individually as"REMAP-1,""REMAP-2,""REMAP-3," <BR> <BR> <BR> "REMAP-4,""REMAP-5,""REMAP-6,""REMAP-7,""REMAP-8,""REMAP-9," "REMAP-10,"<BR> <BR> <BR> "REMAP-11,""REMAP-12,""REMAP-13,""REMAP-14,""REMAP-15,""REMA P-16,"<BR> <BR> <BR> "REMAP-17,""REMAP-18,""REMAP-19,""REMAP-20,""REMAP-21,""REMA P-22," "REMAP-23,""REMAP-24,""REMAP-25,"and"REMAP-26."In one aspect, the invention provides an isolated polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. In one alternative, the invention provides an isolated polypeptide comprising the amino acid sequence of SEQ ID NO: 1-26.

The invention further provides an isolated polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26.

In one alternative, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ ID NO : 1-26. In another alternative, the polynucleotide is selected from the group consisting of SEQ ID NO : 27-52.

Additionally, the invention provides a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. In one alternative, the invention provides a cell transformed with the recombinant polynucleotide. In another alternative, the invention provides a transgenic organism comprising the recombinant polynucleotide.

The invention also provides a method for producing a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. The method comprises a) culturing a cell under conditions suitable for expression of the polypeptide, wherein said cell is transformed with a recombinant polynucleotide comprising a promoter sequence operably linked to a polynucleotide encoding the polypeptide, and b) recovering the polypeptide so expressed.

Additionally, the invention provides an isolated antibody which specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26.

The invention further provides an isolated polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). In one alternative, the polynucleotide comprises at least 60 contiguous nucleotides.

Additionally, the invention provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ IID NO : 27-52, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) hybridizing the sample with a probe comprising at least 20 contiguous nucleotides comprising a sequence complementary to said target polynucleotide in the sample, and which probe specifically hybridizes to said target polynucleotide, under conditions whereby a

hybridization complex is formed between said probe and said target polynucleotide or fragments thereof, and b) detecting the presence or absence of said hybridization complex, and optionally, if present, the amount thereof. In one alternative, the probe comprises at least 60 contiguous nucleotides.

The invention further provides a method for detecting a target polynucleotide in a sample, said target polynucleotide having a sequence of a polynucleotide selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, c) a polynucleotide complementary to the polynucleotide of a), d) a polynucleotide complementary to the polynucleotide of b), and e) an RNA equivalent of a)-d). The method comprises a) amplifying said target polynucleotide or fragment thereof using polymerase chain reaction amplification, and b) detecting the presence or absence of said amplified target polynucleotide or fragment thereof, and, optionally, if present, the amount thereof.

The invention further provides a composition comprising an effective amount of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and a pharmaceutically acceptable excipient. In one embodiment, the composition comprises an amino acid sequence selected from the group consisting of SEQ ID NO : 1- 26. The invention additionally provides a method of treating a disease or condition associated with decreased expression of functional REMAP, comprising administering to a patient in need of such treatment the composition.

The invention also provides a method for screening a compound for effectiveness as an agonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-26. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample. In one alternative, the

invention provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with decreased expression of functional REMAP, comprising administering to a patient in need of such treatment the composition.

Additionally, the invention provides a method for screening a compound for effectiveness as an antagonist of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample. In one alternative, the invention provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient. In another alternative, the invention provides a method of treating a disease or condition associated with overexpression of functional REMAP, comprising administering to a patient in need of such treatment the composition.

The invention further provides a method of screening for a compound that specifically binds to a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. The method comprises a) combining the polypeptide with at least one test compound under suitable conditions, and b) detecting binding of the polypeptide to the test compound, thereby identifying a compound that specifically binds to the polypeptide.

The invention further provides a method of screening for a compound that modulates the activity of a polypeptide selected from the group consisting of a) a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-26. The method comprises a) combining the polypeptide with at least

one test compound under conditions permissive for the activity of the polypeptide, b) assessing the activity of the polypeptide in the presence of the test compound, and c) comparing the activity of the polypeptide in the presence of the test compound with the activity of the polypeptide in the absence of the test compound, wherein a change in the activity of the polypeptide in the presence of the test compound is indicative of a compound that modulates the activity of the polypeptide.

The invention further provides a method for screening a compound for effectiveness in altering expression of a target polynucleotide, wherein said target polynucleotide comprises a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, the method comprising a) exposing a sample comprising the target polynucleotide to a compound, b) detecting altered expression of the target polynucleotide, and c) comparing the expression of the target polynucleotide in the presence of varying amounts of the compound and in the absence of the compound.

The invention further provides a method for assessing toxicity of a test compound, said method comprising a) treating a biological sample containing nucleic acids with the test compound; b) hybridizing the nucleic acids of the treated biological sample with a probe comprising at least 20 contiguous nucleotides of a polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, iii) a polynucleotide having a sequence complementary to i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Hybridization occurs under conditions whereby a specific hybridization complex is formed between said probe and a target polynucleotide in the biological sample, said target polynucleotide selected from the group consisting of i) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 27-52, iii) a polynucleotide complementary to the polynucleotide of i), iv) a polynucleotide complementary to the polynucleotide of ii), and v) an RNA equivalent of i)-iv). Alternatively, the target polynucleotide comprises a fragment of a polynucleotide sequence selected from the group consisting of i)-v) above; c) quantifying the amount of hybridization complex; and d) comparing the amount of hybridization complex in the treated biological sample with the amount of hybridization complex in an untreated biological sample, wherein a difference in the amount of hybridization complex in the treated biological sample is indicative of toxicity of the test compound.

BRIEF DESCRIPTION OF THE TABLES

Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the present invention.

Table 2 shows the GenBank identification number and annotation of the nearest GenBank homolog, and the PROTEOME database identification numbers and annotations of PROTEOME database homologs, for polypeptides of the invention. The probability scores for the matches between each polypeptide and its homolog (s) are also shown.

Table 3 shows structural features of polypeptide sequences of the invention, including predicted motifs and domains, along with the methods, algorithms, and searchable databases used for analysis of the polypeptides.

Table 4 lists the cDNA and/or genomic DNA fragments which were used to assemble polynucleotide sequences of the invention, along with selected fragments of the polynucleotide sequences.

Table 5 shows the representative cDNA library for polynucleotides of the invention.

Table 6 provides an appendix which describes the tissues and vectors used for construction of the cDNA libraries shown in Table 5.

Table 7 shows the tools, programs, and algorithms used to analyze the polynucleotides and polypeptides of the invention, along with applicable descriptions, references, and threshold parameters.

DESCRIPTION OF THE INVENTION Before the present proteins, nucleotide sequences, and methods are described, it is understood that this invention is not limited to the particular machines, materials and methods described, as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims.

It must be noted that as used herein and in the appended claims, the singular forms"a,""an," and"the"include plural reference unless the context clearly dictates otherwise. Thus, for example, a reference to"a host cell"includes a plurality of such host cells, and a reference to"an antibody"is a reference to one or more antibodies and equivalents thereof known to those skilled in the art, and so forth.

Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs.

Although any machines, materials, and methods similar or equivalent to those described herein can be used to practice or test the present invention, the preferred machines, materials and methods are now described. All publications mentioned herein are cited for the purpose of describing and disclosing

the cell lines, protocols, reagents and vectors which are reported in the publications and which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

DEFINITIONS "REMAP"refers to the amino acid sequences of substantially purified REMAP obtained from any species, particularly a mammalian species, including bovine, ovine, porcine, murine, equine, and human, and from any source, whether natural, synthetic, semi-synthetic, or recombinant.

The term"agonist"refers to a molecule which intensifies or mimics the biological activity of REMAP. Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of REMAP either by directly interacting with REMAP or by acting on components of the biological pathway in which REMAP participates.

An"allelic variant"is an alternative form of the gene encoding REMAP. Allelic variants may result from at least one mutation in the nucleic acid sequence and may result in altered mRNAs or in polypeptides whose structure or function may or may not be altered. A gene may have none, one, or many allelic variants of its naturally occurring form. Common mutational changes which give rise to allelic variants are generally ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes may occur alone, or in combination with the others, one or more times in a given sequence.

"Altered"nucleic acid sequences encoding REMAP include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as REMAP or a polypeptide with at least one functional characteristic of REMAP. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding REMAP, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding REMAP. The encoded protein may also be"altered,"and may contain deletions, insertions, or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent REMAP. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of REMAP is retained. For example, negatively charged amino acids may include aspartic acid and glutamic acid, and positively charged amino acids may include lysine and arginine. Amino acids with uncharged polar side chains having similar hydrophilicity values may include: asparagine and glutamine; and serine and threonine.

Amino acids with uncharged side chains having similar hydrophilicity values may include: leucine, isoleucine, and valine; glycine and alanine; and phenylalanine and tyrosine.

The terms"amino acid"and"amino acid sequence"refer to an oligopeptide, peptide,

polypeptide, or protein sequence, or a fragment of any of these, and to naturally occurring or synthetic molecules. Where"amino acid sequence"is recited to refer to a sequence of a naturally occurring protein molecule,"amino acid sequence"and like terms are not meant to limit the amino acid sequence to the complete native amino acid sequence associated with the recited protein molecule.

"Amplification"relates to the production of additional copies of a nucleic acid sequence.

Amplification is generally carried out using polymerase chain reaction (PCR) technologies well known in the art.

The term"antagonist"refers to a molecule which inhibits or attenuates the biological activity of REMAP. Antagonists may include proteins such as antibodies, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of REMAP either by directly interacting with REMAP or by acting on components of the biological pathway in which REMAP participates.

The term"antibody"refers to intact immunoglobulin molecules as well as to fragments thereof, such as Fab, F (ab') 2, and Fv fragments, which are capable of binding an epitopic determinant.

Antibodies that bind REMAP polypeptides can be prepared using intact polypeptides or using fragments containing small peptides of interest as the immunizing antigen. The polypeptide or oligopeptide used to immunize an animal (e. g., a mouse, a rat, or a rabbit) can be derived from the translation of RNA, or synthesized chemically, and can be conjugated to a carrier protein if desired.

Commonly used carriers that are chemically coupled to peptides include bovine serum albumin, thyroglobulin, and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal.

The term"antigenic determinant"refers to that region of a molecule (i. e., an epitope) that makes contact with a particular antibody. When a protein or a fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to antigenic determinants (particular regions or three-dimensional structures on the protein). An antigenic determinant may compete with the intact antigen (i. e., the immunogen used to elicit the immune response) for binding to an antibody.

The term"aptamer"refers to a nucleic acid or oligonucleotide molecule that binds to a specific molecular target. Aptamers are derived from an in vitro evolutionary process (e. g., SELEX (Systematic Evolution of Ligands by EXponential Enrichment), described in U. S. Patent No.

5,270,163), which selects for target-specific aptamer sequences from large combinatorial libraries.

Aptamer compositions may be double-stranded or single-stranded, and may include deoxyribonucleotides, ribonucleotides, nucleotide derivatives, or other nucleotide-like molecules.

The nucleotide components of an aptamer may have modified sugar groups (e. g., the 2'-OH group of a ribonucleotide may be replaced by 2'-F or 2'-NH2), which may improve a desired property, e. g.,

resistance to nucleases or longer lifetime in blood. Aptamers may be conjugated to other molecules, e. g., a high molecular weight carrier to slow clearance of the aptamer from the circulatory system.

Aptamers may be specifically cross-linked to their cognate ligands, e. g., by photo-activation of a cross-linker. (See, e. g., Brody, E. N. and L. Gold (2000) J. Biotechnol. 74: 5-13.) The term"intramer"refers to an aptamer which is expressed in vivo. For example, a vaccinia virus-based RNA expression system has been used to express specific RNA aptamers at high levels in the cytoplasm of leukocytes (Blind, M. et al. (1999) Proc. Natl. Acad. Sci. USA 96: 3606-3610).

The term"spiegelmer"refers to an aptamer which includes L-DNA, L-RNA, or other left- handed nucleotide derivatives or nucleotide-like molecules. Aptamers containing left-handed nucleotides are resistant to degradation by naturally occurring enzymes, which normally act on substrates containing right-handed nucleotides.

The term"antisense"refers to any composition capable of base-pairing with the"sense" (coding) strand of a specific nucleic acid sequence. Antisense compositions may include DNA; RNA; peptide nucleic acid (PNA); oligonucleotides having modified backbone linkages such as phosphorothioates, methylphosphonates, or benzylphosphonates; oligonucleotides having modified sugar groups such as 2'-methoxyethyl sugars or 2'-methoxyethoxy sugars ; or oligonucleotides having modified bases such as 5-methyl cytosine, 2'-deoxyuracil, or 7-deaza-2'-deoxyguanosine. Antisense molecules may be produced by any method including chemical synthesis or transcription. Once introduced into a cell, the complementary antisense molecule base-pairs with a naturally occurring nucleic acid sequence produced by the cell to form duplexes which block either transcription or translation. The designation"negative"or"minus"can refer to the antisense strand, and the designation"positive"or"plus"can refer to the sense strand of a reference DNA molecule.

The term"biologically active"refers to a protein having structural, regulatory, or biochemical functions of a naturally occurring molecule. Likewise,"immunologically active"or"immunogenic" refers to the capability of the natural, recombinant, or synthetic REMAP, or of any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and to bind with specific antibodies.

"Complementary"describes the relationship between two single-stranded nucleic acid sequences that anneal by base-pairing. For example, 5'-AGT-3'pairs with its complement, 3'-TCA-5'.

A"composition comprising a given polynucleotide sequence"and a"composition comprising a given amino acid sequence"refer broadly to any composition containing the given polynucleotide or amino acid sequence. The composition may comprise a dry formulation or an aqueous solution.

Compositions comprising polynucleotide sequences encoding REMAP or fragments of REMAP may be employed as hybridization probes. The probes may be stored in freeze-dried form and may be

associated with a stabilizing agent such as a carbohydrate. In hybridizations, the probe may be deployed in an aqueous solution containing salts (e. g., NaCI), detergents (e. g., sodium dodecyl sulfate; SDS), and other components (e. g., Denhardt's solution, dry milk, salmon sperm DNA, etc.).

"Consensus sequence"refers to a nucleic acid sequence which has been subjected to repeated DNA sequence analysis to resolve uncalled bases, extended using the XL-PCR kit (Applied Biosystems, Foster City CA) in the 5'and/or the 3'direction, and resequenced, or which has been assembled from one or more overlapping cDNA, EST, or genomic DNA fragments using a computer program for fragment assembly, such as the GELVIEW fragment assembly system (GCG, Madison WI) or Phrap (University of Washington, Seattle WA). Some sequences have been both extended and assembled to produce the consensus sequence.

"Conservative amino acid substitutions"are those substitutions that are predicted to least interfere with the properties of the original protein, i. e., the structure and especially the function of the protein is conserved and not significantly changed by such substitutions. The table below shows amino acids which may be substituted for an original amino acid in a protein and which are regarded as conservative amino acid substitutions.

Original Residue Conservative Substitution Ala Gly, Ser Arg His, Lys Asn Asp, Gln, His Asp Asn, Glu Cys Ala, Ser Gln Asn, Glu, His Glu Asp, Gln, His Gly Ala His Asn, Arg, Gln, Glu Ile Leu, Val Leu Ile, Val Lys Arg, Gln, Glu Met Leu, Ile Phe His, Met, Leu, Trp, Tyr Ser Cys, Thr Thr Ser, Val Trp Phe, Tyr Tyr His, Phe, Trp Val Ile, Leu, Thr Conservative amino acid substitutions generally maintain (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a beta sheet or alpha helical conformation, (b) the charge or hydrophobicity of the molecule at the site of the substitution, and/or (c) the bulk of the side chain.

A"deletion"refers to a change in the amino acid or nucleotide sequence that results in the absence of one or more amino acid residues or nucleotides.

The term"derivative"refers to a chemically modified polynucleotide or polypeptide.

Chemical modifications of a polynucleotide can include, for example, replacement of hydrogen by an alkyl, acyl, hydroxyl, or amino group. A derivative polynucleotide encodes a polypeptide which retains at least one biological or immunological function of the natural molecule. A derivative polypeptide is one modified by glycosylation, pegylation, or any similar process that retains at least one biological or immunological function of the polypeptide from which it was derived.

A"detectable label"refers to a reporter molecule or enzyme that is capable of generating a measurable signal and is covalently or noncovalently joined to a polynucleotide or polypeptide.

"Differential expression"refers to increased or upregulated; or decreased, downregulated, or absent gene or protein expression, determined by comparing at least two different samples. Such comparisons may be carried out between, for example, a treated and an untreated sample, or a diseased and a normal sample.

"Exon shuffling"refers to the recombination of different coding regions (exons). Since an exon may represent a structural or functional domain of the encoded protein, new proteins may be assembled through the novel reassortment of stable substructures, thus allowing acceleration of the evolution of new protein functions.

A"fragment"is a unique portion of REMAP or the polynucleotide encoding REMAP which is identical in sequence to but shorter in length than the parent sequence. A fragment may comprise up to the entire length of the defined sequence, minus one nucleotide/amino acid residue. For example, a fragment may comprise from 5 to 1000 contiguous nucleotides or amino acid residues. A fragment used as a probe, primer, antigen, therapeutic molecule, or for other purposes, may be at least 5,10,15,16,20,25,30,40,50,60,75,100,150,250 or at least 500 contiguous nucleotides or amino acid residues in length. Fragments may be preferentially selected from certain regions of a molecule.

For example, a polypeptide fragment may comprise a certain length of contiguous amino acids selected from the first 250 or 500 amino acids (or first 25% or 50%) of a polypeptide as shown in a certain defined sequence. Clearly these lengths are exemplary, and any length that is supported by the specification, including the Sequence Listing, tables, and figures, may be encompassed by the present embodiments.

A fragment of SEQ ID NO : 27-52 comprises a region of unique polynucleotide sequence that specifically identifies SEQ ID NO : 27-52, for example, as distinct from any other sequence in the genome from which the fragment was obtained. A fragment of SEQ ID NO : 27-52 is useful, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ ID NO : 27-52 from related polynucleotide sequences. The precise length of a fragment of SEQ ID NO : 27-52 and the region of SEQ ID NO : 27-52 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.

A fragment of SEQ ID NO: 1-26 is encoded by a fragment of SEQ ID NO : 27-52. A fragment of SEQ ID NO : 1-26 comprises a region of unique amino acid sequence that specifically identifies SEQ ID NO : 1-26. For example, a fragment of SEQ ID NO : 1-26 is useful as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID NO: 1-26. The precise length of a fragment of SEQ ID NO: 1-26 and the region of SEQ ID NO: 1-26 to which the fragment corresponds are routinely determinable by one of ordinary skill in the art based on the intended purpose for the fragment.

A"full length"polynucleotide sequence is one containing at least a translation initiation codon (e. g., methionine) followed by an open reading frame and a translation termination codon. A "full length"polynucleotide sequence encodes a"full length"polypeptide sequence.

"Homology"refers to sequence similarity or, interchangeably, sequence identity, between two or more polynucleotide sequences or two or more polypeptide sequences.

The terms'percent identity"and"% identity,"as applied to polynucleotide sequences, refer to the percentage of residue matches between at least two polynucleotide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.

Percent identity between polynucleotide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN version 3.12e sequence alignment program. This program is part of the LASERGENE software package, a suite of molecular biological analysis programs (DNASTAR, Madison WI). CLUSTAL V is described in Higgins, D. G. and P. M. Sharp (1989) CABIOS 5: 151-153 and in Higgins, D. G. et al. (1992) CABIOS 8: 189-191. For pairwise alignments of polynucleotide sequences, the default parameters are set as follows: Ktuple=2, gap penalty=5, window=4, and"diagonals saved"=4. The"weighted"residue weight table is selected as the default. Percent identity is reported by CLUSTAL V as the"percent similarity"between aligned polynucleotide sequences.

Alternatively, a suite of commonly used and freely available sequence comparison algorithms is provided by the National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) (Altschul, S. F. et al. (1990) J. Mol. Biol. 215: 403-410), which is available from several sources, including the NCBI, Bethesda, MD, and on the Internet at http ://www. ncbi. nlm. nih. gov/BLAST/. The BLAST software suite includes various sequence analysis programs including"blastn,"that is used to align a known polynucleotide sequence with other polynucleotide sequences from a variety of databases. Also available is a tool called"BLAST 2 Sequences"that is used for direct pairwise comparison of two nucleotide sequences."BLAST 2 Sequences"can be accessed and used interactively at http ://www. ncbi. nlm. nih. gov/gorf/bl2. html.

The"BLAST 2 Sequences"tool can be used for both blastn and blastp (discussed below). BLAST programs are commonly used with gap and other parameters set to default settings. For example, to compare two nucleotide sequences, one may use blastn with the"BLAST 2 Sequences"tool Version 2.0.12 (April-21-2000) set at default parameters. Such default parameters may be, for example: Matrix : BLOSUM62 Reward for match : 1 Penalty for mismatch :-2 Open Gap : 5 and Extension Gap : 2 penalties Gap x drop-off.-50 Expect : 10 Word Size : 11 Filter: on Percent identity may be measured over the length of an entire defined sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined sequence, for instance, a fragment of at least 20, at least 30, at least 40, at least 50, at least 70, at least 100, or at least 200 contiguous nucleotides. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures, or Sequence Listing, may be used to describe a length over which percentage identity may be measured.

Nucleic acid sequences that do not show a high degree of identity may nevertheless encode similar amino acid sequences due to the degeneracy of the genetic code. It is understood that changes in a nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acid sequences that all encode substantially the same protein.

The phrases"percent identity"and"% identity,"as applied to polypeptide sequences, refer to the percentage of residue matches between at least two polypeptide sequences aligned using a standardized algorithm. Methods of polypeptide sequence alignment are well-known. Some alignment methods take into account conservative amino acid substitutions. Such conservative substitutions, explained in more detail above, generally preserve the charge and hydrophobicity at the site of substitution, thus preserving the structure (and therefore function) of the polypeptide.

Percent identity between polypeptide sequences may be determined using the default parameters of the CLUSTAL V algorithm as incorporated into the MEGALIGN version 3.12e sequence alignment program (described and referenced above). For pairwise alignments of polypeptide sequences using CLUSTAL V, the default parameters are set as follows: Ktuple=l, gap penalty=3, window=5, and"diagonals saved"=5. The PAM250 matrix is selected as the default residue weight table. As with polynucleotide alignments, the percent identity is reported by

CLUSTAL V as the"percent similarity"between aligned polypeptide sequence pairs.

Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise comparison of two polypeptide sequences, one may use the"BLAST 2 Sequences"tool Version 2.0.12 (April-21-2000) with blastp set at default parameters. Such default parameters may be, for example: Matrix : BLOSUM62 Open Gap : 11 and Extension Gap : 1 penalties Gap x drop-of : 50 Expect : 10 Word Size: 3 Filter: on Percent identity may be measured ove rthe length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ ID number, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide sequence, for instance, a fragment of at least 15, at least 20, at least 30, at least 40, at least 50, at least 70 or at least 150 contiguous residues. Such lengths are exemplary only, and it is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.

"Human artificial chromosomes" (HACs) are linear microchromosomes which may contain DNA sequences of about 6 kb to 10 Mb in size and which contain all of the elements required for chromosome replication, segregation and maintenance.

The term"humanized antibody"refers to an antibody molecule in which the amino acid sequence in the non-antigen binding regions has been altered so that the antibody more closely resembles a human antibody, and still retains its original binding ability.

"Hybridization"refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of complementarity.

Specific hybridization complexes form under permissive annealing conditions and remain hybridized after the"washing"step (s). The washing step (s) is particularly important in determining the stringency of the hybridization process, with more stringent conditions allowing less non-specific binding, i. e., binding between pairs of nucleic acid strands that are not perfectly matched. Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve the desired stringency, and therefore hybridization specificity.

Permissive annealing conditions occur, for example, at 68°C in the presence of about 6 x SSC, about

1 % (w/v) SDS, and about 100 yg/ml sheared, denatured salmon sperm DNA.

Generally, stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. An equation for calculating T. and conditions for nucleic acid hybridization are well known and can be found in Sambrook, J. et al.

(1989) Molecular Cloning : A Labora ! M Manual, 2"ed., vol. 1-3, Cold Spring Harbor Press, Plainview NY; specifically see volume 2, chapter 9.

High stringency conditions for hybridization between polynucleotides of the present invention include wash conditions of 68°C in the presence of about 0.2 x SSC and about 0.1 % SDS, for 1 hour. Alternatively, temperatures of about 65°C, 60°C, 55°C, or 42°C may be used. SSC concentration may be varied from about 0.1 to 2 x SSC, with SDS being present at about 0.1 %.

Typically, blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared and denatured salmon sperm DNA at about 100-200 yg/ml. Organic solvent, such as formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for RNA: DNA hybridizations. Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art. Hybridization, particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is strongly indicative of a similar role for the nucleotides and their encoded polypeptides.

The term"hybridization complex"refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (e. g., Cot or Rot analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (e. g., paper, membranes, filters, chips, pins or glass slides, or any other appropriate substrate to which cells or their nucleic acids have been fixed).

The words"insertion"and"addition"refer to changes in an amino acid or nucleotide sequence resulting in the addition of one or more amino acid residues or nucleotides, respectively.

"Immune response"can refer to conditions associated with inflammation, trauma, immune disorders, or infectious or genetic disease, etc. These conditions can be characterized by expression of various factors, e. g., cytokines, chemokines, and other signaling molecules, which may affect cellular and systemic defense systems.

An"immunogenic fragment"is a polypeptide or oligopeptide fragment of REMAP which is capable of eliciting an immune response when introduced into a living organism, for example, a mammal. The term"immunogenic fragment"also includes any polypeptide or oligopeptide fragment

of REMAP which is useful in any of the antibody production methods disclosed herein or known in the art.

The term"microarray"refers to an arrangement of a plurality of polynucleotides, polypeptides, or other chemical compounds on a substrate.

The terms"element"and"array element"refer to a polynucleotide, polypeptide, or other chemical compound having a unique and defined position on a microarray.

The term"modulate"refers to a change in the activity of REMAP. For example, modulation may cause an increase or a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of REMAP.

The phrases"nucleic acid"and"nucleic acid sequence"refer to a nucleotide, oligonucleotide, polynucleotide, or any fragment thereof. These phrases also refer to DNA or RNA of genomic or synthetic origin which may be single-stranded or double-stranded and may represent the sense or the antisense strand, to peptide nucleic acid (PNA), or to any DNA-like or RNA-like material.

"Operably linked"refers to the situation in which a first nucleic acid sequence is placed in a functional relationship with a second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Operably linked DNA sequences may be in close proximity or contiguous and, where necessary to join two protein coding regions, in the same reading frame.

"Peptide nucleic acid" (PNA) refers to an antisense molecule or anti-gene agent which comprises an oligonucleotide of at least about 5 nucleotides in length linked to a peptide backbone of amino acid residues ending in lysine. The terminal lysine confers solubility to the composition.

PNAs preferentially bind complementary single stranded DNA or RNA and stop transcript elongation, and may be pegylated to extend their lifespan in the cell.

"Post-translational modification"of an REMAP may involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and other modifications known in the art. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cell type depending on the enzymatic milieu of REMAP.

"Probe"refers to nucleic acid sequences encoding REMAP, their complements, or fragments thereof, which are used to detect identical, allelic or related nucleic acid sequences. Probes are isolated oligonucleotides or polynucleotides attached to a detectable label or reporter molecule.

Typical labels include radioactive isotopes, ligands, chemiluminescent agents, and enzymes.

"Primers"are short nucleic acids, usually DNA oligonucleotides, which may be annealed to a target polynucleotide by complementary base-pairing. The primer may then be extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for amplification (and identification) of a nucleic acid sequence, e. g., by the polymerase chain reaction (PCR).

Probes and primers as used in the present invention typically comprise at least 15 contiguous nucleotides of a known sequence. In order to enhance specificity, longer probes and primers may also be employed, such as probes and primers that comprise at least 20,25,30,40,50,60,70,80,90,100, or at least 150 consecutive nucleotides of the disclosed nucleic acid sequences. Probes and primers may be considerably longer than these examples, and it is understood that any length supported by the specification, including the tables, figures, and Sequence Listing, may be used.

Methods for preparing and using probes and primers are described in the references, for example Sambrook, J. et al. (1989) Molecular Cloning : A Laboratory Manual, 2nd ed., vol. 1-3, Cold Spring Harbor Press, Plainview NY; Ausubel, F. M. et al. (1987) Current Protocols in Molecular Biology. Greene Publ. Assoc. & Wiley-Intersciences, New York NY; Innis, M. et al. (1990) PCR Protocols. A Guide to Methods and Applications, Academic Press, San Diego CA. PCR primer pairs can be derived from a known sequence, for example, by using computer programs intended for that purpose such as Primer (Version 0.5,1991, Whitehead Institute for Biomedical Research, Cambridge MA).

Oligonucleotides for use as primers are selected using software known in the art for such purpose. For example, OLIGO 4.06 software is useful for the selection of PCR primer pairs of up to 100 nucleotides each, and for the analysis of oligonucleotides and larger polynucleotides of up to 5,000 nucleotides from an input polynucleotide sequence of up to 32 kilobases. Similar primer selection programs have incorporated additional features for expanded capabilities. For example, the PrimOU primer selection program (available to the public from the Genome Center at University of Texas South West Medical Center, Dallas TX) is capable of choosing specific primers from megabase sequences and is thus useful for designing primers on a genome-wide scope. The Primer3 primer selection program (available to the public from the Whitehead Institute/MIT Center for Genome Research, Cambridge MA) allows the user to input a"mispriming library,"in which sequences to avoid as primer binding sites are user-specified. Primer3 is useful, in particular, for the selection of oligonucleotides for microarrays. (The source code for the latter two primer selection programs may also be obtained from their respective sources and modified to meet the user's specific needs.) The PrimeGen program (available to the public from the UK Human Genome Mapping Project Resource Centre, Cambridge UK) designs primers based on multiple sequence alignments, thereby allowing selection of primers that hybridize to either the most conserved or least conserved regions of aligned nucleic acid sequences. Hence, this program is useful for identification of both unique and conserved oligonucleotides and polynucleotide fragments. The oligonucleotides and polynucleotide fragments identified by any of the above selection methods are useful in hybridization technologies, for example, as PCR or sequencing primers, microarray elements, or specific probes to identify fully or partially complementary polynucleotides in a sample of nucleic acids. Methods of

oligonucleotide selection are not limited to those described above.

A"recombinant nucleic acid"is a sequence that is not naturally occurring or has a sequence that is made by an artificial combination of two or more otherwise separated segments of sequence.

This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, e. g., by genetic engineering techniques such as those described in Sambrook, supra. The term recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion of a portion of the nucleic acid. Frequently, a recombinant nucleic acid may include a nucleic acid sequence operably linked to a promoter sequence. Such a recombinant nucleic acid may be part of a vector that is used, for example, to transform a cell.

Alternatively, such recombinant nucleic acids may be part of a viral vector, e. g., based on a vaccinia virus, that could be use to vaccinate a mammal wherein the recombinant nucleic acid is expressed, inducing a protective immunological response in the mammal.

A"regulatory element"refers to a nucleic acid sequence usually derived from untranslated regions of a gene and includes enhancers, promoters, introns, and 5'and 3'untranslated regions (UTRs). Regulatory elements interact with host or viral proteins which control transcription, translation, or RNA stability.

"Reporter molecules"are chemical or biochemical moieties used for labeling a nucleic acid, amino acid, or antibody. Reporter molecules include radionuclides; enzymes; fluorescent, chemiluminescent, or chromogenic agents; substrates; cofactors; inhibitors; magnetic particles; and other moieties known in the art.

An"RNA equivalent,"in reference to a DNA sequence, is composed of the same linear sequence of nucleotides as the reference DNA sequence with the exception that all occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.

The term"sample"is used in its broadest sense. A sample suspected of containing REMAP, nucleic acids encoding REMAP, or fragments thereof may comprise a bodily fluid; an extract from a cell, chromosome, organelle, or membrane isolated from a cell; a cell; genomic DNA, RNA, or cDNA, in solution or bound to a substrate; a tissue; a tissue print; etc.

The terms"specific binding"and"specifically binding"refer to that interaction between a protein or peptide and an agonist, an antibody, an antagonist, a small molecule, or any natural or synthetic binding composition. The interaction is dependent upon the presence of a particular structure of the protein, e. g., the antigenic determinant or epitope, recognized by the binding molecule. For example, if an antibody is specific for epitope"A,"the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A

and the antibody will reduce the amount of labeled A that binds to the antibody.

The term"substantially purified"refers to nucleic acid or amino acid sequences that are removed from their natural environment and are isolated or separated, and are at least 60% free, preferably at least 75% free, and most preferably at least 90% free from other components with which they are naturally associated.

A"substitution"refers to the replacement of one or more amino acid residues or nucleotides by different amino acid residues or nucleotides, respectively.

"Substrate"refers to any suitable rigid or semi-rigid support including membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, tubing, plates, polymers, microparticles and capillaries. The substrate can have a variety of surface forms, such as wells, trenches, pins, channels and pores, to which polynucleotides or polypeptides are bound.

A"transcript image"or"expression profile"refers to the collective pattern of gene expression by a particular cell type or tissue under given conditions at a given time.

"Transformation"describes a process by which exogenous DNA is introduced into a recipient cell. Transformation may occur under natural or artificial conditions according to various methods well known in the art, and may rely on any known method for the insertion of foreign nucleic acid sequences into a prokaryotic or eukaryotic host cell. The method for transformation is selected based on the type of host cell being transformed and may include, but is not limited to, bacteriophage or viral infection, electroporation, heat shock, lipofection, and particle bombardment. The term "transformed cells"includes stably transformed cells in which the inserted DNA is capable of replication either as an autonomously replicating plasmid or as part of the host chromosome, as well as transiently transformed cells which express the inserted DNA or RNA for limited periods of time.

A"transgenic organism,"as used herein, is any organism, including but not limited to animals and plants, in which one or more of the cells of the organism contains heterologous nucleic acid introduced by way of human intervention, such as by transgenic techniques well known in the art. The nucleic acid is introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. In one alternative, the nucleic acid can be introduced by infection with a recombinant viral vector, such as a lentiviral vector (Lois, C. et al. (2002) Science 295: 868-872). The term genetic manipulation does not include classical cross-breeding, or ion vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. The transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals. The isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation. Techniques for transferring the DNA of the present invention into such organisms are widely known and provided

in references such as Sambrook et al. (1989), supra.

A"variant"of a particular nucleic acid sequence is defined as a nucleic acid sequence having at least 40% sequence identity to the particular nucleic acid sequence over a certain length of one of the nucleic acid sequences using blastn with the"BLAST 2 Sequences"tool Version 2.0.9 (May-07- 1999) set at default parameters. Such a pair of nucleic acids may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length. A variant may be described as, for example, an "allelic" (as defined above),"splice,""species,"or"polymorphic"variant. A splice variant may have significant identity to a reference molecule, but will generally have a greater or lesser number of polynucleotides due to alternate splicing of exons during mRNA processing. The corresponding polypeptide may possess additional functional domains or lack domains that are present in the reference molecule. Species variants are polynucleotide sequences that vary from one species to another. The resulting polypeptides will generally have significant amino acid identity relative to each other. A polymorphic variant is a variation in the polynucleotide sequence of a particular gene between individuals of a given species. Polymorphic variants also may encompass"single nucleotide polymorphisms" (SNPs) in which the polynucleotide sequence varies by one nucleotide base. The presence of SNPs may be indicative of, for example, a certain population, a disease state, or a propensity for a disease state.

A"variant"of a particular polypeptide sequence is defined as a polypeptide sequence having at least 40% sequence identity to the particular polypeptide sequence over a certain length of one of the polypeptide sequences using blastp with the"BLAST 2 Sequences"tool Version 2.0.9 (May-07- 1999) set at default parameters. Such a pair of polypeptides may show, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% or greater sequence identity over a certain defined length of one of the polypeptides.

THE INVENTION The invention is based on the discovery of new human receptors and membrane-associated proteins (REMAP), the polynucleotides encoding REMAP, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative, autoimmune/inflammatory, neurological, metabolic, developmental, endocrine, cardiovascular, reproductive, gastrointestinal, metabolic, genetic, and lipid metabolism disorders, cancer, and viral infections.

Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide sequences of the invention. Each polynucleotide and its corresponding polypeptide are correlated to a

single Incyte project identification number (Incyte Project ID). Each polypeptide sequence is denoted by both a polypeptide sequence identification number (Polypeptide SEQ ID NO:) and an Incyte polypeptide sequence number (Incyte Polypeptide ID) as shown. Each polynucleotide sequence is denoted by both a polynucleotide sequence identification number (Polynucleotide SEQ ID NO:) and an Incyte polynucleotide consensus sequence number (Incyte Polynucleotide ID) as shown. Column 6 shows the Incyte ID numbers of physical, full length clones corresponding to the polypeptide and polynucleotide sequences of the invention. The full length clones encode polypeptides which have at least 95% sequence identity to the polypeptide sequences shown in column 3.

Table 2 shows sequences with homology to the polypeptides of the invention as identified by BLAST analysis against the GenBank protein (genpept) database and the PROTEOME database.

Columns 1 and 2 show the polypeptide sequence identification number (Polypeptide SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for polypeptides of the invention. Column 3 shows the GenBank identification number (GenBank ID NO:) of the nearest GenBank homolog and the PROTEOME database identification numbers (PROTEOME ID NO:) of the nearest PROTEOME database homologs. Column 4 shows the probability scores for the matches between each polypeptide and its homolog (s). Column 5 shows the annotation of the GenBank and PROTEOME database homolog (s) along with relevant citations where applicable, all of which are expressly incorporated by reference herein.

Table 3 shows various structural features of the polypeptides of the invention. Columns 1 and 2 show the polypeptide sequence identification number (SEQ ID NO:) and the corresponding Incyte polypeptide sequence number (Incyte Polypeptide ID) for each polypeptide of the invention.

Column 3 shows the number of amino acid residues in each polypeptide. Column 4 shows potential phosphorylation sites, and column 5 shows potential glycosylation sites, as determined by the MOTIFS program of the GCG sequence analysis software package (Genetics Computer Group, Madison WI). Column 6 shows amino acid residues comprising signature sequences, domains, and motifs. Column 7 shows analytical methods for protein structure/function analysis and in some cases, searchable databases to which the analytical methods were applied.

Together, Tables 2 and 3 summarize the properties of polypeptides of the invention, and these properties establish that the claimed polypeptides are receptors and membrane-associated proteins.

For example, SEQ ID NO : 1 is 86% identical, from residue Mi to residue V1158, to mouse VPS10 domain receptor protein SorCS2 (GenBank ID gl2007720) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2.) The BLAST probability score is 0.0, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ IID NO : 1 also contains BNR repeats as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein family domains. (See

Table 3.) Data from MOTIFS, and additional BLAST analyses against the PRODOM and DOMO databases provide further corroborative evidence that SEQ ID NO : 1 is a member of the VPS 10 domain receptor protein family. In addition, TMAP analysis reveals that SEQ ID NO: 1 contains two transmembrane domains. In another example, SEQ ID NO : 6 is 99% identical, from residue N124 to residue V673, to human melatonin-related receptor (GenBank ID gl326155) as determined by BLAST. (See Table 2.) The BLAST probability score is 0.0. SEQ ID NO : 6 also contains a 7 transmembrane receptor (rhodopsin family) domain as determined by HMM-based PFAM database.

(See Table 3.) Data from BLIMPS, MOTIFS, and PROFILESCAN analyses provide further corroborative evidence that SEQ ID NO : 6 is a rhodopsin family receptor. In another example, SEQ ID NO : 17 is 49% identical, from residue L2 to residue P346, to human Nogo receptor (GenBank ID gl2407653) as determined by BLAST. (See Table 2.) The BLAST probability score is 1.2e-81.

SEQ ID NO : 17 also contains leucine rich repeat domains as determined byHMM-based PFAM database. (See Table 3.) Data from BLIMPS analyses provide further corroborative evidence that SEQ ID NO: 17 is a Nogo receptor. In yet another example, SEQ ID NO: 19 is 100% identical, from residue M103 to residue G409, to a human pregnancy-induced growth inhibitor (GenBank ID g6274473) as determined by BLAST. (See Table 2.) The BLAST probability score is 1.6e-166.

Data from TMAP provides further corroborative evidence that SEQ ID NO : 19 is a transmembrane protein. In a further example, SEQ ID NO : 23 is 82% identical, from residue V413 to residue K2696, and 87% identical from residue M1 to residue G324, to the murine SET domain and multiple PHD finger domain-containing nuclear receptor protein, NSD1 (GenBank ID g3329465) as determined by BLAST. (See Table 2.) The BLAST probability score is 0.0. SEQ ID NO : 23 also contains the above-identified conserved domains of NSD1 as determined by HMM-based PFAM database. (See Table 3.) Data from BLIMPS and MOTIFS analyses provide further corroborative evidence that SEQ ID NO : 23 is a SET-domain-containing receptor. In another example, SEQ ID NO : 24 is 58% identical, from residue D740 to residue L2210, and 45% identical from residue K466 to residue N1311, to human thyroid hormone receptor-associated protein complex component, TRAP240 (GenBank ID g4530437) as determined by BLAST. (See Table 2.) The BLAST probability score is 0.0. Data from MOTIFS analysis provides evidence for the presence of a"P-loop"ATP/GTP-binding site. In yet another example, SEQ ID NO : 25 is 98% identical, from residue V81 to residue G336, and 80% identical from residue M2 to residue Q110, to a murine nuclear orphan receptor/transcription factor (GenBank ID gl869971) as determined by BLAST. (See Table 2.) The BLAST probability score is 3.4e-180. SEQ ID NO : 25 also contains a nuclear hormone receptor ligand-binding domain as determined by searching for statistically significant matches in the HMM-based PFAM database.

(See Table 3.) Data from BLIMPS, MOTIFS, and PROFILESCAN analyses provide further corroborative evidence that SEQ ID NO : 25 is a nuclear hormone receptor. SEQ ID NO : 2-5, SEQ ID

NO : 7-16, SEQ ID NO : 18, SEQ ID NO : 20-22, and SEQ ID NO : 26 were analyzed and annotated in a similar manner. The algorithms and parameters for the analysis of SEQ ID NO: 1-26 are described in Table 7.

As shown in Table 4, the full length polynucleotide sequences of the present invention were assembled using cDNA sequences or coding (exon) sequences derived from genomic DNA, or any combination of these two types of sequences. Column 1 lists the polynucleotide sequence identification number (Polynucleotide SEQ ID NO:), the corresponding Incyte polynucleotide consensus sequence number (Incyte ID) for each polynucleotide of the invention, and the length of each polynucleotide sequence in basepairs. Column 2 shows the nucleotide start (5') and stop (3') positions of the cDNA and/or genomic sequences used to assemble the full length polynucleotide sequences of the invention, and of fragments of the polynucleotide sequences which are useful, for example, in hybridization or amplification technologies that identify SEQ ID NO : 27-52 or that distinguish between SEQ ID NO : 27-52 and related polynucleotide sequences.

The polynucleotide fragments described in Column 2 of Table 4 may refer specifically, for example, to Incyte cDNAs derived from tissue-specific cDNA libraries or from pooled cDNA libraries. Alternatively, the polynucleotide fragments described in column 2 may refer to GenBank cDNAs or ESTs which contributed to the assembly of the full length polynucleotide sequences. In addition, the polynucleotide fragments described in column 2 may identify sequences derived from the ENSEMBL (The Sanger Centre, Cambridge, UK) database (i. e., those sequences including the designation"ENST"). Alternatively, the polynucleotide fragments described in column 2 may be derived from the NCBI RefSeq Nucleotide Sequence Records Database (i. e., those sequences including the designation"NM"or"NT") or the NCBI RefSeq Protein Sequence Records (i. e., those sequences including the designation"NP"). Alternatively, the polynucleotide fragments described in column 2 may refer to assemblages of both cDNA and Genscan-predicted exons brought together by an"exon stitching"algorithm. For example, a polynucleotide sequence identified as -N3-N4 represents a"stitched"sequence in which XXXXXX is the identification number of the cluster of sequences to which the algorithm was applied, and YYYYY is the number of the prediction generated by the algorithm, and N, 2 3, if present, represent specific exons that may have been manually edited during analysis (See Example V). Alternatively, the polynucleotide fragments in column 2 may refer to assemblages of exons brought together by an "exon-stretching"algorithm. For example, a polynucleotide sequence identified as E71E_gAAAAA_gBBBBB_l_N is a"stretched"sequence, with XXXXXX being the Incyte project identification number, gAAAAA being the GenBank identification number of the human genomic sequence to which the"exon-stretching"algorithm was applied, gBBBBB being the GenBank identification number or NCBI RefSeq identification number of the nearest GenBank

protein homolog, and N referring to specific exons (See Example V). In instances where a RefSeq sequence was used as a protein homolog for the"exon-stretching"algorithm, a RefSeq identifier (denoted by"NM,""NP,"or"NT") may be used in place of the GenBank identifier (i. e., gBBBBB).

Alternatively, a prefix identifies component sequences that were hand-edited, predicted from genomic DNA sequences, or derived from a combination of sequence analysis methods. The following Table lists examples of component sequence prefixes and corresponding sequence analysis methods associated with the prefixes (see Example IV and Example V). Prefix Type of analysis and/or examples of programs GNN, GFG, Exon prediction from genomic sequences using, for example, ENST GENSCAN (Stanford University, CA, USA) or FGENES (Computer Genomics Group, The Sanger Centre, Cambridge, UK). GBI Hand-edited analysis of genomic sequences. FL Stitched or stretched genomic sequences (see Example V). INCY Full length transcript and exon prediction from mapping of EST sequences to the genome. Genomic location and EST composition data are combined to predict the exons and resulting transcript.

In some cases, Incyte cDNA coverage redundant with the sequence coverage shown in Table 4 was obtained to confirm the final consensus polynucleotide sequence, but the relevant Incyte cDNA identification numbers are not shown.

Table 5 shows the representative cDNA libraries for those full length polynucleotide sequences which were assembled using Incyte cDNA sequences. The representative cDNA library is the Incyte cDNA library which is most frequently represented by the Incyte cDNA sequences which were used to assemble and confirm the above polynucleotide sequences. The tissues and vectors which were used to construct the cDNA libraries shown in Table 5 are described in Table 6.

The invention also encompasses REMAP variants. A preferred REMAP variant is one which has at least about 80%, or alternatively at least about 90%, or even at least about 95% amino acid sequence identity to the REMAP amino acid sequence, and which contains at least one functional or structural characteristic of REMAP.

The invention also encompasses polynucleotides which encode REMAP. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO : 27-52, which encodes REMAP. The polynucleotide sequences of SEQ ID NO : 27-52, as presented in the Sequence Listing, embrace the equivalent RNA sequences, wherein occurrences of the nitrogenous base thymine are replaced with uracil, and the sugar backbone is composed of ribose instead of deoxyribose.

The invention also encompasses a variant of a polynucleotide sequence encoding REMAP.

In particular, such a variant polynucleotide sequence will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to the polynucleotide sequence encoding REMAP. A particular aspect of the invention encompasses a variant of a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO : 27-52 which has at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to a nucleic acid sequence selected from the group consisting of SEQ ID NO : 27-52. Any one of the polynucleotide variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of REMAP.

In addition, or in the alternative, a polynucleotide variant of the invention is a splice variant of a polynucleotide sequence encoding REMAP. A splice variant may have portions which have significant sequence identity to the polynucleotide sequence encoding REMAP, but will generally have a greater or lesser number of polynucleotides due to additions or deletions of blocks of sequence arising from alternate splicing of exons during mRNA processing. A splice variant may have less than about 70%, or alternatively less than about 60%, or alternatively less than about 50% polynucleotide sequence identity to the polynucleotide sequence encoding REMAP over its entire length; however, portions of the splice variant will have at least about 70%, or alternatively at least about 85%, or alternatively at least about 95%, or alternatively 100% polynucleotide sequence identity to portions of the polynucleotide sequence encoding REMAP. For example, a polynucleotide comprising a sequence of SEQ ID NO : 39 is a splice variant of a polynucleotide comprising a sequence of SEQ ID NO : 52. Any one of the splice variants described above can encode an amino acid sequence which contains at least one functional or structural characteristic of REMAP.

It will be appreciated by those skilled in the art that as a result of the degeneracy of the genetic code, a multitude of polynucleotide sequences encoding REMAP, some bearing minimal similarity to the polynucleotide sequences of any known and naturally occurring gene, may be produced. Thus, the invention contemplates each and every possible variation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide sequence of naturally occurring REMAP, and all such variations are to be considered as being specifically disclosed.

Although nucleotide sequences which encode REMAP and its variants are generally capable of hybridizing to the nucleotide sequence of the naturally occurring REMAP under appropriately selected conditions of stringency, it may be advantageous to produce nucleotide sequences encoding REMAP or its derivatives possessing a substantially different codon usage, e. g., inclusion of non- naturally occurring codons. Codons may be selected to increase the rate at which expression of the

peptide occurs in a particular prokaryotic or eukaryotic host in accordance with the frequency with which particular codons are utilized by the host. Other reasons for substantially altering the nucleotide sequence encoding REMAP and its derivatives without altering the encoded amino acid sequences include the production of RNA transcripts having more desirable properties, such as a greater half-life, than transcripts produced from the naturally occurring sequence.

The invention also encompasses production of DNA sequences which encode REMAP and REMAP derivatives, or fragments thereof, entirely by synthetic chemistry. After production, the synthetic sequence may be inserted into any of the many available expression vectors and cell systems using reagents well known in the art. Moreover, synthetic chemistry may be used to introduce mutations into a sequence encoding REMAP or any fragment thereof.

Also encompassed by the invention are polynucleotide sequences that are capable of hybridizing to the claimed polynucleotide sequences, and, in particular, to those shown in SEQ ID NO : 27-52 and fragments thereof under various conditions of stringency. (See, e. g., Wahl, G. M. and S. L. Berger (1987) Methods Enzymol. 152: 399-407 ; Kimmel, A. R. (1987) Methods Enzymol.

152: 507-511.) Hybridization conditions, including annealing and wash conditions, are described in "Definitions." Methods for DNA sequencing are well known in the art and may be used to practice any of the embodiments of the invention. The methods may employ such enzymes as the Klenow fragment of DNA polymerase I, SEQUENASE (US Biochemical, Cleveland OH), Taq polymerase (Applied Biosystems), thermostable T7 polymerase (Amersham Biosciences, Piscataway NJ), or combinations of polymerases and proofreading exonucleases such as those found in the ELONGASE amplification system (Invitrogen, Carlsbad CA). Preferably, sequence preparation is automated with machines such as the MICROLAB 2200 liquid transfer system (Hamilton, Reno NV), PTC200 thermal cycler (MJ Research, Watertown MA) and ABI CATALYST 800 thermal cycler (Applied Biosystems).

Sequencing is then carried out using either the ABI 373 or 377 DNA sequencing system (Applied Biosystems), the MEGABACE 1000 DNA sequencing system (Amersham Biosciences), or other systems known in the art. The resulting sequences are analyzed using a variety of algorithms which are well known in the art. (See, e. g., Ausubel, F. M. (1997) Short Protocols in Molecular Biology, John Wiley & Sons, New York NY, unit 7.7; Meyers, R. A. (1995) Molecular Biology and Biotechnology, Wiley VCH, New York NY, pp. 856-853.) The nucleic acid sequences encoding REMAP may be extended utilizing a partial nucleotide sequence and employing various PCR-based methods known in the art to detect upstream sequences, such as promoters and regulatory elements. For example, one method which may be employed, restriction-site PCR, uses universal and nested primers to amplify unknown sequence from genomic DNA within a cloning vector. (See, e. g., Sarkar, G. (1993) PCR Methods Applic. 2: 318-322.)

Another method, inverse PCR, uses primers that extend in divergent directions to amplify unknown sequence from a circularized template. The template is derived from restriction fragments comprising a known genomic locus and surrounding sequences. (See, e. g., Triglia, T. et al. (1988) Nucleic Acids Res. 16: 8186.) A third method, capture PCR, involves PCR amplification of DNA fragments adjacent to known sequences in human and yeast artificial chromosome DNA. (See, e. g., Lagerstrom, M. et al. (1991) PCR Methods Applic. 1: 111-119.) In this method, multiple restriction enzyme digestions and ligations may be used to insert an engineered double-stranded sequence into a region of unknown sequence before performing PCR. Other methods which may be used to retrieve unknown sequences are known in the art. (See, e. g., Parker, J. D. et al. (1991) Nucleic Acids Res.

19: 3055-3060). Additionally, one may use PCR, nested primers, and PROMOTERFINDER libraries (Clontech, Palo Alto CA) to walk genomic DNA. This procedure avoids the need to screen libraries and is useful in finding intron/exon junctions. For all PCR-based methods, primers may be designed using commercially available software, such as OLIGO 4.06 primer analysis software (National Biosciences, Plymouth MN) or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the template at temperatures of about 68°C to 72°C.

When screening for full length cDNAs, it is preferable to use libraries that have been size-selected to include larger cDNAs. In addition, random-primed libraries, which often include sequences containing the 5'regions of genes, are preferable for situations in which an oligo d (T) library does not yield a full-length cDNA. Genomic libraries may be useful for extension of sequence into 5'non-transcribed regulatory regions.

Capillary electrophoresis systems which are commercially available may be used to analyze the size or confirm the nucleotide sequence of sequencing or PCR products. In particular, capillary sequencing may employ flowable polymers for electrophoretic separation, four different nucleotide- specific, laser-stimulated fluorescent dyes, and a charge coupled device camera for detection of the emitted wavelengths. Output/light intensity may be converted to electrical signal using appropriate software (e. g., GENOTYPER and SEQUENCE NAVIGATOR, Applied Biosystems), and the entire process from loading of samples to computer analysis and electronic data display may be computer controlled. Capillary electrophoresis is especially preferable for sequencing small DNA fragments which may be present in limited amounts in a particular sample.

In another embodiment of the invention, polynucleotide sequences or fragments thereof which encode REMAP may be cloned in recombinant DNA molecules that direct expression of REMAP, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences which encode substantially the same or a functionally equivalent amino acid sequence may be produced and used to express REMAP.

The nucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter REMAP-encoding sequences for a variety of purposes including, but not limited to, modification of the cloning, processing, and/or expression of the gene product. DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. For example, oligonucleotide- mediated site-directed mutagenesis may be used to introduce mutations that create new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, and so forth.

The nucleotides of the present invention may be subjected to DNA shuffling techniques such as MOLECULARBREEDING (Maxygen Inc., Santa Clara CA; described in U. S. Patent No.

5,837,458; Chang, C.-C. et al. (1999) Nat. Biotechnol. 17: 793-797; Christians, F. C. et al. (1999) Nat.

Biotechnol. 17 : 259-264; and Crameri, A. et al. (1996) Nat. Biotechnol. 14: 315-319) to alter or improve the biological properties of REMAP, such as its biological or enzymatic activity or its ability to bind to other molecules or compounds. DNA shuffling is a process by which a library of gene variants is produced using PCR-mediated recombination of gene fragments. The library is then subjected to selection or screening procedures that identify those gene variants with the desired properties. These preferred variants may then be pooled and further subjected to recursive rounds of DNA shuffling and selection/screening. Thus, genetic diversity is created through"artificial" breeding and rapid molecular evolution. For example, fragments of a single gene containing random point mutations may be recombined, screened, and then reshuffled until the desired properties are optimized. Alternatively, fragments of a given gene may be recombined with fragments of homologous genes in the same gene family, either from the same or different species, thereby maximizing the genetic diversity of multiple naturally occurring genes in a directed and controllable manner.

In another embodiment, sequences encoding REMAP may be synthesized, in whole or in part, using chemical methods well known in the art. (See, e. g., Caruthers, M. H. et al. (1980) Nucleic Acids Symp. Ser. 7: 215-223; and Horn, T. et al. (1980) Nucleic Acids Symp. Ser. 7: 225-232.) Alternatively, REMAP itself or a fragment thereof may be synthesized using chemical methods. For example, peptide synthesis can be performed using various solution-phase or solid-phase techniques.

(See, e. g., Creighton, T. (1984) Proteins, Structures and Molecular Properties, WH Freeman, New York NY, pp. 55-60; and Roberge, J. Y. et al. (1995) Science 269: 202-204.) Automated synthesis may be achieved using the ABI 431A peptide synthesizer (Applied Biosystems). Additionally, the amino acid sequence of REMAP, or any part thereof, may be altered during direct synthesis and/or combined with sequences from other proteins, or any part thereof, to produce a variant polypeptide or a polypeptide having a sequence of a naturally occurring polypeptide.

The peptide may be substantially purified by preparative high performance liquid

chromatography. (See, e. g., Chiez, R. M. and F. Z. Regnier (1990) Methods Enzymol. 182 : 392-421.) The composition of the synthetic peptides may be confirmed by amino acid analysis or by sequencing. (See, e. g., Creighton, supra, pp. 28-53.) In order to express a biologically active REMAP, the nucleotide sequences encoding REMAP or derivatives thereof may be inserted into an appropriate expression vector, i. e., a vector which contains the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host. These elements include regulatory sequences, such as enhancers, constitutive and inducible promoters, and 5'and 3'untranslated regions in the vector and in polynucleotide sequences encoding REMAP. Such elements may vary in their strength and specificity. Specific initiation signals may also be used to achieve more efficient translation of sequences encoding REMAP. Such signals include the ATG initiation codon and adjacent sequences, e. g. the Kozak sequence. In cases where sequences encoding REMAP and its initiation codon and upstream regulatory sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous translational control signals including an in- frame ATG initiation codon should be provided by the vector. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate for the particular host cell system used.

(See, e. g., Scharf, D. et al. (1994) Results Probl. Cell Differ. 20: 125-162.) Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding REMAP and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. (See, e. g., Sambrook, J. et al. (1989) Molecular Cloning, Laboratory Manual, Cold Spring Harbor Press, Plainview NY, ch. 4,8, and 16-17; Ausubel, F. M. et al. (1995) Current Protocols in Molecular Biology, John Wiley & Sons, New York NY, ch. 9,13, and 16.) A variety of expression vector/host systems may be utilized to contain and express sequences encoding REMAP. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e. g., baculovirus); plant cell systems transformed with viral expression vectors (e. g., cauliflower mosaic virus, CaMV, or tobacco mosaic virus, TMV) or with bacterial expression vectors (e. g., Ti or pBR322 plasmids) ; or animal cell systems. (See, e. g., Sambrook, supra ; Ausubel, supra ; Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264 : 5503-5509; Engelhard, E. K. et al. (1994) Proc. Natl.

Acad. Sci. USA 91: 3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther. 7: 1937-1945; Takamatsu,

N. (1987) EMBO J. 6: 307-311; The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York NY, pp. 191-196; Logan, J. and T. Shenk (1984) Proc. Natl. Acad. Sci. USA 81: 3655-3659; and Harrington, J. J. et al. (1997) Nat. Genet. 15: 345-355.) Expression vectors derived from retroviruses, adenoviruses, or herpes or vaccinia viruses, or from various bacterial plasmids, may be used for delivery of nucleotide sequences to the targeted organ, tissue, or cell population.

(See, e. g., Di Nicola, M. et al. (1998) Cancer Gen. Ther. 5 (6): 350-356; Yu, M. et al. (1993) Proc.

Natl. Acad. Sci. USA 90 (13) : 6340-6344 ; Buller, R. M. et al. (1985) Nature 317 (6040): 813-815; McGregor, D. P. et al. (1994) Mol. Immunol. 31 (3): 219-226; and Verma, I. M. and N. Somia (1997) Nature 389: 239-242.) The invention is not limited by the host cell employed.

In bacterial systems, a number of cloning and expression vectors may be selected depending upon the use intended for polynucleotide sequences encoding REMAP. For example, routine cloning, subcloning, and propagation of polynucleotide sequences encoding REMAP can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Invitrogen). Ligation of sequences encoding REMAP into the vector's multiple cloning site disrupts the lacZ gene, allowing a colorimetric screening procedure for identification of transformed bacteria containing recombinant molecules. In addition, these vectors may be useful for in vitro transcription, dideoxy sequencing, single strand rescue with helper phage, and creation of nested deletions in the cloned sequence. (See, e. g., Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem.

264: 5503-5509.) When large quantities of REMAP are needed, e. g. for the production of antibodies, vectors which direct high level expression of REMAP may be used. For example, vectors containing the strong, inducible SP6 or T7 bacteriophage promoter may be used.

Yeast expression systems may be used for production of REMAP. A number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH promoters, may be used in the yeast Saccharomyces cerevisiae or Pic7ia pastors. In addition, such vectors direct either the secretion or intracellular retention of expressed proteins and enable integration of foreign sequences into the host genome for stable propagation. (See, e. g., Ausubel, 1995, supra ; Bitter, G. A. et al. (1987) Methods Enzymol. 153: 516-544; and Scorer, C. A. et al. (1994) Bio/Technology 12: 181-184.) Plant systems may also be used for expression of REMAP. Transcription of sequences encoding REMAP may be driven by viral promoters, e. g., the 35S and 19S promoters of CaMV used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J.

6: 307-311). Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used. (See, e. g., Coruzzi, G. et al. (1984) EMBO J. 3: 1671-1680; Broglie, R. et al.

(1984) Science 224: 838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17: 85-105.) These constructs can be introduced into plant cells by direct DNA transformation or

pathogen-mediated transfection. (See, e. g., The McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York NY, pp. 191-196.) In mammalian cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, sequences encoding REMAP may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain infective virus which expresses REMAP in host cells. (See, e. g., Logan, J. and T. Shenk (1984) Proc.

Natl. Acad. Sci. USA 81: 3655-3659.) In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells. SV40 or EBV- based vectors may also be used for high-level protein expression.

Human artificial chromosomes (HACs) may also be employed to deliver larger fragments of DNA than can be contained in and expressed from a plasmid. HACs of about 6 kb to 10 Mb are constructed and delivered via conventional delivery methods (liposomes, polycationic amino polymers, or vesicles) for therapeutic purposes. (See, e. g., Harrington, J. J. et al. (1997) Nat. Genet.

15:345-355.) For long term production of recombinant proteins in mammalian systems, stable expression of REMAP in cell lines is preferred. For example, sequences encoding REMAP can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector.

Following the introduction of the vector, cells may be allowed to grow for about 1 to 2 days in enriched media before being switched to selective media. The purpose of the selectable marker is to confer resistance to a selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be propagated using tissue culture techniques appropriate to the cell type.

Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase and adenine phosphoribosyltransferase genes, for use in tk-and apr cells, respectively. (See, e. g., Wigler, M. et al. (1977) Cell 11: 223-232; Lowy, I. et al. (1980) Cell 22: 817-823.) Also, antimetabolite, antibiotic, or herbicide resistance can be used as the basis for selection. For example, dhfr confers resistance to methotrexate; neo confers resistance to the aminoglycosides neomycin and G-418; and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively. (See, e. g., Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. USA 77: 3567-3570; Colbere-Garapin, F. et al. (1981) J. Mol. Biol. 150: 1-14.) Additional selectable genes have been described, e. g., trpB and hisD, which alter cellular requirements for metabolites. (See, e. g., Hartman, S. C. and R. C. Mulligan (1988) Proc.

Natl. Acad. Sci. USA 85: 8047-8051.) Visible markers, e. g., anthocyanins, green fluorescent proteins

(GFP ; Clontech), B glucuronidase and its substrate B-glucuronide, or luciferase and its substrate luciferin may be used. These markers can be used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system.

(See, e. g., Rhodes, C. A. (1995) Methods Mol. Biol. 55: 121-131.) Although the presence/absence of marker gene expression suggests that the gene of interest is also present, the presence and expression of the gene may need to be confirmed. For example, if the sequence encoding REMAP is inserted within a marker gene sequence, transformed cells containing sequences encoding REMAP can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a sequence encoding REMAP under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.

In general, host cells that contain the nucleic acid sequence encoding REMAP and that express REMAP may be identified by a variety of procedures known to those of skill in the art.

These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations, PCR amplification, and protein bioassay or immunoassay techniques which include membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein sequences.

Immunological methods for detecting and measuring the expression of REMAP using either specific polyclonal or monoclonal antibodies are known in the art. Examples of such techniques include enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS). A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering epitopes on REMAP is preferred, but a competitive binding assay may be employed. These and other assays are well known in the art. (See, e. g., Hampton, R. et al. (1990) Serological Methods, a Laboratory Manual, APS Press, St. Paul MN, Sect. IV; Coligan, J. E. et al. (1997) Current Protocols in Immunology, Greene Pub. Associates and Wiley-Interscience, New York NY ; and Pound, J. D. (1998) Immunochemical Protocols, Humana Press, Totowa NJ.) A wide variety of labels and conjugation techniques are known by those skilled in the art and may be used in various nucleic acid and amino acid assays. Means for producing labeled hybridization or PCR probes for detecting sequences related to polynucleotides encoding REMAP include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.

Alternatively, the sequences encoding REMAP, or any fragments thereof, may be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by addition of an appropriate RNA polymerase such as T7, T3, or SP6 and labeled nucleotides. These procedures may be conducted using a variety of commercially available kits, such as those provided by Amersham Biosciences, Promega (Madison

WI), and US Biochemical. Suitable reporter molecules or labels which may be used for ease of detection include radionuclides, enzymes, fluorescent, chemiluminescent, or chromogenic agents, as well as substrates, cofactors, inhibitors, magnetic particles, and the like.

Host cells transformed with nucleotide sequences encoding REMAP may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a transformed cell may be secreted or retained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides which encode REMAP may be designed to contain signal sequences which direct secretion of REMAP through a prokaryotic or eukaryotic cell membrane.

In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a"prepro"or "pro"form of the protein may also be used to specify protein targeting, folding, and/or activity.

Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e. g., CHO, HeLa, MDCK, HEK293, and WI38) are available from the American Type Culture Collection (ATCC, Manassas VA) and may be chosen to ensure the correct modification and processing of the foreign protein.

In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences encoding REMAP may be ligated to a heterologous sequence resulting in translation of a fusion protein in any of the aforementioned host systems : For example, a chimeric REMAP protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide libraries for inhibitors of REMAP activity. Heterologous protein and peptide moieties may also facilitate purification of fusion proteins using commercially available affinity matrices. Such moieties include, but are not limited to, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutinin (HA). GST, MBP, Trx, CBP, and 6-His enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively. FLAG, c-myc, and hemagglutinin (HA) enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags. A fusion protein may also be engineered to contain a proteolytic cleavage site located between the REMAP encoding sequence and the heterologous protein sequence, so that REMAP may be cleaved away from the heterologous moiety following purification. Methods for fusion protein expression and purification are discussed in Ausubel (1995, supra, ch. 10). A variety of commercially available kits may also be used to facilitate expression and

purification of fusion proteins.

In a further embodiment of the invention, synthesis of radiolabeled REMAP may be achieved in vitro using the TNT rabbit reticulocyte lysate or wheat germ extract system (Promega). These systems couple transcription and translation of protein-coding sequences operably associated with the T7, T3, or SP6 promoters. Translation takes place in the presence of a radiolabeled amino acid precursor, for example, 35S-methionine.

REMAP of the present invention or fragments thereof may be used to screen for compounds that specifically bind to REMAP. At least one and up to a plurality of test compounds may be screened for specific binding to REMAP. Examples of test compounds include antibodies, oligonucleotides, proteins (e. g., ligands or receptors), or small molecules. In one embodiment, the compound thus identified is closely related to the natural ligand of REMAP, e. g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner. (See, e. g., Coligan, J. E. et al. (1991) Current Protocols in Immunology 1 (2): Chapter 5.) In another embodiment, the compound thus identified is a natural ligand of a receptor REMAP. (See, e. g., Howard, A. D. et al. (2001) Trends Pharmacol. Sci. 22: 132-140; Wise, A. et al. (2002) Drug Discovery Today 7: 235- 246.) In other embodiments, the compound can be closely related to the natural receptor to which REMAP binds, at least a fragment of the receptor, or a fragment of the receptor including all or a portion of the ligand binding site or binding pocket. For example, the compound may be a receptor for REMAP which is capable of propagating a signal, or a decoy receptor for REMAP which is not capable of propagating a signal (Ashkenazi, A. and V. M. Divit (1999) Curr. Opin. Cell Biol. 11: 255- 260; Mantovani, A. et al. (2001) Trends Immunol. 22: 328-336). The compound can be rationally designed using known techniques. Examples of such techniques include those used to construct the compound etanercept (ENBREL; Immunex Corp., Seattle WA), which is efficacious for treating rheumatoid arthritis in humans. Etanercept is an engineered p75 tumor necrosis factor (TNF) receptor dimer linked to the Fc portion of human IgGI (Taylor, P. C. et al. (2001) Curr. Opin.

Immunol. 13: 611-616).

In one embodiment, screening for compounds which specifically bind to, stimulate, or inhibit REMAP involves producing appropriate cells which express REMAP, either as a secreted protein or on the cell membrane. Preferred cells include cells from mammals, yeast, Drosophila, or E. coli.

Cells expressing REMAP or cell membrane fractions which contain REMAP are then contacted with a test compound and binding, stimulation, or inhibition of activity of either REMAP or the compound is analyzed.

An assay may simply test binding of a test compound to the polypeptide, wherein binding is detected by a fluorophore, radioisotope, enzyme conjugate, or other detectable label. For example,

the assay may comprise the steps of combining at least one test compound with REMAP, either in solution or affixed to a solid support, and detecting the binding of REMAP to the compound.

Alternatively, the assay may detect or measure binding of a test compound in the presence of a labeled competitor. Additionally, the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound (s) may be free in solution or affixed to a solid support.

An assay can be used to assess the ability of a compound to bind to its natural ligand and/or to inhibit the binding of its natural ligand to its natural receptors. Examples of such assays include radio-labeling assays such as those described in U. S. Patent No. 5,914,236 and U. S. Patent No.

6,372,724. In a related embodiment, one or more amino acid substitutions can be introduced into a polypeptide compound (such as a receptor) to improve or alter its ability to bind to its natural ligands.

(See, e. g., Matthews, D. J. and J. A. Wells. (1994) Chem. Biol. 1: 25-30.) In another related embodiment, one or more amino acid substitutions can be introduced into a polypeptide compound (such as a ligand) to improve or alter its ability to bind to its natural receptors. (See, e. g., Cunningham, B. C. and J. A. Wells (1991) Proc. Natl. Acad. Sci. USA 88 : 3407-3411; Lowman, H. B. et al. (1991) J. Biol. Chem. 266: 10982-10988.) REMAP of the present invention or fragments thereof may be used to screen for compounds that modulate the activity of REMAP. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for REMAP activity, wherein REMAP is combined with at least one test compound, and the activity of REMAP in the presence of a test compound is compared with the activity of REMAP in the absence of the test compound. A change in the activity of REMAP in the presence of the test compound is indicative of a compound that modulates the activity of REMAP. Alternatively, a test compound is combined with an in vitro or cell-free system comprising REMAP under conditions suitable for REMAP activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of REMAP may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened.

In another embodiment, polynucleotides encoding REMAP or their mammalian homologs may be"knocked out"in an animal model system using homologous recombination in embryonic stem (ES) cells. Such techniques are well known in the art and are useful for the generation of animal models of human disease. (See, e. g., U. S. Patent No. 5,175,383 and U. S. Patent No. 5,767,337.) For example, mouse ES cells, such as the mouse 129/SvJ cell line, are derived from the early mouse embryo and grown in culture. The ES cells are transformed with a vector containing the gene of interest disrupted by a marker gene, e. g., the neomycin phosphotransferase gene (neo; Capecchi, M. R.

(1989) Science 244: 1288-1292). The vector integrates into the corresponding region of the host

genome by homologous recombination. Alternatively, homologous recombination takes place using the Cre-loxP system to knockout a gene of interest in a tissue-or developmental stage-specific manner (Marth, J. D. (1996) Clin. Invest. 97: 1999-2002; Wagner, K. U. et al. (1997) Nucleic Acids Res. 25: 4323-4330). Transformed ES cells are identified and microinjected into mouse cell blastocysts such as those from the C57BL/6 mouse strain. The blastocysts are surgically transferred to pseudopregnant dams, and the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains. Transgenic animals thus generated may be tested with potential therapeutic or toxic agents.

Polynucleotides encoding REMAP may also be manipulated in vitro in ES cells derived from human blastocysts. Human ES cells have the potential to differentiate into at least eight separate cell lineages including endoderm, mesoderm, and ectodermal cell types. These cell lineages differentiate into, for example, neural cells, hematopoietic lineages, and cardiomyocytes (Thomson, J. A. et al.

(1998) Science 282: 1145-1147).

Polynucleotides encoding REMAP can also be used to create"knockin"humanized animals (pigs) or transgenic animals (mice or rats) to model human disease. With knockin technology, a region of a polynucleotide encoding REMAP is injected into animal ES cells, and the injected sequence integrates into the animal cell genome. Transformed cells are injected into blastulae, and the blastulae are implanted as described above. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain information on treatment of a human disease.

Alternatively, a mammal inbred to overexpress REMAP, e. g., by secreting REMAP in its milk, may also serve as a convenient source of that protein (Janne, J. et al. (1998) Biotechnol. Annu. Rev. 4: 55- 74).

THERAPEUTICS Chemical and structural similarity, e. g., in the context of sequences and motifs, exists between regions of REMAP and receptors and membrane-associated proteins. In addition, examples of tissues expressing REMAP can be found in Table 6 and can also be found in Example XI.

Therefore, REMAP appears to play a role in cell proliferative, autoimmune/inflammatory, neurological, metabolic, developmental, endocrine, cardiovascular, reproductive, gastrointestinal, metabolic, genetic, and lipid metabolism disorders, cancer, and viral infections. In the treatment of disorders associated with increased REMAP expression or activity, it is desirable to decrease the expression or activity of REMAP. In the treatment of disorders associated with decreased REMAP expression or activity, it is desirable to increase the expression or activity of REMAP.

Therefore, in one embodiment, REMAP or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of REMAP. Examples of such disorders include, but are not limited to, a cell proliferative

disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy- candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves'disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjögren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma ; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann- Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia,

catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; a metabolic disorder such as Addison's disease, cerebrotendinous xanthomatosis, congenital adrenal hyperplasia, coumarin resistance, cystic fibrosis, fatty hepatocirrhosis, fructose-1,6-diphosphatase deficiency, galactosemia, goiter, glucagonoma, glycogen storage diseases, hereditary fructose intolerance, hyperadrenalism, hypoadrenalism, hyperparathyroidism, hypoparathyroidism, hypercholesterolemia, hyperthyroidism, hypoglycemia, hypothyroidism, hyperlipidemia, hyperlipemia, lipid myopathies, lipodystrophies, lysosomal storage diseases, mannosidosis, neuraminidase deficiency, obesity, osteoporosis, phenylketonuria, pseudovitamin D-deficiency rickets, disorders of carbohydrate metabolism such as congenital type II dyserythropoietic anemia, diabetes, insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, galactose epimerase deficiency, glycogen storage diseases, lysosomal storage diseases, fructosuria, pentosuria, and inherited abnormalities of pyruvate metabolism, disorders of lipid metabolism such as fatty liver, cholestasis, primary biliary cirrhosis, carnitine deficiency, carnitine palmitoyltansferase deficiency, myoadenylate deaminase deficiency, hypertriglyceridemia, lipid storage disorders such Fabry's disease, Gaucher's disease, Niemann-Pick's disease, metachromatic leukodystrophy, adrenoleukodystrophy, GM2 gangliosidosis, and ceroid lipofuscinosis, abetalipoproteinemia, Tangier disease, hyperlipoproteinemia, lipodystrophy, lipomatoses, acute panniculitis, disseminated fat necrosis, adiposis dolorosa, lipoid adrenal hyperplasia, minimal change disease, lipomas, atherosclerosis, hypercholesterolemia, hypercholesterolemia with hypertriglyceridemia, primary hypoalphalipoproteinemia, hypothyroidism, renal disease, liver disease, lecithin: cholesterol acyltransferase deficiency, cerebrotendinous xanthomatosis, sitosterolemia, hypocholesterolemia, Tay-Sachs disease, Sandhoff's disease, hyperlipidemia, hyperlipemia, and lipid myopathies, and disorders of copper metabolism such as Menke's disease, Wilson's disease, and Ehlers-Danlos syndrome type IX diabetes; a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, a seizure disorder such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss; an endocrine disorder such as a disorder of the hypothalamus and/or pituitary resulting from lesions such as a primary brain tumor, adenoma, infarction associated with pregnancy, hypophysectomy, aneurysm, vascular malformation, thrombosis, infection, immunological disorder, and complication

due to head trauma, a disorder associated with hypopituitarism including hypogonadism, Sheehan syndrome, diabetes insipidus, Kallman's disease, Hand-Schuller-Christian disease, Letterer-Siwe disease, sarcoidosis, empty sella syndrome, and dwarfism, a disorder associated with hyperpituitarism including acromegaly, giantism, and syndrome of inappropriate antidiuretic hormone (ADH) secretion (SIADH) often caused by benign adenoma, a disorder associated with hypothyroidism including goiter, myxedema, acute thyroiditis associated with bacterial infection, subacute thyroiditis associated with viral infection, autoimmune thyroiditis (Hashimoto's disease), and cretinism, a disorder associated with hyperthyroidism including thyrotoxicosis and its various forms, Grave's disease, pretibial myxedema, toxic multinodular goiter, thyroid carcinoma, and Plummer's disease, a disorder associated with hyperparathyroidism including Conn disease (chronic hypercalemia), a pancreatic disorder such as Type I or Type It diabetes mellitus and associated complications, a disorder associated with the adrenals such as hyperplasia, carcinoma, or adenoma of the adrenal cortex, hypertension associated with alkalosis, amyloidosis, hypokalemia, Cushing's disease, Liddle's syndrome, and Arnold-Healy-Gordon syndrome, pheochromocytoma tumors, and Addison's disease, a disorder associated with gonadal steroid hormones such as: in women, abnormal prolactin production, infertility, endometriosis, perturbation of the menstrual cycle, polycystic ovarian disease, hyperprolactinemia, isolated gonadotropin deficiency, amenorrhea, galactorrhea, hermaphroditism, hirsutism and virilization, breast cancer, and, in post-menopausal women, osteoporosis, and, in men, Leydig cell deficiency, male climacteric phase, and germinal cell aplasia, a hypergonadal disorder associated with Leydig cell tumors, androgen resistance associated with absence of androgen receptors, and syndrome of 5 a-reductase ; a cardiovascular disorder such as arteriovenous fistula, atherosclerosis, hypertension, vasculitis, Raynaud's disease, aneurysms, arterial dissections, varicose veins, thrombophlebitis and phlebothrombosis, vascular tumors, and complications of thrombolysis, balloon angioplasty, vascular replacement, coronary artery bypass graft surgery, congestive heart failure, ischemic heart disease, angina pectoris, myocardial infarction, hypertensive heart disease, degenerative valvular heart disease, calcific aortic valve stenosis, congenitally bicuspid aortic valve, mitral annular calcification, mitral valve prolapse, rheumatic fever and rheumatic heart disease, infective endocarditis, nonbacterial thrombotic endocarditis, endocarditis of systemic lupus erythematosus, carcinoid heart disease, cardiomyopathy, myocarditis, pericarditis, neoplastic heart disease, congenital heart disease, complications of cardiac transplantation, congenital lung anomalies, atelectasis, pulmonary congestion and edema, pulmonary embolism, pulmonary hemorrhage, pulmonary infarction, pulmonary hypertension, vascular sclerosis, obstructive pulmonary disease, restrictive pulmonary disease, chronic obstructive pulmonary disease, emphysema, chronic bronchitis, bronchial asthma, bronchiectasis, bacterial pneumonia, viral and mycoplasmal pneumonia, lung abscess, pulmonary tuberculosis, diffuse interstitial diseases, pneumoconioses, sarcoidosis, idiopathic

pulmonary fibrosis, desquamative interstitial pneumonitis, hypersensitivity pneumonitis, pulmonary eosinophilia bronchiolitis obliterans-organizing pneumonia, diffuse pulmonary hemorrhage syndromes, Goodpasture's syndromes, idiopathic pulmonary hemosiderosis, pulmonary involvement in collagen-vascular disorders, pulmonary alveolar proteinosis, lung tumors, inflammatory and noninflammatory pleural effusions, pneumothorax, pleural tumors, drug-induced lung disease, radiation-induced lung disease, and complications of lung transplantation ; a reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, ectopic pregnancy, teratogenesis, cancer of the breast, fibrocystic breast disease, galactorrhea, a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puberty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumours; a gastrointestinal disorder such as dysphagia, peptic esophagitis, esophageal spasm, esophageal stricture, esophageal carcinoma, dyspepsia, indigestion, gastritis, gastric carcinoma, anorexia, nausea, emesis, gastroparesis, antral or pyloric edema, abdominal angina, pyrosis, gastroenteritis, intestinal obstruction, infections of the intestinal tract, peptic ulcer, cholelithiasis, cholecystitis, cholestasis, pancreatitis, pancreatic carcinoma, biliary tract disease, hepatitis, hyperbilirubinemia, cirrhosis, passive congestion of the liver, hepatoma, infectious colitis, ulcerative colitis, ulcerative proctitis, Crohn's disease, Whipple's disease, Mallory-Weiss syndrome, colonic carcinoma, colonic obstruction, irritable bowel syndrome, short bowel syndrome, diarrhea, constipation, gastrointestinal hemorrhage, acquired immunodeficiency syndrome (AIDS) enteropathy, jaundice, hepatic encephalopathy, hepatorenal syndrome, hepatic steatosis, hemochromatosis, Wilson's disease, alpha- antitrypsin deficiency, Reye's syndrome, primary sclerosing cholangitis, liver infarction, portal vein obstruction and thrombosis, centrilobular necrosis, peliosis hepatis, hepatic vein thrombosis, veno- occlusive disease, preeclampsia, eclampsia, acute fatty liver of pregnancy, intrahepatic cholestasis of pregnancy, and hepatic tumors including nodular hyperplasias, adenomas, and carcinomas; a genetic disorder such as adrenoleukodystrophy, Alport's syndrome, choroideremia, Duchenne and Becker muscular dystrophy, Down's syndrome, cystic fibrosis, chronic granulomatous disease, Gaucher's disease, Huntington's chorea, Marfan's syndrome, muscular dystrophy, myotonic dystrophy, pycnodysostosis, Refsum's syndrome, retinoblastoma, sickle cell anemia, thalassemia, Werner syndrome, von Willebrand's disease, Wilms'tumor, Zellweger syndrome, peroxisomal acyl-CoA

oxidase deficiency, peroxisomal thiolase deficiency, peroxisomal bifunctional protein deficiency, mitochondrial carnitine palmitoyl transferase and carnitine deficiency, mitochondrial very-long-chain acyl-CoA dehydrogenase deficiency, mitochondrial medium-chain acyl-CoA dehydrogenase deficiency, mitochondrial short-chain acyl-CoA dehydrogenase deficiency, mitochondrial electron transport flavoprotein and electron transport flavoprotein : ubiquinone oxidoreductase deficiency, mitochondrial trifunctional protein deficiency, and mitochondrial short-chain 3-hydroxyacyl-CoA dehydrogenase deficiency; and a lipid metabolism disorder such as fatty liver, cholestasis, primary biliary cirrhosis, carnitine deficiency, carnitine palmitoyltransferase deficiency, myoadenylate deaminase deficiency, hypertriglyceridemia, lipid storage disorders such Fabry's disease, Gaucher's disease, Niemann-Pick's disease, metachromatic leukodystrophy, adrenoleukodystrophy, GM2 gangliosidosis, and ceroid lipofuscinosis, abetalipoproteinemia, Tangier disease, hyperlipoproteinemia, diabetes mellitus, lipodystrophy, lipomatoses, acute panniculitis, disseminated fat necrosis, adiposis dolorosa, lipoid adrenal hyperplasia, minimal change disease, lipomas, atherosclerosis, hypercholesterolemia, hypercholesterolemia with hypertriglyceridemia, primary hypoalphalipoproteinemia, hypothyroidism, renal disease, liver disease, lecithin: cholesterol acyltransferase deficiency, cerebrotendinous xanthomatosis, sitosterolemia, hypocholesterolemia, Tay-Sachs disease, Sandhoff's disease, hyperlipidemia, hyperlipemia, and lipid myopathies; cancer such as adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; and an infection by a viral agent classified as adenovirus, arenavirus, bunyavirus, calicivirus, coronavirus, filovirus, hepadnavirus, herpesvirus, flavivirus, orthomyxovirus, parvovirus, papovavirus, paramyxovirus, picornavirus, poxvirus, reovirus, retrovirus, rhabdovirus, and tongavirus.

In another embodiment, a vector capable of expressing REMAP or a fragment or derivative thereof may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of REMAP including, but not limited to, those described above.

In a further embodiment, a composition comprising a substantially purified REMAP in conjunction with a suitable pharmaceutical carrier may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of REMAP including, but not limited to, those provided above.

In still another embodiment, an agonist which modulates the activity of REMAP may be administered to a subject to treat or prevent a disorder associated with decreased expression or activity of REMAP including, but not limited to, those listed above.

In a further embodiment, an antagonist of REMAP may be administered to a subject to treat

or prevent a disorder associated with increased expression or activity of REMAP. Examples of such disorders include, but are not limited to, those cell proliferative, autoimmune/inflammatory, neurological, metabolic, developmental, endocrine, cardiovascular, reproductive, gastrointestinal, metabolic, genetic, and lipid metabolism disorders, cancer, and viral infections described above. In one aspect, an antibody which specifically binds REMAP may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express REMAP.

In an additional embodiment, a vector expressing the complement of the polynucleotide encoding REMAP may be administered to a subject to treat or prevent a disorder associated with increased expression or activity of REMAP including, but not limited to, those described above.

In other embodiments, any of the proteins, antagonists, antibodies, agonists, complementary sequences, or vectors of the invention may be administered in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.

An antagonist of REMAP may be produced using methods which are generally known in the art. In particular, purified REMAP may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind REMAP. Antibodies to REMAP may also be generated using methods that are well known in the art. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric, and single chain antibodies, Fab fragments, and fragments produced by a Fab expression library. Neutralizing antibodies (i. e., those which inhibit dimer formation) are generally preferred for therapeutic use. Single chain antibodies (e. g., from camels or llamas) may be potent enzyme inhibitors and may have advantages in the design of peptide mimetics, and in the development of immuno-adsorbents and biosensors (Muyldermans, S. (2001) J.

Biotechnol. 74: 277-302).

For the production of antibodies, various hosts including goats, rabbits, rats, mice, camels, dromedaries, llamas, humans, and others may be immunized by injection with REMAP or with any fragment or oligopeptide thereof which has immunogenic properties. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, KLH, and dinitrophenol.

Among adjuvants used in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are especially preferable.

It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to REMAP have an amino acid sequence consisting of at least about 5 amino acids, and generally will consist of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the natural protein. Short stretches of REMAP amino acids may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced.

Monoclonal antibodies to REMAP may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (See, e. g., Kohler, G. et al. (1975) Nature 256: 495-497; Kozbor, D. et al. (1985) J.

Immunol. Methods 81: 31-42; Cote, R. J. et al. (1983) Proc. Natl. Acad. Sci. USA 80: 2026-2030; and Cole, S. P. et al. (1984) Mol. Cell Biol. 62: 109-120.) In addition, techniques developed for the production of"chimeric antibodies,"such as the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity, can be used. (See, e. g., Morrison, S. L. et al. (1984) Proc.

Natl. Acad. Sci. USA 81: 6851-6855; Neuberger, M. S. et al. (1984) Nature 312: 604-608; and Takeda, S. et al. (1985) Nature 314: 452-454.) Alternatively, techniques described for the production of single chain antibodies may be adapted, using methods known in the art, to produce REMAP-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries. (See, e. g., Burton, D. R. (1991) Proc. Natl. Acad. Sci. USA 88: 10134-10137.) Antibodies may also be produced by inducing irez vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (See, e. g., Orlandi, R. et al. (1989) Proc. Natl. Acad. Sci. USA 86: 3833-3837; Winter, G. et al. (1991) Nature 349: 293-299.) Antibody fragments which contain specific binding sites for REMAP may also be generated.

For example, such fragments include, but are not limited to, F (ab') 2 fragments produced by pepsin digestion of the antibody molecule and Fab fragments generated by reducing the disulfide bridges of the F (ab') 2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (See, e. g., Huse, W. D. et al. (1989) Science 246: 1275-1281.) Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between REMAP and its

specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to two non-interfering REMAP epitopes is generally used, but a competitive binding assay may also be employed (Pound, supra).

Various methods such as Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for REMAP. Affinity is expressed as an association constant, Ka which is defined as the molar concentration of REMAP-antibody complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions.

The Ka determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple REMAP epitopes, represents the average affinity, or avidity, of the antibodies for REMAP. The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular REMAP epitope, represents a true measure of affinity. High-affinity antibody preparations with Ka ranging from about 109 to 10l2 L/mole are preferred for use in immunoassays in which the REMAP-antibody complex must withstand rigorous manipulations. Low-affinity antibody preparations with Ka ranging from about 106 to 10'L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of REMAP, preferably in active form, from the antibody (Catty, D. (1988) Antibodies, Volume I : A Practical Approach, IRL Press, Washington DC; Liddell, J. E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies, John Wiley & Sons, New York NY).

The titer and avidity of polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications. For example, a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml, preferably 5-10 mg specific antibody/ml, is generally employed in procedures requiring precipitation of REMAP-antibody complexes. Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available. (See, e. g., Catty, supra, and Coligan et al. supra.) In another embodiment of the invention, the polynucleotides encoding REMAP, or any fragment or complement thereof, may be used for therapeutic purposes. In one aspect, modifications of gene expression can be achieved by designing complementary sequences or antisense molecules (DNA, RNA, PNA, or modified oligonucleotides) to the coding or regulatory regions of the gene encoding REMAP. Such technology is well known in the art, and antisense oligonucleotides or larger fragments can be designed from various locations along the coding or control regions of sequences encoding REMAP. (See, e. g., Agrawal, S., ed. (1996) Antisense Therapeutics, Humana Press Inc., Totawa NJ.) In therapeutic use, any gene delivery system suitable for introduction of the antisense sequences into appropriate target cells can be used. Antisense sequences can be delivered

intracellularly in the form of an expression plasmid which, upon transcription, produces a sequence complementary to at least a portion of the cellular sequence encoding the target protein. (See, e. g., Slater, J. E. et al. (1998) J. Allergy Clin. Immunol. 102 (3): 469-475; and Scanlon, K. J. et al. (1995) 9 (13): 1288-1296.) Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors. (See, e. g., Miller, A. D. (1990) Blood 76: 271; Ausubel, supra ; Uckert, W. and W. Walther (1994) Pharmacol. Ther. 63 (3): 323-347.) Other gene delivery mechanisms include liposome-derived systems, artificial viral envelopes, and other systems known in the art. (See, e. g., Rossi, J. J. (1995) Br. Med. Bull. 51 (1) : 217-225; Boado, R. J. et al. (1998) J. Pharm. Sci. 87 (11): 1308-1315; and Morris, M. C. et al. (1997) Nucleic Acids Res.

25 (14): 2730-2736.) In another embodiment of the invention, polynucleotides encoding REMAP may be used for somatic or germline gene therapy. Gene therapy may be performed to (i) correct a genetic deficiency (e. g., in the cases of severe combined immunodeficiency (SCID)-Xl disease characterized by X- linked inheritance (Cavazzana-Calvo, M. et al. (2000) Science 288: 669-672), severe combined immunodeficiency syndrome associated with an inherited adenosine deaminase (ADA) deficiency (Blaese, R. M. et al. (1995) Science 270: 475-480; Bordignon, C. et al. (1995) Science 270: 470-475), cystic fibrosis (Zabner, J. et al. (1993) Cell 75 : 207-216; Crystal, R. G. et al. (1995) Hum. Gene Therapy 6: 643-666; Crystal, R. G. et al. (1995) Hum. Gene Therapy 6: 667-703), thalassamias, familial hypercholesterolemia, and hemophilia resulting from Factor VIII or Factor IX deficiencies (Crystal, R. G. (1995) Science 270: 404-410; Verma, I. M. and N. Somia (1997) Nature 389: 239-242)), (ii) express a conditionally lethal gene product (e. g., in the case of cancers which result from unregulated cell proliferation), or (iii) express a protein which affords protection against intracellular parasites (e. g., against human retroviruses, such as human immunodeficiency virus (HIV) (Baltimore, D.

(1988) Nature 335: 395-396; Poeschla, E. et al. (1996) Proc. Natl. Acad. Sci. USA 93: 11395-11399), hepatitis B or C virus (HBV, HCV); fungal parasites, such as Candida albicans and Paracoccidioides brasiliensis ; and protozoan parasites such as Plasmodium falciparum and Trypanosoma cruzi). In the case where a genetic deficiency in REMAP expression or regulation causes disease, the expression of REMAP from an appropriate population of transduced cells may alleviate the clinical manifestations caused by the genetic deficiency.

In a further embodiment of the invention, diseases or disorders caused by deficiencies in REMAP are treated by constructing mammalian expression vectors encoding REMAP and introducing these vectors by mechanical means into REMAP-deficient cells. Mechanical transfer technologies for use with cells in vivo or ex vitro include (i) direct DNA microinjection into individual cells, (ii) ballistic gold particle delivery, (iii) liposome-mediated transfection, (iv) receptor- mediated gene transfer, and (v) the use of DNA transposons (Morgan, R. A. and W. F. Anderson

(1993) Annu. Rev. Biochem. 62: 191-217; Ivics, Z. (1997) Cell 91: 501-510; Boulay, J-L. and H.

Recipon (1998) Curr. Opin. Biotechnol. 9: 445-450).

Expression vectors that may be effective for the expression of REMAP include, but are not limited to, the PCDNA 3.1, EPITAG, PRCCMV2, PREP, PVAX, PCR2-TOPOTA vectors (Invitrogen, Carlsbad CA), PCMV-SCRIPT, PCMV-TAG, PEGSH/PERV (Stratagene, La Jolla CA), and PTET-OFF, PTET-ON, PTRE2, PTRE2-LUC, PTK-HYG (Clontech, Palo Alto CA). REMAP may be expressed using (i) a constitutively active promoter, (e. g., from cytomegalovirus (CMV), Rous sarcoma virus (RSV), SV40 virus, thymidine kinase (TK), or p-actin genes), (ii) an inducible promoter (e. g., the tetracycline-regulated promoter (Gossen, M. and H. Bujard (1992) Proc. Natl.

Acad. Sci. USA 89: 5547-5551; Gossen, M. et al. (1995) Science 268: 1766-1769; Rossi, F. M. V. and H. M. Blau (1998) Curr. Opin. Biotechnol. 9: 451-456), commercially available in the T-REX plasmid (Invitrogen)) ; the ecdysone-inducible promoter (available in the plasmids PVGRXR and PIND ; Invitrogen) ; the FK506/rapamycin inducible promoter; or the RU486/mifepristone inducible promoter (Rossi, F. M. V. and H. M. Blau, supra)), or (iii) a tissue-specific promoter or the native promoter of the endogenous gene encoding REMAP from a normal individual.

Commercially available liposome transformation kits (e. g., the PERFECT LIPID TRANSFECTION KIT, available from Invitrogen) allow one with ordinary skill in the art to deliver polynucleotides to target cells in culture and require minimal effort to optimize experimental parameters. In the alternative, transformation is performed using the calcium phosphate method (Graham, F. L. and A. J. Eb (1973) Virology 52: 456-467), or by electroporation (Neumann, E. et al.

(1982) EMBO J. 1: 841-845). The introduction of DNA to primary cells requires modification of these standardized mammalian transfection protocols.

In another embodiment of the invention, diseases or disorders caused by genetic defects with respect to REMAP expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding REMAP under the control of an independent promoter or the retrovirus long terminal repeat (LTR) promoter, (ii) appropriate RNA packaging signals, and (iii) a Rev-responsive element (RRE) along with additional retrovirus cis-acting RNA sequences and coding sequences required for efficient vector propagation. Retrovirus vectors (e. g., PFB and PFBNEO) are commercially available (Stratagene) and are based on published data (Riviere, I. et al. (1995) Proc.

Natl. Acad. Sci. USA 92: 6733-6737), incorporated by reference herein. The vector is propagated in an appropriate vector producing cell line (VPCL) that expresses an envelope gene with a tropism for receptors on the target cells or a promiscuous envelope protein such as VSVg (Armentano, D. et al.

(1987) J. Virol. 61: 1647-1650; Bender, M. A. et al. (1987) J. Virol. 61: 1639-1646; Adam, M. A. and A. D. Miller (1988) J. Virol. 62: 3802-3806; Dull, T. et al. (1998) J. Virol. 72: 8463-8471; Zufferey, R. et al. (1998) J. Virol. 72: 9873-9880). U. S. Patent No. 5,910,434 to Rigg ("Method for obtaining

retrovirus packaging cell lines producing high transducing efficiency retroviral supernatant") discloses a method for obtaining retrovirus packaging cell lines and is hereby incorporated by reference. Propagation of retrovirus vectors, transduction of a population of cells (e. g., CD4+ T- cells), and the return of transduced cells to a patient are procedures well known to persons skilled in the art of gene therapy and have been well documented (Ranga, U. et al. (1997) J. Virol. 71: 7020- 7029; Bauer, G. et al. (1997) Blood 89: 2259-2267; Bonyhadi, M. L. (1997) J. Virol. 71: 4707-4716; Ranga, U. et al. (1998) Proc. Natl. Acad. Sci. USA 95: 1201-1206; Su, L. (1997) Blood 89: 2283- 2290).

In the alternative, an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding REMAP to cells which have one or more genetic abnormalities with respect to the expression of REMAP. The construction and packaging of adenovirus-based vectors are well known to those with ordinary skill in the art. Replication defective adenovirus vectors have proven to be versatile for importing genes encoding immunoregulatory proteins into intact islets in the pancreas (Csete, M. E. et al. (1995) Transplantation 27: 263-268). Potentially useful adenoviral vectors are described in U. S. Patent No. 5,707,618 to Armentano ("Adenovirus vectors for gene therapy"), hereby incorporated by reference. For adenoviral vectors, see also Antinozzi, P. A. et al. (1999) Annu. Rev. Nutr. 19: 511-544 and Verma, I. M. and N. Somia (1997) Nature 18: 389: 239-242, both incorporated by reference herein.

In another alternative, a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding REMAP to target cells which have one or more genetic abnormalities with respect to the expression of REMAP. The use of herpes simplex virus (HSV)-based vectors may be especially valuable for introducing REMAP to cells of the central nervous system, for which HSV has a tropism. The construction and packaging of herpes-based vectors are well known to those with ordinary skill in the art. A replication-competent herpes simplex virus (HSV) type 1-based vector has been used to deliver a reporter gene to the eyes of primates (Liu, X. et al. (1999) Exp. Eye Res.

169: 385-395). The construction of a HSV-1 virus vector has also been disclosed in detail in U. S.

Patent No. 5,804,413 to DeLuca ("Herpes simplex virus strains for gene transfer"), which is hereby incorporated by reference. U. S. Patent No. 5,804,413 teaches the use of recombinant HSV d92 which consists of a genome containing at least one exogenous gene to be transferred to a cell under the control of the appropriate promoter for purposes including human gene therapy. Also taught by this patent are the construction and use of recombinant HSV strains deleted for ICP4, ICP27 and ICP22.

For HSV vectors, see also Goins, W. F. et al. (1999) J. Virol. 73: 519-532 and Xu, H. et al. (1994) Dev. Biol. 163: 152-161, hereby incorporated by reference. The manipulation of cloned herpesvirus sequences, the generation of recombinant virus following the transfection of multiple plasmids containing different segments of the large herpesvirus genomes, the growth and propagation of

herpesvirus, and the infection of cells with herpesvirus are techniques well known to those of ordinary skill in the art.

In another alternative, an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding REMAP to target cells. The biology of the prototypic alphavirus, Semliki Forest Virus (SFV), has been studied extensively and gene transfer vectors have been based on the SFV genome (Garoff, H. and K.-J. Li (1998) Curr. Opin. Biotechnol. 9: 464-469). During alphavirus RNA replication, a subgenomic RNA is generated that normally encodes the viral capsid proteins. This subgenomic RNA replicates to higher levels than the full length genomic RNA, resulting in the overproduction of capsid proteins relative to the viral proteins with enzymatic activity (e. g., protease and polymerase). Similarly, inserting the coding sequence for REMAP into the alphavirus genome in place of the capsid-coding region results in the production of a large number of REMAP-coding RNAs and the synthesis of high levels of REMAP in vector transduced cells. While alphavirus infection is typically associated with cell lysis within a few days, the ability to establish a persistent infection in hamster normal kidney cells (BHK-21) with a variant of Sindbis virus (SIN) indicates that the lytic replication of alphaviruses can be altered to suit the needs of the gene therapy application (Dryga, S. A. et al. (1997) Virology 228: 74-83). The wide host range of alphaviruses will allow the introduction of REMAP into a variety of cell types. The specific transduction of a subset of cells in a population may require the sorting of cells prior to transduction. The methods of manipulating infectious cDNA clones of alphaviruses, performing alphavirus cDNA and RNA transfections, and performing alphavirus infections, are well known to those with ordinary skill in the art.

Oligonucleotides derived from the transcription initiation site, e. g., between about positions - 10 and +10 from the start site, may also be employed to inhibit gene expression. Similarly, inhibition can be achieved using triple helix base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or regulatory molecules. Recent therapeutic advances using triplex DNA have been described in the literature. (See, e. g., Gee, J. E. et al. (1994) in Huber, B. E. and B. I. Carr, Molecular and Immunologic Approaches, Futura Publishing, Mt. Kisco NY, pp. 163- 177.) A complementary sequence or antisense molecule may also be designed to block translation of mRNA by preventing the transcript from binding to ribosomes.

Ribozymes, enzymatic RNA molecules, may also be used to catalyze the specific cleavage of RNA. The mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. For example, engineered hammerhead motif ribozyme molecules may specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding REMAP.

Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, including the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides, corresponding to the region of the target gene containing the cleavage site, may be evaluated for secondary structural features which may render the oligonucleotide inoperable. The suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays.

Complementary ribonucleic acid molecules and ribozymes of the invention may be prepared by any method known in the art for the synthesis of nucleic acid molecules. These include techniques for chemically synthesizing oligonucleotides such as solid phase phosphoramidite chemical synthesis.

Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding REMAP. Such DNA sequences may be incorporated into a wide variety of vectors with suitable RNA polymerase promoters such as T7 or SP6. Alternatively, these cDNA constructs that synthesize complementary RNA, constitutively or inducibly, can be introduced into cell lines, cells, or tissues.

RNA molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5'and/or 3' ends of the molecule, or the use of phosphorothioate or 2'0-methyl rather than phosphodiesterase linkages within the backbone of the molecule. This concept is inherent in the production of PNAs and can be extended in all of these molecules by the inclusion of nontraditional bases such as inosine, queosine, and wybutosine, as well as acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases.

An additional embodiment of the invention encompasses a method for screening for a compound which is effective in altering expression of a polynucleotide encoding REMAP.

Compounds which may be effective in altering expression of a specific polynucleotide may include, but are not limited to, oligonucleotides, antisense oligonucleotides, triple helix-forming oligonucleotides, transcription factors and other polypeptide transcriptional regulators, and non- macromolecular chemical entities which are capable of interacting with specific polynucleotide sequences. Effective compounds may alter polynucleotide expression by acting as either inhibitors or promoters of polynucleotide expression. Thus, in the treatment of disorders associated with increased REMAP expression or activity, a compound which specifically inhibits expression of the polynucleotide encoding REMAP may be therapeutically useful, and in the treatment of disorders associated with decreased REMAP expression or activity, a compound which specifically promotes expression of the polynucleotide encoding REMAP may be therapeutically useful.

At least one, and up to a plurality, of test compounds may be screened for effectiveness in altering expression of a specific polynucleotide. A test compound may be obtained by any method commonly known in the art, including chemical modification of a compound known to be effective in altering polynucleotide expression; selection from an existing, commercially-available or proprietary library of naturally-occurring or non-natural chemical compounds; rational design of a compound based on chemical and/or structural properties of the target polynucleotide; and selection from a library of chemical compounds created combinatorially or randomly. A sample comprising a polynucleotide encoding REMAP is exposed to at least one test compound thus obtained. The sample may comprise, for example, an intact or permeabilized cell, or an in vitro cell-free or reconstituted biochemical system. Alterations in the expression of a polynucleotide encoding REMAP are assayed by any method commonly known in the art. Typically, the expression of a specific nucleotide is detected by hybridization with a probe having a nucleotide sequence complementary to the sequence of the polynucleotide encoding REMAP. The amount of hybridization may be quantified, thus forming the basis for a comparison of the expression of the polynucleotide both with and without exposure to one or more test compounds. Detection of a change in the expression of a polynucleotide exposed to a test compound indicates that the test compound is effective in altering the expression of the polynucleotide. A screen for a compound effective in altering expression of a specific polynucleotide can be carried out, for example, using a Schizosaccharomyces ponzbe gene expression system (Atkins, D. et al. (1999) U. S. Patent No.

5,932,435; Arndt, G. M. et al. (2000) Nucleic Acids Res. 28: E15) or a human cell line such as HeLa cell (Clarke, M. L. et al. (2000) Biochem. Biophys. Res. Commun. 268: 8-13). A particular embodiment of the present invention involves screening a combinatorial library of oligonucleotides (such as deoxyribonucleotides, ribonucleotides, peptide nucleic acids, and modified oligonucleotides) for antisense activity against a specific polynucleotide sequence (Bruice, T. W. et al. (1997) U. S.

Patent No. 5,686,242; Bruice, T. W. et al. (2000) U. S. Patent No. 6,022,691).

Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient.

Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See, e. g., Goldman, C. K. et al. (1997) Nat.

Biotechnol. 15: 462-466.) Any of the therapeutic methods described above may be applied to any subject in need of such therapy, including, for example, mammals such as humans, dogs, cats, cows, horses, rabbits, and monkeys.

An additional embodiment of the invention relates to the administration of a composition

which generally comprises an active ingredient formulated with a pharmaceutically acceptable excipient. Excipients may include, for example, sugars, starches, celluloses, gums, and proteins.

Various formulations are commonly known and are thoroughly discussed in the latest edition of Remington's Pharmaceutical Sciences (Maack Publishing, Easton PA). Such compositions may consist of REMAP, antibodies to REMAP, and mimetics, agonists, antagonists, or inhibitors of REMAP.

The compositions utilized in this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, pulmonary, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, or rectal means.

Compositions for pulmonary administration may be prepared in liquid or dry powder form.

These compositions are generally aerosolized immediately prior to inhalation by the patient. In the case of small molecules (e. g. traditional low molecular weight organic drugs), aerosol delivery of fast-acting formulations is well-known in the art. In the case of macromolecules (e. g. larger peptides and proteins), recent developments in the field of pulmonary delivery via the alveolar region of the lung have enabled the practical delivery of drugs such as insulin to blood circulation (see, e. g., Patton, J. S. et al., U. S. Patent No. 5,997,848). Pulmonary delivery has the advantage of administration without needle injection, and obviates the need for potentially toxic penetration enhancers.

Compositions suitable for use in the invention include compositions wherein the active ingredients are contained in an effective amount to achieve the intended purpose. The determination of an effective dose is well within the capability of those skilled in the art.

Specialized forms of compositions may be prepared for direct intracellular delivery of macromolecules comprising REMAP or fragments thereof. For example, liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule. Alternatively, REMAP or a fragment thereof may be joined to a short cationic N- terminal portion from the HIV Tat-1 protein. Fusion proteins thus generated have been found to transduce into the cells of all tissues, including the brain, in a mouse model system (Schwarze, S. R. et al. (1999) Science 285: 1569-1572).

For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays, e. g., of neoplastic cells, or in animal models such as mice, rats, rabbits, dogs, monkeys, or pigs. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.

A therapeutically effective dose refers to that amount of active ingredient, for example REMAP or fragments thereof, antibodies of REMAP, and agonists, antagonists or inhibitors of

REMAP, which ameliorates the symptoms or condition. Therapeutic efficacy and toxicity may be determined by standard pharmaceutical procedures in cell cultures or with experimental animals, such as by calculating the ED50 (the dose therapeutically effective in 50% of the population) or LD50 (the dose lethal to 50% of the population) statistics. The dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the LD50/ED50 ratio. Compositions which exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and animal studies are used to formulate a range of dosage for human use. The dosage contained in such compositions is preferably within a range of circulating concentrations that includes the ED50 with little or no toxicity.

The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.

The exact dosage will be determined by the practitioner, in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active moiety or to maintain the desired effect. Factors which may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination (s), reaction sensitivities, and response to therapy. Long-acting compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation.

Normal dosage amounts may vary from about 0.1 Mg to 100,000, ug, up to a total dose of about 1 gram, depending upon the route of administration. Guidance as to particular dosages and methods of delivery is provided in the literature and generally available to practitioners in the art.

Those skilled in the art will employ different formulations for nucleotides than for proteins or their inhibitors. Similarly, delivery of polynucleotides or polypeptides will be specific to particular cells, conditions, locations, etc.

DIAGNOSTICS In another embodiment, antibodies which specifically bind REMAP may be used for the diagnosis of disorders characterized by expression of REMAP, or in assays to monitor patients being treated with REMAP or agonists, antagonists, or inhibitors of REMAP. Antibodies useful for diagnostic purposes may be prepared in the same manner as described above for therapeutics.

Diagnostic assays for REMAP include methods which utilize the antibody and a label to detect REMAP in human body fluids or in extracts of cells or tissues. The antibodies may be used with or without modification, and may be labeled by covalent or non-covalent attachment of a reporter molecule. A wide variety of reporter molecules, several of which are described above, are known in the art and may be used.

A variety of protocols for measuring REMAP, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of REMAP expression.

Normal or standard values for REMAP expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to REMAP under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of REMAP expressed in subject, control, and disease samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease.

In another embodiment of the invention, the polynucleotides encoding REMAP may be used for diagnostic purposes. The polynucleotides which may be used include oligonucleotide sequences, complementary RNA and DNA molecules, and PNAs. The polynucleotides may be used to detect and quantify gene expression in biopsied tissues in which expression of REMAP may be correlated with disease. The diagnostic assay may be used to determine absence, presence, and excess expression of REMAP, and to monitor regulation of REMAP levels during therapeutic intervention.

In one aspect, hybridization with PCR probes which are capable of detecting polynucleotide sequences, including genomic sequences, encoding REMAP or closely related molecules may be used to identify nucleic acid sequences which encode REMAP. The specificity of the probe, whether it is made from a highly specific region, e. g., the 5'regulatory region, or from a less specific region, e. g., a conserved motif, and the stringency of the hybridization or amplification will determine whether the probe identifies only naturally occurring sequences encoding REMAP, allelic variants, or related sequences.

Probes may also be used for the detection of related sequences, and may have at least 50% sequence identity to any of the REMAP encoding sequences. The hybridization probes of the subject invention may be DNA or RNA and may be derived from the sequence of SEQ ID NO : 27-52 or from genomic sequences including promoters, enhancers, and introns of the REMAP gene.

Means for producing specific hybridization probes for DNAs encoding REMAP include the cloning of polynucleotide sequences encoding REMAP or REMAP derivatives into vectors for the production of mRNA probes. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides. Hybridization probes may be labeled by a variety of reporter groups, for example, by radionuclides such ås 32p or 35S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, and the like.

Polynucleotide sequences encoding REMAP may be used for the diagnosis of disorders associated with expression of REMAP. Examples of such disorders include, but are not limited to, a cell proliferative disorder such as actinic keratosis, arteriosclerosis, atherosclerosis, bursitis, cirrhosis, hepatitis, mixed connective tissue disease (MCTD), myelofibrosis, paroxysmal nocturnal

hemoglobinuria, polycythemia vera, psoriasis, primary thrombocythemia, and cancers including adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; an autoimmune/inflammatory disorder such as acquired immunodeficiency syndrome (AIDS), Addison's disease, adult respiratory distress syndrome, allergies, ankylosing spondylitis, amyloidosis, anemia, asthma, atherosclerosis, autoimmune hemolytic anemia, autoimmune thyroiditis, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), bronchitis, cholecystitis, contact dermatitis, Crohn's disease, atopic dermatitis, dermatomyositis, diabetes mellitus, emphysema, episodic lymphopenia with lymphocytotoxins, erythroblastosis fetalis, erythema nodosum, atrophic gastritis, glomerulonephritis, Goodpasture's syndrome, gout, Graves' disease, Hashimoto's thyroiditis, hypereosinophilia, irritable bowel syndrome, multiple sclerosis, myasthenia gravis, myocardial or pericardial inflammation, osteoarthritis, osteoporosis, pancreatitis, polymyositis, psoriasis, Reiter's syndrome, rheumatoid arthritis, scleroderma, Sjogren's syndrome, systemic anaphylaxis, systemic lupus erythematosus, systemic sclerosis, thrombocytopenic purpura, ulcerative colitis, uveitis, Werner syndrome, complications of cancer, hemodialysis, and extracorporeal circulation, viral, bacterial, fungal, parasitic, protozoal, and helminthic infections, and trauma; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann- Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial

frontotemporal dementia; a metabolic disorder such as Addison's disease, cerebrotendinous xanthomatosis, congenital adrenal hyperplasia, coumarin resistance, cystic fibrosis, fatty hepatocirrhosis, fructose-1, 6-diphosphatase deficiency, galactosemia, goiter, glucagonoma, glycogen storage diseases, hereditary fructose intolerance, hyperadrenalism, hypoadrenalism, hyperparathyroidism, hypoparathyroidism, hypercholesterolemia, hyperthyroidism, hypoglycemia, hypothyroidism, hyperlipidemia, hyperlipemia, lipid myopathies, lipodystrophies, lysosomal storage diseases, mannosidosis, neuraminidase deficiency, obesity, osteoporosis, phenylketonuria, pseudovitamin D-deficiency rickets, disorders of carbohydrate metabolism such as congenital type II dyserythropoietic anemia, diabetes, insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, galactose epimerase deficiency, glycogen storage diseases, lysosomal storage diseases, fructosuria, pentosuria, and inherited abnormalities of pyruvate metabolism, disorders of lipid metabolism such as fatty liver, cholestasis, primary biliary cirrhosis, camitine deficiency, carnitine palmitoyltransferase deficiency, myoadenylate deaminase deficiency, hypertriglyceridemia, lipid storage disorders such Fabry's disease, Gaucher's disease, Niemann-Pick's disease, metachromatic leukodystrophy, adrenoleukodystrophy, GM2 gangliosidosis, and ceroid lipofuscinosis, abetalipoproteinemia, Tangier disease, hyperlipoproteinemia, lipodystrophy, lipomatoses, acute panniculitis, disseminated fat necrosis, adiposis dolorosa, lipoid adrenal hyperplasia, minimal change disease, lipomas, atherosclerosis, hypercholesterolemia, hypercholesterolemia with hypertriglyceridemia, primary hypoalphalipoproteinemia, hypothyroidism, renal disease, liver disease, lecithin: cholesterol acyltransferase deficiency, cerebrotendinous xanthomatosis, sitosterolemia, hypocholesterolemia, Tay-Sachs disease, Sandhoff's disease, hyperlipidemia, hyperlipemia, and lipid myopathies, and disorders of copper metabolism such as Menke's disease, Wilson's disease, and Ehlers-Danlos syndrome type IX diabetes; a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms' tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, a seizure disorder such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss; and an endocrine disorder such as a disorder of the hypothalamus and/or pituitary resulting from lesions such as a primary brain tumor, adenoma, infarction associated with pregnancy, hypophysectomy, aneurysm, vascular malformation, thrombosis, infection, immunological disorder, and complication due to head trauma, a disorder associated with hypopituitarism including hypogonadism, Sheehan syndrome, diabetes insipidus, Kallman's disease, Hand-Schuller-Christian disease, Letterer-Siwe

disease, sarcoidosis, empty sella syndrome, and dwarfism, a disorder associated with hyperpituitarism including acromegaly, giantism, and syndrome of inappropriate antidiuretic hormone (ADH) secretion (SIADH) often caused by benign adenoma, a disorder associated with hypothyroidism including goiter, myxedema, acute thyroiditis associated with bacterial infection, subacute thyroiditis associated with viral infection, autoimmune thyroiditis (Hashimoto's disease), and cretinism, a disorder associated with hyperthyroidism including thyrotoxicosis and its various forms, Grave's disease, pretibial myxedema, toxic multinodular goiter, thyroid carcinoma, and Plummer's disease, a disorder associated with hyperparathyroidism including Conn disease (chronic hypercalemia), a pancreatic disorder such as Type I or Type II diabetes mellitus and associated complications, a disorder associated with the adrenals such as hyperplasia, carcinoma, or adenoma of the adrenal cortex, hypertension associated with alkalosis, amyloidosis, hypokalemia, Cushing's disease, Liddle's syndrome, and Arnold-Healy-Gordon syndrome, pheochromocytoma tumors, and Addison's disease, a disorder associated with gonadal steroid hormones such as: in women, abnormal prolactin production, infertility, endometriosis, perturbation of the menstrual cycle, polycystic ovarian disease, hyperprolactinemia, isolated gonadotropin deficiency, amenorrhea, galactorrhea, hermaphroditism, hirsutism and virilization, breast cancer, and, in post-menopausal women, osteoporosis, and, in men, Leydig cell deficiency, male climacteric phase, and germinal cell aplasia, a hypergonadal disorder associated with Leydig cell tumors, androgen resistance associated with absence of androgen receptors, and syndrome of 5 a-reductase ; a cardiovascular disorder such as arteriovenous fistula, atherosclerosis, hypertension, vasculitis, Raynaud's disease, aneurysms, arterial dissections, varicose veins, thrombophlebitis and phlebothrombosis, vascular tumors, and complications of thrombolysis, balloon angioplasty, vascular replacement, coronary artery bypass graft surgery, congestive heart failure, ischemic heart disease, angina pectoris, myocardial infarction, hypertensive heart disease, degenerative valvular heart disease, calcific aortic valve stenosis, congenitally bicuspid aortic valve, mitral annular calcification, mitral valve prolapse, rheumatic fever and rheumatic heart disease, infective endocarditis, nonbacterial thrombotic endocarditis, endocarditis of systemic lupus erythematosus, carcinoid heart disease, cardiomyopathy, myocarditis, pericarditis, neoplastic heart disease, congenital heart disease, complications of cardiac transplantation, congenital lung anomalies, atelectasis, pulmonary congestion and edema, pulmonary embolism, pulmonary hemorrhage, pulmonary infarction, pulmonary hypertension, vascular sclerosis, obstructive pulmonary disease, restrictive pulmonary disease, chronic obstructive pulmonary disease, emphysema, chronic bronchitis, bronchial asthma, bronchiectasis, bacterial pneumonia, viral and mycoplasmal pneumonia, lung abscess, pulmonary tuberculosis, diffuse interstitial diseases, pneumoconioses, sarcoidosis, idiopathic pulmonary fibrosis, desquamative interstitial pneumonitis, hypersensitivity pneumonitis, pulmonary eosinophilia bronchiolitis obliterans-organizing pneumonia, diffuse pulmonary hemorrhage

syndromes, Goodpasture's syndromes, idiopathic pulmonary hemosiderosis, pulmonary involvement in collagen-vascular disorders, pulmonary alveolar proteinosis, lung tumors, inflammatory and noninflammatory pleural effusions, pneumothorax, pleural tumors, drug-induced lung disease, radiation-induced lung disease, and complications of lung transplantation; a reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, ectopic pregnancy, teratogenesis, cancer of the breast, fibrocystic breast disease, galactorrhea, a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puberty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumours; a gastrointestinal disorder such as dysphagia, peptic esophagitis, esophageal spasm, esophageal stricture, esophageal carcinoma, dyspepsia, indigestion, gastritis, gastric carcinoma, anorexia, nausea, emesis, gastroparesis, antral or pyloric edema, abdominal angina, pyrosis, gastroenteritis, intestinal obstruction, infections of the intestinal tract, peptic ulcer, cholelithiasis, cholecystitis, cholestasis, pancreatitis, pancreatic carcinoma, biliary tract disease, hepatitis, hyperbilirubinemia, cirrhosis, passive congestion of the liver, hepatoma, infectious colitis, ulcerative colitis, ulcerative proctitis, Crohn's disease, Whipple's disease, Mallory-Weiss syndrome, colonic carcinoma, colonic obstruction, irritable bowel syndrome, short bowel syndrome, diarrhea, constipation, gastrointestinal hemorrhage, acquired immunodeficiency syndrome (AIDS) enteropathy, jaundice, hepatic encephalopathy, hepatorenal syndrome, hepatic steatosis, hemochromatosis, Wilson's disease, alphal- antitrypsin deficiency, Reye's syndrome, primary sclerosing cholangitis, liver infarction, portal vein obstruction and thrombosis, centrilobular necrosis, peliosis hepatis, hepatic vein thrombosis, veno- occlusive disease, preeclampsia, eclampsia, acute fatty liver of pregnancy, intrahepatic cholestasis of pregnancy, and hepatic tumors including nodular hyperplasias, adenomas, and carcinomas; a genetic disorder such as adrenoleukodystrophy, Alport's syndrome, choroideremia, Duchenne and Becker muscular dystrophy, Down's syndrome, cystic fibrosis, chronic granulomatous disease, Gaucher's disease, Huntington's chorea, Marfan's syndrome, muscular dystrophy, myotonic dystrophy, pycnodysostosis, Refsum's syndrome, retinoblastoma, sickle cell anemia, thalassemia, Werner syndrome, von Willebrand's disease, Wilms'tumor, Zellweger syndrome, peroxisomal acyl-CoA oxidase deficiency, peroxisomal thiolase deficiency, peroxisomal bifunctional protein deficiency, mitochondrial carnitine palmitoyl transferase and carnitine deficiency, mitochondrial very-long-chain

acyl-CoA dehydrogenase deficiency, mitochondrial medium-chain acyl-CoA dehydrogenase deficiency, mitochondrial short-chain acyl-CoA dehydrogenase deficiency, mitochondrial electron transport flavoprotein and electron transport flavoprotein: ubiquinone oxidoreductase deficiency, mitochondrial trifunctional protein deficiency, and mitochondrial short-chain 3-hydroxyacyl-CoA dehydrogenase deficiency; and a lipid metabolism disorder such as fatty liver, cholestasis, primary biliary cirrhosis, carnitine deficiency, carnitine palmitoyltransferase deficiency, myoadenylate deaminase deficiency, hypertriglyceridemia, lipid storage disorders such Fabry's disease, Gaucher's disease, Niemann-Pick's disease, metachromatic leukodystrophy, adrenoleukodystrophy, GM2 gangliosidosis, and ceroid lipofuscinosis, abetalipoproteinemia, Tangier disease, hyperlipoproteinemia, diabetes mellitus, lipodystrophy, lipomatoses, acute panniculitis, disseminated fat necrosis, adiposis dolorosa, lipoid adrenal hyperplasia, minimal change disease, lipomas, atherosclerosis, hypercholesterolemia, hypercholesterolemia with hypertriglyceridemia, primary hypoalphalipoproteinemia, hypothyroidism, renal disease, liver disease, lecithin: cholesterol acyltransferase deficiency, cerebrotendinous xanthomatosis, sitosterolemia, hypocholesterolemia, Tay-Sachs disease, Sandhoff's disease, hyperlipidemia, hyperlipemia, and lipid myopathies; cancer such as adenocarcinoma, leukemia, lymphoma, melanoma, myeloma, sarcoma, teratocarcinoma, and, in particular, cancers of the adrenal gland, bladder, bone, bone marrow, brain, breast, cervix, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; and an infection by a viral agent classified as adenovirus, arenavirus, bunyavirus, calicivirus, coronavirus, filovirus, hepadnavirus, herpesvirus, flavivirus, orthomyxovirus, parvovirus, papovavirus, paramyxovirus, picornavirus, poxvirus, reovirus, retrovirus, rhabdovirus, and tongavirus. The polynucleotide sequences encoding REMAP may be used in Southern or northern analysis, dot blot, or other membrane-based technologies; in PCR technologies; in dipstick, pin, and multiformat ELISA-like assays; and in microarrays utilizing fluids or tissues from patients to detect altered REMAP expression. Such qualitative or quantitative methods are well known in the art.

In a particular aspect, the nucleotide sequences encoding REMAP may be useful in assays that detect the presence of associated disorders, particularly those mentioned above. The nucleotide sequences encoding REMAP may be labeled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridization complexes. After a suitable incubation period, the sample is washed and the signal is quantified and compared with a standard value. If the amount of signal in the patient sample is significantly altered in comparison to a control sample then the presence of altered levels of nucleotide sequences encoding REMAP in the sample indicates the presence of the associated disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or to

monitor the treatment of an individual patient.

In order to provide a basis for the diagnosis of a disorder associated with expression of REMAP, a normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or human, with a sequence, or a fragment thereof, encoding REMAP, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with values from an experiment in which a known amount of a substantially purified polynucleotide is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder.

Once the presence of a disorder is established and a treatment protocol is initiated, hybridization assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in the normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.

With respect to cancer, the presence of an abnormal amount of transcript (either under-or overexpressed) in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.

Additional diagnostic uses for oligonucleotides designed from the sequences encoding REMAP may involve the use of PCR. These oligomers may be chemically synthesized, generated enzymatically, or produced in vitro. Oligomers will preferably contain a fragment of a polynucleotide encoding REMAP, or a fragment of a polynucleotide complementary to the polynucleotide encoding REMAP, and will be employed under optimized conditions for identification of a specific gene or condition. Oligomers may also be employed under less stringent conditions for detection or quantification of closely related DNA or RNA sequences.

In a particular aspect, oligonucleotide primers derived from the polynucleotide sequences encoding REMAP may be used to detect single nucleotide polymorphisms (SNPs). SNPs are substitutions, insertions and deletions that are a frequent cause of inherited or acquired genetic disease in humans. Methods of SNP detection include, but are not limited to, single-stranded conformation polymorphism (SSCP) and fluorescent SSCP (fSSCP) methods. In SSCP, oligonucleotide primers derived from the polynucleotide sequences encoding REMAP are used to amplify DNA using the polymerase chain reaction (PCR). The DNA may be derived, for example,

from diseased or normal tissue, biopsy samples, bodily fluids, and the like. SNPs in the DNA cause differences in the secondary and tertiary structures of PCR products in single-stranded form, and these differences are detectable using gel electrophoresis in non-denaturing gels. In fSCCP, the oligonucleotide primers are fluorescently labeled, which allows detection of the amplimers in high- throughput equipment such as DNA sequencing machines. Additionally, sequence database analysis methods, termed in silico SNP (isSNP), are capable of identifying polymorphisms by comparing the sequence of individual overlapping DNA fragments which assemble into a common consensus sequence. These computer-based methods filter out sequence variations due to laboratory preparation of DNA and sequencing errors using statistical models and automated analyses of DNA sequence chromatograms. In the alternative, SNPs may be detected and characterized by mass spectrometry using, for example, the high throughput MASSARRAY system (Sequenom, Inc., San Diego CA).

SNPs may be used to study the genetic basis of human disease. For example, at least 16 common SNPs have been associated with non-insulin-dependent diabetes mellitus. SNPs are also useful for examining differences in disease outcomes in monogenic disorders, such as cystic fibrosis, sickle cell anemia, or chronic granulomatous disease. For example, variants in the mannose-binding lectin, MBL2, have been shown to be correlated with deleterious pulmonary outcomes in cystic fibrosis. SNPs also have utility in pharmacogenomics, the identification of genetic variants that influence a patient's response to a drug, such as life-threatening toxicity. For example, a variation in N-acetyl transferase is associated with a high incidence of peripheral neuropathy in response to the anti-tuberculosis drug isoniazid, while a variation in the core promoter of the ALOX5 gene results in diminished clinical response to treatment with an anti-asthma drug that targets the 5-lipoxygenase pathway. Analysis of the distribution of SNPs in different populations is useful for investigating genetic drift, mutation, recombination, and selection, as well as for tracing the origins of populations and their migrations. (Taylor, J. G. et al. (2001) Trends Mol. Med. 7: 507-512; Kwok, P.-Y. and Z. Gu (1999) Mol. Med. Today 5: 538-543; Nowotny, P. et al. (2001) Curr. Opin. Neurobiol. 11: 637-641.) Methods which may also be used to quantify the expression of REMAP include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves. (See, e. g., Melby, P. C. et al. (1993) J. Immunol. Methods 159: 235-244; Duplaa, C. et al. (1993) Anal. Biochem. 212: 229-236.) The speed of quantitation of multiple samples may be accelerated by running the assay in a high-throughput format where the oligomer or polynucleotide of interest is presented in various dilutions and a spectrophotometric or colorimetric response gives rapid quantitation.

In further embodiments, oligonucleotides or longer fragments derived from any of the polynucleotide sequences described herein may be used as elements on a microarray. The microarray can be used in transcript imaging techniques which monitor the relative expression levels of large

numbers of genes simultaneously as described below. The microarray may also be used to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic basis of a disorder, to diagnose a disorder, to monitor progression/regression of disease as a function of gene expression, and to develop and monitor the activities of therapeutic agents in the treatment of disease. In particular, this information may be used to develop a pharmacogenomic profile of a patient in order to select the most appropriate and effective treatment regimen for that patient. For example, therapeutic agents which are highly effective and display the fewest side effects may be selected for a patient based on his/her pharmacogenomic profile.

In another embodiment, REMAP, fragments of REMAP, or antibodies specific for REMAP may be used as elements on a microarray. The microarray may be used to monitor or measure protein-protein interactions, drug-target interactions, and gene expression profiles, as described above.

A particular embodiment relates to the use of the polynucleotides of the present invention to generate a transcript image of a tissue or cell type. A transcript image represents the global pattern of gene expression by a particular tissue or cell type. Global gene expression patterns are analyzed by quantifying the number of expressed genes and their relative abundance under given conditions and at a given time. (See Seilhamer et al.,"Comparative Gene Transcript Analysis,"U. S. Patent No.

5,840,484, expressly incorporated by reference herein.) Thus a transcript image may be generated by hybridizing the polynucleotides of the present invention or their complements to the totality of transcripts or reverse transcripts of a particular tissue or cell type. In one embodiment, the hybridization takes place in high-throughput format, wherein the polynucleotides of the present invention or their complements comprise a subset of a plurality of elements on a microarray. The resultant transcript image would provide a profile of gene activity.

Transcript images may be generated using transcripts isolated from tissues, cell lines, biopsies, or other biological samples. The transcript image may thus reflect gene expression in vivo, as in the case of a tissue or biopsy sample, or in vitro, as in the case of a cell line.

Transcript images which profile the expression of the polynucleotides of the present invention may also be used in conjunction with in vitro model systems and preclinical evaluation of pharmaceuticals, as well as toxicological testing of industrial and naturally-occurring environmental compounds. All compounds induce characteristic gene expression patterns, frequently termed molecular fingerprints or toxicant signatures, which are indicative of mechanisms of action and toxicity (Nuwaysir, E. F. et al. (1999) Mol. Carcinog. 24: 153-159; Steiner, S. and N. L. Anderson (2000) Toxicol. Lett. 112-113: 467-471, expressly incorporated by reference herein). If a test compound has a signature similar to that of a compound with known toxicity, it is likely to share

those toxic properties. These fingerprints or signatures are most useful and refined when they contain expression information from a large number of genes and gene families. Ideally, a genome-wide measurement of expression provides the highest quality signature. Even genes whose expression is not altered by any tested compounds are important as well, as the levels of expression of these genes are used to normalize the rest of the expression data. The normalization procedure is useful for comparison of expression data after treatment with different compounds. While the assignment of gene function to elements of a toxicant signature aids in interpretation of toxicity mechanisms, knowledge of gene function is not necessary for the statistical matching of signatures which leads to prediction of toxicity. (See, for example, Press Release 00-02 from the National Institute of Environmental Health Sciences, released February 29,2000, available at http ://www. niehs. nih. gov/oc/news/toxchip. htm.) Therefore, it is important and desirable in toxicological screening using toxicant signatures to include all expressed gene sequences.

In one embodiment, the toxicity of a test compound is assessed by treating a biological sample containing nucleic acids with the test compound. Nucleic acids that are expressed in the treated biological sample are hybridized with one or more probes specific to the polynucleotides of the present invention, so that transcript levels corresponding to the polynucleotides of the present invention may be quantified. The transcript levels in the treated biological sample are compared with levels in an untreated biological sample. Differences in the transcript levels between the two samples are indicative of a toxic response caused by the test compound in the treated sample.

Another particular embodiment relates to the use of the polypeptide sequences of the present invention to analyze the proteome of a tissue or cell type. The term proteome refers to the global pattern of protein expression in a particular tissue or cell type. Each protein component of a proteome can be subjected individually to further analysis. Proteome expression patterns, or profiles, are analyzed by quantifying the number of expressed proteins and their relative abundance under given conditions and at a given time. A profile of a cell's proteome may thus be generated by separating and analyzing the polypeptides of a particular tissue or cell type. In one embodiment, the separation is achieved using two-dimensional gel electrophoresis, in which proteins from a sample are separated by isoelectric focusing in the first dimension, and then according to molecular weight by sodium dodecyl sulfate slab gel electrophoresis in the second dimension (Steiner and Anderson, supra). The proteins are visualized in the gel as discrete and uniquely positioned spots, typically by staining the gel with an agent such as Coomassie Blue or silver or fluorescent stains. The optical density of each protein spot is generally proportional to the level of the protein in the sample. The optical densities of equivalently positioned protein spots from different samples, for example, from biological samples either treated or untreated with a test compound or therapeutic agent, are compared to identify any changes in protein spot density related to the treatment. The proteins in the

spots are partially sequenced using, for example, standard methods employing chemical or enzymatic cleavage followed by mass spectrometry. The identity of the protein in a spot may be determined by comparing its partial sequence, preferably of at least 5 contiguous amino acid residues, to the polypeptide sequences of the present invention. In some cases, further sequence data may be obtained for definitive protein identification.

A proteomic profile may also be generated using antibodies specific for REMAP to quantify the levels of REMAP expression. In one embodiment, the antibodies are used as elements on a microarray, and protein expression levels are quantified by exposing the microarray to the sample and detecting the levels of protein bound to each array element (Lueking, A. et al. (1999) Anal. Biochem.

270: 103-111; Mendoze, L. G. et al. (1999) Biotechniques 27: 778-788). Detection may be performed by a variety of methods known in the art, for example, by reacting the proteins in the sample with a thiol-or amino-reactive fluorescent compound and detecting the amount of fluorescence bound at each array element.

Toxicant signatures at the proteome level are also useful for toxicological screening, and should be analyzed in parallel with toxicant signatures at the transcript level. There is a poor correlation between transcript and protein abundances for some proteins in some tissues (Anderson, N. L. and J. Seilhamer (1997) Electrophoresis 18: 533-537), so proteome toxicant signatures may be useful in the analysis of compounds which do not significantly affect the transcript image, but which alter the proteomic profile. In addition, the analysis of transcripts in body fluids is difficult, due to rapid degradation of mRNA, so proteomic profiling may be more reliable and informative in such cases.

In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins that are expressed in the treated biological sample are separated so that the amount of each protein can be quantified. The amount of each protein is compared to the amount of the corresponding protein in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated sample. Individual proteins are identified by sequencing the amino acid residues of the individual proteins and comparing these partial sequences to the polypeptides of the present invention.

In another embodiment, the toxicity of a test compound is assessed by treating a biological sample containing proteins with the test compound. Proteins from the biological sample are incubated with antibodies specific to the polypeptides of the present invention. The amount of protein recognized by the antibodies is quantified. The amount of protein in the treated biological sample is compared with the amount in an untreated biological sample. A difference in the amount of protein between the two samples is indicative of a toxic response to the test compound in the treated

sample.

Microarrays may be prepared, used, and analyzed using methods known in the art. (See, e. g., Brennan, T. M. et al. (1995) U. S. Patent No. 5,474,796; Schena, M. et al. (1996) Proc. Natl. Acad. Sci.

USA 93: 10614-10619; Baldeschweiler et al. (1995) PCT application W095/251116; Shalon, D. et al.

(1995) PCT application W095/35505 ; Heller, R. A. et al. (1997) Proc. Natl. Acad. Sci. USA 94: 2150- 2155; and Heller, M. J. et al. (1997) U. S. Patent No. 5,605,662.) Various types of microarrays are well known and thoroughly described in DNA Microarrays : A Practical Approach, M. Schena, ed.

(1999) Oxford University Press, London, hereby expressly incorporated by reference.

In another embodiment of the invention, nucleic acid sequences encoding REMAP may be used to generate hybridization probes useful in mapping the naturally occurring genomic sequence.

Either coding or noncoding sequences may be used, and in some instances, noncoding sequences may be preferable over coding sequences. For example, conservation of a coding sequence among members of a multi-gene family may potentially cause undesired cross hybridization during chromosomal mapping. The sequences may be mapped to a particular chromosome, to a specific region of a chromosome, or to artificial chromosome constructions, e. g., human artificial chromosomes (HACs), yeast artificial chromosomes (YACs), bacterial artificial chromosomes (BACs), bacterial P1 constructions, or single chromosome cDNA libraries. (See, e. g., Harrington, J. J. et al. (1997) Nat. Genet. 15: 345-355; Price, C. M. (1993) Blood Rev. 7: 127-134; and Trask, B. J.

(1991) Trends Genet. 7: 149-154.) Once mapped, the nucleic acid sequences of the invention may be used to develop genetic linkage maps, for example, which correlate the inheritance of a disease state with the inheritance of a particular chromosome region or restriction fragment length polymorphism (RFLP). (See, for example, Lander, E. S. and D. Botstein (1986) Proc. Natl. Acad. Sci. USA 83: 7353- 7357.) Fluorescent in situ hybridization (FISH) may be correlated with other physical and genetic map data. (See, e. g., Heinz-Ulrich, et al. (1995) in Meyers, supra, pp. 965-968.) Examples of genetic map data can be found in various scientific journals or at the Online Mendelian Inheritance in Man (OMIM) World Wide Web site. Correlation between the location of the gene encoding REMAP on a physical map and a specific disorder, or a predisposition to a specific disorder, may help define the region of DNA associated with that disorder and thus may further positional cloning efforts.

In situ hybridization of chromosomal preparations and physical mapping techniques, such as linkage analysis using established chromosomal markers, may be used for extending genetic maps.

Often the placement of a gene on the chromosome of another mammalian species, such as mouse, may reveal associated markers even if the exact chromosomal locus is not known. This information is valuable to investigators searching for disease genes using positional cloning or other gene discovery techniques. Once the gene or genes responsible for a disease or syndrome have been crudely

localized by genetic linkage to a particular genomic region, e. g., ataxia-telangiectasia to 1 lq22-23, any sequences mapping to that area may represent associated or regulatory genes for further investigation. (See, e. g., Gatti, R. A. et al. (1988) Nature 336: 577-580.) The nucleotide sequence of the instant invention may also be used to detect differences in the chromosomal location due to translocation, inversion, etc., among normal, carrier, or affected individuals.

In another embodiment of the invention, REMAP, its catalytic or immunogenic fragments, or oligopeptides thereof can be used for screening libraries of compounds in any of a variety of drug screening techniques. The fragment employed in such screening may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The formation of binding complexes between REMAP and the agent being tested may be measured.

Another technique for drug screening provides for high throughput screening of compounds having suitable binding affinity to the protein of interest. (See, e. g., Geysen, et al. (1984) PCT application W084/03564.) In this method, large numbers of different small test compounds are synthesized on a solid substrate. The test compounds are reacted with REMAP, or fragments thereof, and washed. Bound REMAP is then detected by methods well known in the art. Purified REMAP can also be coated directly onto plates for use in the aforementioned drug screening techniques.

Alternatively, non-neutralizing antibodies can be used to capture the peptide and immobilize it on a solid support.

In another embodiment, one may use competitive drug screening assays in which neutralizing antibodies capable of binding REMAP specifically compete with a test compound for binding REMAP. In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with REMAP.

In additional embodiments, the nucleotide sequences which encode REMAP may be used in any molecular biology techniques that have yet to be developed, provided the new techniques rely on properties of nucleotide sequences that are currently known, including, but not limited to, such properties as the triplet genetic code and specific base pair interactions.

Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.

The disclosures of all patents, applications and publications, mentioned above and below, including U. S. Ser. No. 60/292,197, U. S. Ser. No. 60/297,012, U. S. Ser. No. 60/300,495, U. S. Ser.

No. 60/300,582, U. S. Ser. No. 60/301,992, and U. S. Ser. No. 60/340, 542, are expressly incorporated by reference herein.

EXAMPLES I. Construction of cDNA Libraries Incyte cDNAs were derived from cDNA libraries described in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA). Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL (Invitrogen), a monophasic solution of phenol and guanidine isothiocyanate. The resulting lysates were centrifuged over CsCI cushions or extracted with chloroform. RNA was precipitated from the lysates with either isopropanol or sodium acetate and ethanol, or by other routine methods.

Phenol extraction and precipitation of RNA were repeated as necessary to increase RNA purity. In some cases, RNA was treated with DNase. For most libraries, poly (A) + RNA was isolated using oligo d (T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA purification kit (QIAGEN). Alternatively, RNA was isolated directly from tissue lysates using other RNA isolation kits, e. g., the POLY (A) PURE mRNA purification kit (Ambion, Austin TX).

In some cases, Stratagene was provided with RNA and constructed the corresponding cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen), using the recommended procedures or similar methods known in the art. (See, e. g., Ausubel, 1997, supra, units 5.1-6.6.) Reverse transcription was initiated using oligo d (T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Biosciences) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e. g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Invitrogen), PCDNA2.1 plasmid (Invitrogen, Carlsbad CA), PBK-CMV plasmid (Stratagene), PCR2-TOPOTA plasmid (Invitrogen), PCMV-ICIS plasmid (Stratagene), pIGEN (Incyte Genomics, Palo Alto CA), pRARE (Incyte Genomics), or pINCY (Incyte Genomics), or derivatives thereof. Recombinant plasmids were transformed into competent E. coli cells including XLl-Blue, XLl-BlueMRF, or SOLR from Stratagene or DHSa, DH10B, or ElectroMAX DH10B from Invitrogen.

II. Isolation of cDNA Clones Plasmids obtained as described in Example I were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using at least one of the following: a Magic or WIZARD Minipreps DNA purification system (Promega); an

AGTC Miniprep purification kit (Edge Biosystems, Gaithersburg MD); and QIAWELL 8 Plasmid, QIAWELL 8 Plus Plasmid, QIAWELL 8 Ultra Plasmid purification systems or the R. E. A. L. PREP 96 plasmid purification kit from QIAGEN. Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4°C.

Alternatively, plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao, V. B. (1994) Anal. Biochem. 216: 1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN I fluorescence scanner (Labsystems Oy, Helsinki, Finland).

III. Sequencing and Analysis Incyte cDNA recovered in plasmids as described in Example II were sequenced as follows.

Sequencing reactions were processed using standard methods or high-throughput instrumentation such as the ABI CATALYST 800 (Applied Biosystems) thermal cycler or the PTC-200 thermal cycler (MJ Research) in conjunction with the HYDRA microdispenser (Robbins Scientific) or the MICROLAB 2200 (Hamilton) liquid transfer system. cDNA sequencing reactions were prepared using reagents provided by Amersham Biosciences or supplied in ABI sequencing kits such as the ABI PRISM BIGDYE Terminator cycle sequencing ready reaction kit (Applied Biosystems).

Electrophoretic separation of cDNA sequencing reactions and detection of labeled polynucleotides were carried out using the MEGABACE 1000 DNA sequencing system (Amersham Biosciences); the ABI PRISM 373 or 377 sequencing system (Applied Biosystems) in conjunction with standard ABI protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods (reviewed in Ausubel, 1997, supra, unit 7.7). Some of the cDNA sequences were selected for extension using the techniques disclosed in Example VIII.

The polynucleotide sequences derived from Incyte cDNAs were validated by removing vector, linker, and poly (A) sequences and by masking ambiguous bases, using algorithms and programs based on BLAST, dynamic programming, and dinucleotide nearest neighbor analysis. The Incyte cDNA sequences or translations thereof were then queried against a selection of public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases, and BLOCKS, PRINTS, DOMO, PRODOM; PROTEOME databases with sequences from Homo sapiens, Rattus norvegicus, Mus musculus, Caenorhabditis elegans, Saccharomyces cerevisiae, Schizosaccharomvces pombe, and Candida albicans (Incyte Genomics, Palo Alto CA); hidden Markov model (HMM)-based protein family databases such as PFAM, INCY, and TIGRFAM (Haft, D. H. et al. (2001) Nucleic Acids Res. 29: 41-43); and HMM-based protein domain databases such as

SMART (Schultz et al. (1998) Proc. Natl. Acad. Sci. USA 95: 5857-5864; Letunic, I. et al. (2002) Nucleic Acids Res. 30: 242-244). (HMM is a probabilistic approach which analyzes consensus primary structures of gene families. See, for example, Eddy, S. R. (1996) Curr. Opin. Struct. Biol.

6: 361-365.) The queries were performed using programs based on BLAST, FASTA, BLIMPS, and HMMER. The Incyte cDNA sequences were assembled to produce full length polynucleotide sequences. Alternatively, GenBank cDNAs, GenBank ESTs, stitched sequences, stretched sequences, or Genscan-predicted coding sequences (see Examples IV and V) were used to extend Incyte cDNA assemblages to full length. Assembly was performed using programs based on Phred, Phrap, and Consed, and cDNA assemblages were screened for open reading frames using programs based on GeneMark, BLAST, and FASTA. The full length polynucleotide sequences were translated to derive the corresponding full length polypeptide sequences. Alternatively, a polypeptide of the invention may begin at any of the methionine residues of the full length translated polypeptide. Full length polypeptide sequences were subsequently analyzed by querying against databases such as the GenBank protein databases (genpept), SwissProt, the PROTEOME databases, BLOCKS, PRINTS, DOMO, PRODOM, Prosite, hidden Markov model (HMM)-based protein family databases such as PFAM, INCY, and TIGRFAM; and HMM-based protein domain databases such as SMART. Full length polynucleotide sequences are also analyzed using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA) and LASERGENE software (DNASTAR).

Polynucleotide and polypeptide sequence alignments are generated using default parameters specified by the CLUSTAL algorithm as incorporated into the MEGALIGN multisequence alignment program (DNASTAR), which also calculates the percent identity between aligned sequences.

Table 7 summarizes the tools, programs, and algorithms used for the analysis and assembly of Incyte cDNA and full length sequences and provides applicable descriptions, references, and threshold parameters. The first column of Table 7 shows the tools, programs, and algorithms used, the second column provides brief descriptions thereof, the third column presents appropriate references, all of which are incorporated by reference herein in their entirety, and the fourth column presents, where applicable, the scores, probability values, and other parameters used to evaluate the strength of a match between two sequences (the higher the score or the lower the probability value, the greater the identity between two sequences).

The programs described above for the assembly and analysis of full length polynucleotide and polypeptide sequences were also used to identify polynucleotide sequence fragments from SEQ ID NO : 27-52. Fragments from about 20 to about 4000 nucleotides which are useful in hybridization and amplification technologies are described in Table 4, column 2.

IV. Identification and Editing of Coding Sequences from Genomic DNA Putative receptors and membrane-associated proteins were initially identified by running the

Genscan gene identification program against public genomic sequence databases (e. g., gbpri and gbhtg). Genscan is a general-purpose gene identification program which analyzes genomic DNA sequences from a variety of organisms (See Burge, C. and S. Karlin (1997) J. Mol. Biol. 268: 78-94, and Burge, C. and S. Karlin (1998) Curr. Opin. Struct. Biol. 8: 346-354). The program concatenates predicted exons to form an assembled cDNA sequence extending from a methionine to a stop codon.

The output of Genscan is a FASTA database of polynucleotide and polypeptide sequences. The maximum range of sequence for Genscan to analyze at once was set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode receptors and membrane-associated proteins, the encoded polypeptides were analyzed by querying against PFAM models for receptors and membrane- associated proteins. Potential receptors and membrane-associated proteins were also identified by homology to Incyte cDNA sequences that had been annotated as receptors and membrane-associated proteins. These selected Genscan-predicted sequences were then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences were then edited by comparison to the top BLAST hit from genpept to correct errors in the sequence predicted by Genscan, such as extra or omitted exons. BLAST analysis was also used to find any Incyte cDNA or public cDNA coverage of the Genscan-predicted sequences, thus providing evidence for transcription. When Incyte cDNA coverage was available, this information was used to correct or confirm the Genscan predicted sequence. Full length polynucleotide sequences were obtained by assembling Genscan-predicted coding sequences with Incyte cDNA sequences and/or public cDNA sequences using the assembly process described in Example III. Alternatively, full length polynucleotide sequences were derived entirely from edited or unedited Genscan-predicted coding sequences.

V. Assembly of Genomic Sequence Data with cDNA Sequence Data "Stitched"Sequences Partial cDNA sequences were extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example III were mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster was analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that were subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval was present on more than one sequence in the cluster were identified, and intervals thus identified were considered to be equivalent by transitivity. For example, if an interval was present on a cDNA and two genomic sequences, then all three intervals were considered to be equivalent. This process allows unrelated but consecutive genomic sequences to be brought together, bridged by cDNA sequence. Intervals

thus identified were then"stitched"together by the stitching algorithm in the order that they appear along their parent sequences to generate the longest possible sequence, as well as sequence variants.

Linkages between intervals which proceed along one type of parent sequence (cDNA to cDNA or genomic sequence to genomic sequence) were given preference over linkages which change parent type (cDNA to genomic sequence). The resultant stitched sequences were translated and compared by BLAST analysis to the genpept and gbpri public databases. Incorrect exons predicted by Genscan were corrected by comparison to the top BLAST hit from genpept. Sequences were further extended with additional cDNA sequences, or by inspection of genomic DNA, when necessary.

"Stretched"Sequences Partial DNA sequences were extended to full length with an algorithm based on BLAST analysis. First, partial cDNAs assembled as described in Example III were queried against public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases using the BLAST program. The nearest GenBank protein homolog was then compared by BLAST analysis to either Incyte cDNA sequences or GenScan exon predicted sequences described in Example IV. A chimeric protein was generated by using the resultant high-scoring segment pairs (HSPs) to map the translated sequences onto the GenBank protein homolog. Insertions or deletions may occur in the chimeric protein with respect to the original GenBank protein homolog. The GenBank protein homolog, the chimeric protein, or both were used as probes to search for homologous genomic sequences from the public human genome databases. Partial DNA sequences were therefore"stretched"or extended by the addition of homologous genomic sequences. The resultant stretched sequences were examined to determine whether it contained a complete gene.

VI. Chromosomal Mapping of REMAP Encoding Polynucleotides The sequences which were used to assemble SEQ ID NO : 27-52 were compared with sequences from the Incyte LIFESEQ database and public domain databases using BLAST and other implementations of the Smith-Waterman algorithm. Sequences from these databases that matched SEQ ID NO : 27-52 were assembled into clusters of contiguous and overlapping sequences using assembly algorithms such as Phrap (Table 7). Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Généthon were used to determine if any of the clustered sequences had been previously mapped. Inclusion of a mapped sequence in a cluster resulted in the assignment of all sequences of that cluster, including its particular SEQ ID NO:, to that map location.

Map locations are represented by ranges, or intervals, of human chromosomes. The map position of an interval, in centiMorgans, is measured relative to the terminus of the chromosome's p- arm. (The centiMorgan (cM) is a unit of measurement based on recombination frequencies between chromosomal markers. On average, 1 cM is roughly equivalent to 1 megabase (Mb) of DNA in

humans, although this can vary widely due to hot and cold spots of recombination.) The cM distances are based on genetic markers mapped by Généthon which provide boundaries for radiation hybrid markers whose sequences were included in each of the clusters. Human genome maps and other resources available to the public, such as the NCBI"GeneMap'99"World Wide Web site (http://www. ncbi. nlm. nih. gov/genemap/), can be employed to determine if previously identified disease genes map within or in proximity to the intervals indicated above.

VII. Analysis of Polynucleotide Expression Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound. (See, e. g., Sambrook, supra, ch. 7; Ausubel (1995) supra, ch. 4 and 16.) Analogous computer techniques applying BLAST were used to search for identical or related molecules in cDNA databases such as GenBank or LIFESEQ (Incyte Genomics). This analysis is much faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or similar. The basis of the search is the product score, which is defined as: BLAST Score x Percent Identity 5 x minimum {length (Seq. 1), length (Seq. 2)} The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. The product score is a normalized value between 0 and 100, and is calculated as follows: the BLAST score is multiplied by the percent nucleotide identity and the product is divided by (5 times the length of the shorter of the two sequences). The BLAST score is calculated by assigning a score of +5 for every base that matches in a high-scoring segment pair (HSP), and-4 for every mismatch. Two sequences may share more than one HSP (separated by gaps). If there is more than one HSP, then the pair with the highest BLAST score is used to calculate the product score. The product score represents a balance between fractional overlap and quality in a BLAST alignment. For example, a product score of 100 is produced only for 100% identity over the entire length of the shorter of the two sequences being compared. A product score of 70 is produced either by 100% identity and 70% overlap at one end, or by 88% identity and 100% overlap at the other. A product score of 50 is produced either by 100% identity and 50% overlap at one end, or 79% identity and 100% overlap.

Alternatively, polynucleotide sequences encoding REMAP are analyzed with respect to the tissue sources from which they were derived. For example, some full length sequences are

assembled, at least in part, with overlapping Incyte cDNA sequences (see Example HI). Each cDNA sequence is derived from a cDNA library constructed from a human tissue. Each human tissue is classified into one of the following organ/tissue categories: cardiovascular system; connective tissue; digestive system; embryonic structures; endocrine system; exocrine glands; genitalia, female; genitalia, male; germ cells; hemic and immune system; liver; musculoskeletal system; nervous system; pancreas; respiratory system; sense organs; skin; stomatognathic system; unclassified/mixed ; or urinary tract. The number of libraries in each category is counted and divided by the total number of libraries across all categories. Similarly, each human tissue is classified into one of the following disease/condition categories: cancer, cell line, developmental, inflammation, neurological, trauma, cardiovascular, pooled, and other, and the number of libraries in each category is counted and divided by the total number of libraries across all categories. The resulting percentages reflect the tissue-and disease-specific expression of cDNA encoding REMAP. cDNA sequences and cDNA library/tissue information are found in the LIFESEQ GOLD database (Incyte Genomics, Palo Alto CA).

VIII. Extension of REMAP Encoding Polynucleotides Full length polynucleotide sequences were also produced by extension of an appropriate fragment of the full length molecule using oligonucleotide primers designed from this fragment. One primer was synthesized to initiate 5'extension of the known fragment, and the other primer was synthesized to initiate 3'extension of the known fragment. The initial primers were designed using OLIGO 4.06 software (National Biosciences), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68°C to about 72°C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.

Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed.

High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the PTC-200 thermal cycler (MJ Research, Inc.). The reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg2+, (NH4) 2SO4, and 2-mercaptoethanol, Taq DNA polymerase (Amersham Biosciences), ELONGASE enzyme (Invitrogen), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min ; Step 4: 68°C, 2 min ; Step 5: Steps 2,3, and 4 repeated 20 times; Step 6: 68°C, 5 min ; Step 7: storage at 4°C. In the alternative, the parameters for primer pair T7 and SK+ were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2,3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7 : storage at 4°C.

The concentration of DNA in each well was determined by dispensing 100 nul PICOGREEN

quantitation reagent (0.25% (v/v) PICOGREEN; Molecular Probes, Eugene OR) dissolved in IX TE and 0.5 nul of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton MA), allowing the DNA to bind to the reagent. The plate was scanned in a Pluoroskan II (Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5, ul to 10, ul aliquot of the reaction mixture was analyzed by electrophoresis on a 1 % agarose gel to determine which reactions were successful in extending the sequence.

The extended nucleotides were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC 18 vector (Amersham Biosciences). For shotgun sequencing, the digested nucleotides were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with Agar ACE (Promega). Extended clones were religated using T4 ligase (New England Biolabs, Beverly MA) into pUC 18 vector (Amersham Biosciences), treated with Pfu DNA polymerase (Stratagene) to fill-in restriction site overhangs, and transfected into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and individual colonies were picked and cultured overnight at 37°C in 384-well plates in LB/2x carb liquid media.

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

In like manner, full length polynucleotide sequences are verified using the above procedure or are used to obtain 5'regulatory sequences using the above procedure along with oligonucleotides designed for such extension, and an appropriate genomic library.

IX. Identification of Single Nucleotide Polymorphisms in REMAP Encoding Polynucleotides Common DNA sequence variants known as single nucleotide polymorphisms (SNPs) were identified in SEQ ID NO : 27-52 using the LIFESEQ database (Incyte Genomics). Sequences from the same gene were clustered together and assembled as described in Example III, allowing the identification of all sequence variants in the gene. An algorithm consisting of a series of filters was

used to distinguish SNPs from other sequence variants. Preliminary filters removed the majority of basecall errors by requiring a minimum Phred quality score of 15, and removed sequence alignment errors and errors resulting from improper trimming of vector sequences, chimeras, and splice variants. An automated procedure of advanced chromosome analysis analysed the original chromatogram files in the vicinity of the putative SNP. Clone error filters used statistically generated algorithms to identify errors introduced during laboratory processing, such as those caused by reverse transcriptase, polymerase, or somatic mutation. Clustering error filters used statistically generated algorithms to identify errors resulting from clustering of close homologs or pseudogenes, or due to contamination by non-human sequences. A final set of filters removed duplicates and SNPs found in immunoglobulins or T-cell receptors.

Certain SNPs were selected for further characterization by mass spectrometry using the high throughput MASSARRAY system (Sequenom, Inc.) to analyze allele frequencies at the SNP sites in four different human populations. The Caucasian population comprised 92 individuals (46 male, 46 female), including 83 from Utah, four French, three Venezualan, and two Amish individuals. The African population comprised 194 individuals (97 male, 97 female), all African Americans. The Hispanic population comprised 324 individuals (162 male, 162 female), all Mexican Hispanic. The Asian population comprised 126 individuals (64 male, 62 female) with a reported parental breakdown of 43% Chinese, 31% Japanese, 13% Korean, 5% Vietnamese, and 8% other Asian. Allele frequencies were first analyzed in the Caucasian population; in some cases those SNPs which showed no allelic variance in this population were not further tested in the other three populations.

X. Labeling and Use of Individual Hybridization Probes Hybridization probes derived from SEQ ID NO : 27-52 are employed to screen cDNAs, genomic DNAs, or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base pairs, is specifically described, essentially the same procedure is used with larger nucleotide fragments. Oligonucleotides are designed using state-of-the-art software such as OLIGO 4.06 software (National Biosciences) and labeled by combining 50 pmol of each oligomer, 250, uCi of [Y-32P] adenosine triphosphate (Amersham Biosciences), and T4 polynucleotide kinase (DuPont NEN, Boston MA). The labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Biosciences). An aliquot containing 101 counts per minute of the labeled probe is used in a typical membrane-based hybridization analysis of human genomic DNA digested with one of the following endonucleases: Ase I, Bgl ici, Eco RI, Pst I, Xba I, or Pvu IN (DuPont NEN).

The DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham NH). Hybridization is carried out for 16 hours at 40°C. To remove nonspecific signals, blots are sequentially washed at room temperature

under conditions of up to, for example, 0.1 x saline sodium citrate and 0.5% sodium dodecyl sulfate.

Hybridization patterns are visualized using autoradiography or an alternative imaging means and compared.

XI. Microarrays The linkage or synthesis of array elements upon a microarray can be achieved utilizing photolithography, piezoelectric printing (ink-jet printing, See, e. g., Baldeschweiler, supra.), mechanical microspotting technologies, and derivatives thereof. The substrate in each of the aforementioned technologies should be uniform and solid with a non-porous surface (Schena (1999), supra). Suggested substrates include silicon, silica, glass slides, glass chips, and silicon wafers.

Alternatively, a procedure analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures. A typical array may be produced using available methods and machines well known to those of ordinary skill in the art and may contain any appropriate number of elements. (See, e. g., Schena, M. et al. (1995) Science 270: 467-470 ; Shalon, D. et al. (1996) Genome Res. 6: 639-645; Marshall, A. and J. Hodgson (1998) Nat. Biotechnol. 16: 27-31.) Full length cDNAs, Expressed Sequence Tags (ESTs), or fragments or oligomers thereof may comprise the elements of the microarray. Fragments or oligomers suitable for hybridization can be selected using software well known in the art such as LASERGENE software (DNASTAR). The array elements are hybridized with polynucleotides in a biological sample. The polynucleotides in the biological sample are conjugated to a fluorescent label or other molecular tag for ease of detection.

After hybridization, nonhybridized nucleotides from the biological sample are removed, and a fluorescence scanner is used to detect hybridization at each array element. Alternatively, laser desorbtion and mass spectrometry may be used for detection of hybridization. The degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed. In one embodiment, microarray preparation and usage is described in detail below.

Tissue or Cell Sample Preparation Total RNA is isolated from tissue samples using the guanidinium thiocyanate method and poly (A) + RNA is purified using the oligo- (dT) cellulose method. Each poly (A) + RNA sample is reverse transcribed using MMLV reverse-transcriptase, 0.05 pg/, ul oligo-(dT) primer (21mer), 1X first strand buffer, 0.03 units/yl RNase inhibitor, 500 yM dATP, 500, uM dGTP, 500, uM dTTP, 40 juM dCTP, 40 juM dCTP-Cy3 (BDS) or dCTP-Cy5 (Amersham Biosciences). The reverse transcription reaction is performed in a 25 ml volume containing 200 ng poly (A) RNA with GEMBRIGHT kits (Incyte). Specific control poly (A) + RNAs are synthesized by in vitro transcription from non-coding yeast genomic DNA. After incubation at 37° C for 2 hr, each reaction sample (one

with Cy3 and another with Cy5 labeling) is treated with 2.5 ml of 0. 5M sodium hydroxide and incubated for 20 minutes at 850 C to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (CLONTECH Laboratories, Inc.

(CLONTECH), Palo Alto CA) and after combining, both reaction samples are ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The sample is then dried to completion using a SpeedVAC (Savant Instruments Inc., Holbrook NY) and resuspended in 14 y1 5X SSC/0.2% SDS.

Microarrav Preparation Sequences of the present invention are used to generate array elements. Each array element is amplified from bacterial cells containing vectors with cloned cDNA inserts. PCR amplification uses primers complementary to the vector sequences flanking the cDNA insert. Array elements are amplified in thirty cycles of PCR from an initial quantity of 1-2 ng to a final quantity greater than 5 g. Amplified array elements are then purified using SEPHACRYL-400 (Amersham Biosciences).

Purified array elements are immobilized on polymer-coated glass slides. Glass microscope slides (Corning) are cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments. Glass slides are etched in 4% hydrofluoric acid (VWR Scientific Products Corporation (VWR), West Chester PA), washed extensively in distilled water, and coated with 0.05% aminopropyl silane (Sigma) in 95% ethanol. Coated slides are cured in a 110°C oven.

Array elements are applied to the coated glass substrate using a procedure described in U. S.

Patent No. 5,807,522, incorporated herein by reference. 1/. tl of the array element DNA, at an average concentration of 100 ng/ttl, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 nl of array element sample per slide.

Microarrays are W-crosslinked using a STRATALINKER UV-crosslinker (Stratagene).

Microarrays are washed at room temperature once in 0.2% SDS and three times in distilled water.

Non-specific binding sites are blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (PBS) (Tropix, Inc., Bedford MA) for 30 minutes at 60° C followed by washes in 0.2% SDS and distilled water as before.

Hybridization Hybridization reactions contain 9 y1 of sample mixture consisting of 0.2 jUg each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer. The sample mixture is heated to 65° C for 5 minutes and is aliquoted onto the microarray surface and covered with an 1.8 cm2 coverslip. The arrays are transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber is kept at 100% humidity internally by the addition of 140 jul of 5X SSC in a corner of the chamber. The chamber containing the arrays is

incubated for about 6.5 hours at 60° C. The arrays are washed for 10 min at 45 ° C in a first wash buffer (1X SSC, 0.1% SDS), three times for 10 minutes each at 45°C in a second wash buffer (0. 1X SSC), and dried.

Detection Reporter-labeled hybridization complexes are detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Inc., Santa Clara CA) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of Cy5. The excitation laser light is focused on the array using a 20X microscope objective (Nikon, Inc., Melville NY). The slide containing the array is placed on a computer-controlled X-Y stage on the microscope and raster- scanned past the objective. The 1.8 cm x 1.8 cm array used in the present example is scanned with a resolution of 20 micrometers.

In two separate scans, a mixed gas multiline laser excites the two fluorophores sequentially.

Emitted light is split, based on wavelength, into two photomultiplier tube detectors (PMT R1477, Hamamatsu Photonics Systems, Bridgewater NJ) corresponding to the two fluorophores. Appropriate filters positioned between the array and the photomultiplier tubes are used to filter the signals. The emission maxima of the fluorophores used are 565 nm for Cy3 and 650 nm for Cy5. Each array is typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus is capable of recording the spectra from both fluorophores simultaneously.

The sensitivity of the scans is typically calibrated using the signal intensity generated by a cDNA control species added to the sample mixture at a known concentration. A specific location on the array contains a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1: 100,000. When two samples from different sources (e. g., representing test and control cells), each labeled with a different fluorophore, are hybridized to a single array for the purpose of identifying genes that are differentially expressed, the calibration is done by labeling samples of the calibrating cDNA with the two fluorophores and adding identical amounts of each to the hybridization mixture.

The output of the photomultiplier tube is digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Inc., Norwood MA) installed in an IBM-compatible PC computer. The digitized data are displayed as an image where the signal intensity is mapped using a linear 20-color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal). The data is also analyzed quantitatively. Where two different fluorophores are excited and measured simultaneously, the data are first corrected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using each fluorophore's emission spectrum.

A grid is superimposed over the fluorescence signal image such that the signal from each spot is centered in each element of the grid. The fluorescence signal within each element is then

integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis is the GEMTOOLS gene expression analysis program (Incyte).

Expression SEQ ID NO : 30, and SEQ ID NO : 48-50 showed differential expression, as determined by microarray analysis. Array elements that exhibited at least about a two-fold change in expression, a signal-to-background ratio of at least 2.5, and an element spot size of at least 40% were identified as differentially expressed using the GEMTOOLS program (Incyte Genomics).

For example, to evaluate the variation in gene expression in the peripheral blood mononuclear cells (PBMCs) (12% B lymphocytes, 40% T lymphocytes, 20% NK cells, 25% monocytes, and 3% various cells that include dendritic and progenitor cells) from healthy donors in response to Staphylococcal enterotoxin B (SEB), the PBMCs from 7 healthy volunteer donors were stimulated in vitro with SEB for 24 and 72 hours. The SEB treated PBMCs from each donor were compared to PBMCs from the same donor, kept in culture for 24 hours in the absence of SEB. The expression of SEQ ID NO : 30 was increased by at least two fold in PBMCs treated with SEB.

Therefore SEQ ID NO : 30 is useful in diagnostic assays for inflammatory responses.

In another example, the degree of complementarity and the relative abundance of each polynucleotide which hybridizes to an element on the microarray may be assessed. In one embodiment, microarray preparation and usage is described in detail below. For example, SEQ ID NO : 48 showed differential expression in breast tumor cell lines which were harvested from donors with ductal carcinoma. Normal human mammary epithelial cells (HMEC) and breast ductal carcinoma cells (BT-474 and BT-483) were grown in basal media in the absence of growth factors and hormones for 24 hours prior to comparison. Cells were lysed in Trizol and the total RNA fraction was recovered according to manufacturer protocols. Poly (A) mRNA was purified using a standard oligo-dT selection method. Cy3 and Cy5 probes were prepared according to the standard operating procedure developed at Incyte's microarray facility. The gene expression profile of HMEC cells was compared to that of BT-474 and BT-483. The expression of SEQ ID NO : 48 was increased by at least two-fold in breast tumor cell lines (BT-474 and BT-483) versus normal breast epithelial cells (HMEC) as determined by microarray analysis. Therefore, SEQ ID NO : 48 is useful in diagnostic assays for detection of breast cancer.

In a further example, microarray analysis was used to compare the expression of SEQ ID NO : 49, encoding the polypeptide of SEQ ID NO : 23, in untreated primary blood mononuclear cells (PBMC) versus PBMC treated with the glucocorticoid, dexamethasone, (1 yM), lipopolysaccharide (LPS) (1 yg/rnl), or both compounds. Pooled lymphocytes from four donors were treated (or mock- treated) for 2,4,24, or 72 hours. PBMC treated with either dexamethasone, LPS, or both compounds showed greater than two-fold down-regulation of SEQ ID NO : 49 during the early time points of the

experiment (i. e., 2 and 4 hours) and a less pronounced down-regulation at later time points. These results demonstrate that SEQ ID NO : 49 expression levels can be used to monitor the early effects of compounds that are toxic to lymphoctes (e. g., LPS) or that mediate inflammatory response by affecting PBMC response (e. g., dexamethasone).

In a related experiment, a B-cell lymphoblast cell line (RPMI 6666) derived from cells obtained from a Hodgkin's disease patient were untreated or treated with 1, ug/ml LPS for 0.5,1,2,4, or 8 hours. The expression levels of SEQ ID NO : 49 decreased steadily throughout the course of the experiment and were decreased greater than two-fold at the 4 and 8-hour time points. In the same experiment, the expression of SEQ ID NO : 50, which encodes the polypeptide of SEQ ID NO : 24, was reduced greater than 20-fold at the 8-hour time point while showing no difference compared to untreated cells at the earlier time points. These results suggest that SEQ ID NO : 49 and SEQ ID NO : 50 are useful for monitoring the effects of toxic compounds such as LPS on B-cell lines.

XII. Complementary Polynucleotides Sequences complementary to the REMAP-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring REMAP. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using OLIGO 4.06 software (National Biosciences) and the coding sequence of REMAP.

To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5' sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to the REMAP-encoding transcript.

XIII. Expression of REMAP Expression and purification of REMAP is achieved using bacterial or virus-based expression systems. For expression of REMAP in bacteria, cDNA is subcloned into an appropriate vector containing an antibiotic resistance gene and an inducible promoter that directs high levels of cDNA transcription. Examples of such promoters include, but are not limited to, the trp-lac (tac) hybrid promoter and the T5 or T7 bacteriophage promoter in conjunction with the lac operator regulatory element. Recombinant vectors are transformed into suitable bacterial hosts, e. g., BL21 (DE3).

Antibiotic resistant bacteria express REMAP upon induction with isopropyl beta-D- thiogalactopyranoside (1PTG). Expression of REMAP in eukaryotic cells is achieved by infecting insect or mammalian cell lines with recombinant Autographica californica nuclear polyhedrosis virus (AcMNPV), commonly known as baculovirus. The nonessential polyhedrin gene of baculovirus is replaced with cDNA encoding REMAP by either homologous recombination or bacterial-mediated transposition involving transfer plasmid intermediates. Viral infectivity is maintained and the strong

polyhedrin promoter drives high levels of cDNA transcription. Recombinant baculovirus is used to infect Spodoptera frugiperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases.

Infection of the latter requires additional genetic modifications to baculovirus. (See Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. USA 91: 3224-3227; Sandig, V. et al. (1996) Hum. Gene Ther.

7: 1937-1945.) In most expression systems, REMAP is synthesized as a fusion protein with, e. g., glutathione S-transferase (GST) or a peptide epitope tag, such as FLAG or 6-His, permitting rapid, single-step, affinity-based purification of recombinant fusion protein from crude cell lysates. GST, a 26- kilodalton enzyme from Schistosoma japonicum, enables the purification of fusion proteins on immobilized glutathione under conditions that maintain protein activity and antigenicity (Amersham Biosciences). Following purification, the GST moiety can be proteolytically cleaved from REMAP at specifically engineered sites. FLAG, an 8-amino acid peptide, enables immunoaffinity purification using commercially available monoclonal and polyclonal anti-FLAG antibodies (Eastman Kodak). 6- His, a stretch of six consecutive histidine residues, enables purification on metal-chelate resins (QIAGEN). Methods for protein expression and purification are discussed in Ausubel (1995, supra, ch. 10 and 16). Purified REMAP obtained by these methods can be used directly in the assays shown in Examples XVII, XVIII, and XIX, where applicable.

XIV. Functional Assays REMAP function is assessed by expressing the sequences encoding REMAP at physiologically elevated levels in mammalian cell culture systems. cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA expression. Vectors of choice include PCMV SPORT plasmid (Invitrogen, Carlsbad CA) and PCR3.1 plasmid (Invitrogen), both of which contain the cytomegalovirus promoter. 5-10 tg of recombinant vector are transiently transfected into a human cell line, for example, an endothelial or hematopoietic cell line, using either liposome formulations or electroporation. 1-2, ug of an additional plasmid containing sequences encoding a marker protein are co-transfected. Expression of a marker protein provides a means to distinguish transfected cells from nontransfected cells and is a reliable predictor of cDNA expression from the recombinant vector. Marker proteins of choice include, e. g., Green Fluorescent Protein (GFP; Clontech), CD64, or a CD64-GFP fusion protein.

Flow cytometry (FCM), an automated, laser optics-based technique, is used to identify transfected cells expressing GFP or CD64-GFP and to evaluate the apoptotic state of the cells and other cellular properties. FCM detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell death. These events include changes in nuclear DNA content as measured by staining of DNA with propidium iodide; changes in cell size and granularity as measured by forward light scatter and 90 degree side light scatter; down-regulation of DNA synthesis as

measured by decrease in bromodeoxyuridine uptake; alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell surface. Methods in flow cytometry are discussed in Ormerod, M. G. (1994) Flow Cytometrx, Oxford, New York NY.

The influence of REMAP on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding REMAP and either CD64 or CD64-GFP.

CD64 and CD64-GFP are expressed on the surface of transfected cells and bind to conserved regions of human immunoglobulin G (IgG). Transfected cells are efficiently separated from nontransfected cells using magnetic beads coated with either human IgG or antibody against CD64 (DYNAL, Lake Success NY). mRNA can be purified from the cells using methods well known by those of skill in the art. Expression of mRNA encoding REMAP and other genes of interest can be analyzed by northern analysis or microarray techniques.

XV. Production of REMAP Specific Antibodies REMAP substantially purified using polyacrylamide gel electrophoresis (PAGE; see, e. g., Harrington, M. G. (1990) Methods Enzymol. 182: 488-495), or other purification techniques, is used to immunize animals (e. g., rabbits, mice, etc.) and to produce antibodies using standard protocols.

Alternatively, the REMAP amino acid sequence is analyzed using LASERGENE software (DNASTAR) to determine regions of high immunogenicity, and a corresponding oligopeptide is synthesized and used to raise antibodies by means known to those of skill in the art. Methods for selection of appropriate epitopes, such as those near the C-terminus or in hydrophilic regions are well described in the art. (See, e. g., Ausubel, 1995, supra, ch. 11.) Typically, oligopeptides of about 15 residues in length are synthesized using an ABI 431A peptide synthesizer (Applied Biosystems) using FMOC chemistry and coupled to KLH (Sigma- Aldrich, St. Louis MO) by reaction with N-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS) to increase immunogenicity. (See, e. g., Ausubel, 1995, supra.) Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide and anti-REMAP activity by, for example, binding the peptide or REMAP to a substrate, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radio-iodinated goat anti-rabbit IgG.

XVI. Purification of Naturally Occurring REMAP Using Specific Antibodies Naturally occurring or recombinant REMAP is substantially purified by immunoaffinity chromatography using antibodies specific for REMAP. An immunoaffinity column is constructed by covalently coupling anti-REMAP antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Biosciences). After the coupling, the resin is blocked and

washed according to the manufacturer's instructions.

Media containing REMAP are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of REMAP (e. g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/REMAP binding (e. g., a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and REMAP is collected.

XVII. Identification of Molecules Which Interact with REMAP Molecules which interact with REMAP may include agonists and antagonists, as well as molecules involved in signal transduction, such as G proteins. REMAP, or a fragment thereof, is labeled with Bolton-Hunter reagent. (See, e. g., Bolton A. E. and W. M. Hunter (1973) Biochem. J.

133: 529-539.) A fragment of REMAP includes, for example, a fragment comprising one or more of the three extracellular loops, the extracellular N-terminal region, or the third intracellular loop.

Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled REMAP, washed, and any wells with labeled REMAP complex are assayed. Data obtained using different concentrations of REMAP are used to calculate values for the number, affinity, and association of REMAP with the candidate molecules.

Alternatively, molecules interacting with REMAP are analyzed using the yeast two-hybrid system as described in Fields, S. and O. Song (1989) Nature 340: 245-246, or using commercially available kits based on the two-hybrid system, such as the MATCHMAKER system (Clontech).

REMAP may also be used in the PATHCALLING process (CuraGen Corp., New Haven CT) which employs the yeast two-hybrid system in a high-throughput manner to determine all interactions between the proteins encoded by two large libraries of genes (Nandabalan, K. et al. (2000) U. S.

Patent No. 6,057,101).

Potential REMAP agonists or antagonists may be tested for activation or inhibition of REMAP receptor activity using the assays described in sections XVII and XVIII. Candidate molecules may be selected from known GPCR agonists or antagonists, peptide libraries, or combinatorial chemical libraries.

Methods for detecting interactions of REMAP with intracellular signal transduction molecules such as G proteins are based on the premise that internal segments or cytoplasmic domains from an orphan G protein-coupled seven transmembrane receptor may be exchanged with the analogous domains of a known G protein-coupled seven transmembrane receptor and used to identify the G-proteins and downstream signaling pathways activated by the orphan receptor domains (Kobilka, B. K. et al. (1988) Science 240: 1310-1316). In an analogous fashion, domains of the orphan receptor may be cloned as a portion of a fusion protein and used in binding assays to demonstrate interactions with specific G proteins. Studies have shown that the third intracellular loop of G

protein-coupled seven transmembrane receptors is important for G protein interaction and signal transduction (Conklin, B. R. et al. (1993) Cell 73: 631-641). For example, the DNA fragment corresponding to the third intracellular loop of REMAP may be amplified by the polymerase chain reaction (PCR) and subcloned into a fusion vector such as pGEX (Pharmacia Biotech). The construct is transformed into an appropriate bacterial host, induced, and the fusion protein is purified from the cell lysate by glutathione-Sepharose 4B (Pharmacia Biotech) affinity chromatography.

For in vitro binding assays, cell extracts containing G proteins are prepared by extraction with 50 mM Tris, pH 7.8,1 mM EGTA, 5 mM MgCl2, 20 mM CHAPS, 20% glycerol, 10 yg of both aprotinin and leupeptin, and 20 Al of 50 mM phenylmethylsulfonyl fluoride. The lysate is incubated on ice for 45 min with constant stirring, centrifuged at 23, 000 g for 15 min at 4°C, and the supernatant is collected. 750 tug of cell extract is incubated with glutathione S-transferase (GST) fusion protein beads for 2 h at 4°C. The GST beads are washed five times with phosphate-buffered saline. Bound G subunits are detected by [32P] ADP-ribosylation with pertussis or cholera toxins. The reactions are terminated by the addition of SDS sample buffer (4.6% (w/v) SDS, 10% (v/v) (3-mercaptoethanol, 20% (w/v) glycerol, 95.2 mM Tris-HCI, pH 6.8,0.01% (w/v) bromphenol blue).

The [32P] ADP-labeled proteins are separated on 10% SDS-PAGE gels, and autoradiographed. The separated proteins in these gels are transferred to nitrocellulose paper, blocked with blotto (5% nonfat dried milk, 50 mM Tris-HCl (pH 8.0), 2 mM CaCl2, 80 mM NaCl, 0.02% NaN3, and 0.2% Nonidet P-40) for 1 hour at room temperature, followed by incubation for 1.5 hours with Ga subtype selective antibodies (1: 500; Calbiochem-Novabiochem). After three washes, blots are incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit immunoglobulin (1: 2000, Cappel, Westchester PA) and visualized by the chemiluminescence-based ECL method (Amersham Corp.).

XVIII. Demonstration of REMAP Activity An assay for REMAP activity measures the expression of REMAP on the cell surface. cDNA encoding REMAP is transfected into an appropriate mammalian cell line. Cell surface proteins are labeled with biotin as described (de la Fuente, M. A. et al. (1997) Blood 90: 2398-2405).

Immunoprecipitations are performed using REMAP-specific antibodies, and immunoprecipitated samples are analyzed using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting techniques. The ratio of labeled immunoprecipitant to unlabeled immunoprecipitant is proportional to the amount of REMAP expressed on the cell surface.

In the alternative, an assay for REMAP activity is based on a prototypical assay for ligand/receptor-mediated modulation of cell proliferation. This assay measures the rate of DNA synthesis in Swiss mouse 3T3 cells. A plasmid containing polynucleotides encoding REMAP is added to quiescent 3T3 cultured cells using transfection methods well known in the art. The transiently transfected cells are then incubated in the presence of [3Wthymidine, a radioactive DNA

precursor molecule. Varying amounts of REMAP ligand are then added to the cultured cells.

Incorporation of [Hjthymidine into acid-precipitable DNA is measured over an appropriate time interval using a radioisotope counter, and the amount incorporated is directly proportional to the amount of newly synthesized DNA. A linear dose-response curve over at least a hundred-fold REMAP ligand concentration range is indicative of receptor activity. One unit of activity per milliliter is defined as the concentration of REMAP producing a 50% response level, where 100% represents maximal incorporation of [3H] thymidine into acid-precipitable DNA (McKay, 1. and I.

Leigh, eds. (1993) Growth Factors: A Practical Approach, Oxford University Press, New York NY, p.

73.) In a further alternative, the assay for REMAP activity is based upon the ability of GPCR family proteins to modulate G protein-activated second messenger signal transduction pathways (e. g., cAMP ; Gaudin, P. et al. (1998) J. Biol. Chem. 273: 4990-4996). A plasmid encoding full length REMAP is transfected into a mammalian cell line (e. g., Chinese hamster ovary (CHO) or human embryonic kidney (HEK-293) cell lines) using methods well-known in the art. Transfected cells are grown in 12-well trays in culture medium for 48 hours, then the culture medium is discarded, and the attached cells are gently washed with PBS. The cells are then incubated in culture medium with or without ligand for 30 minutes, then the medium is removed and cells lysed by treatment with 1 M perchloric acid. The cAMP levels in the lysate are measured by radioimmunoassay using methods well-known in the art. Changes in the levels of cAMP in the lysate from cells exposed to ligand compared to those without ligand are proportional to the amount of REMAP present in the transfected cells.

To measure changes in inositol phosphate levels, the cells are grown in 24-well plates containing 1x105 cells/well and incubated with inositol-free media and [3H] myoinositol, 2, uCi/well, for 48 hr. The culture medium is removed, and the cells washed with buffer containing 10 mM LiCl followed by addition of ligand. The reaction is stopped by addition of perchloric acid. Inositol phosphates are extracted and separated on Dowex AG1-X8 (Bio-Rad) anion exchange resin, and the total labeled inositol phosphates counted by liquid scintillation. Changes in the levels of labeled inositol phosphate from cells exposed to ligand compared to those without ligand are proportional to the amount of REMAP present in the transfected cells.

In a further alternative, the ion conductance capacity of REMAP is demonstrated using an electrophysiological assay. REMAP is expressed by transforming a mammalian cell line such as COS7, HeLa or CHO with a eukaryotic expression vector encoding REMAP. Eukaryotic expression vectors are commercially available, and the techniques to introduce them into cells are well known to those skilled in the art. A small amount of a second plasmid, which expresses any one of a number of marker genes such as (3-galactosidase, is co-transformed into the cells in order to allow rapid

identification of those cells which have taken up and expressed the foreign DNA. The cells are incubated for 48-72 hours after transformation under conditions appropriate for the cell line to allow expression and accumulation of REMAP and ß-galactosidase. Transformed cells expressing ß- galactosidase are stained blue when a suitable colorimetric substrate is added to the culture media under conditions that are well known in the art. Stained cells are tested for differences in membrane conductance due to various ions by electrophysiological techniques that are well known in the art.

Untransformed cells, and/or cells transformed with either vector sequences alone or ß-galactosidase sequences alone, are used as controls and tested in parallel. The contribution of REMAP to cation or anion conductance can be shown by incubating the cells using antibodies specific for either REMAP.

The respective antibodies will bind to the extracellular side of REMAP, thereby blocking the pore in the ion channel, and the associated conductance.

In a further alternative, REMAP transport activity is assayed by measuring uptake of labeled substrates into Xenopus laevis oocytes. Oocytes at stages V and VI are injected with REMAP mRNA (10 ng per oocyte) and incubated for 3 days at 18 °C in OR2 medium (82.5 mM NaCl, 2.5 mM KC1, 1 mM CaCl2, 1 mM MgCl2, 1 mM Na2HPO4, 5 mM Hepes, 3.8 mM NaOH, 50 jig/ml gentamycin, pH 7.8) to allow expression of REMAP protein. Oocytes are then transferred to standard uptake medium (100 mM NaCl, 2 mM KC1, 1 mM CaCl2, 1 mM MgCl2, 10 mM Hepes/Tris pH 7.5). Uptake of various substrates (e. g., amino acids, sugars, drugs, and neurotransmitters) is initiated by adding a 3H substrate to the oocytes. After incubating for 30 minutes, uptake is terminated by washing the oocytes three times in Na+-free medium, measuring the incorporated 3H, and comparing with controls. REMAP activity is proportional to the level of internalized 3H substrate.

In a further alternative, REMAP protein kinase (PK) activity is measured by phosphorylation of a protein substrate using gamma-labeled [32P]-ATP and quantitation of the incorporated radioactivity using a gamma radioisotope counter. REMAP is incubated with the protein substrate, [3zP]-ATP, and an appropriate kinase buffer. The 31p incorporated into the product is separated from free [P]-ATP by electrophoresis and the incorporated 32p is counted. The amount of 32p recovered is proportional to the PK activity of REMAP in the assay. A determination of the specific amino acid residue phosphorylated is made by phosphoamino acid analysis of the hydrolyzed protein.

Transcriptional regulatory activity of REMAP is measured by its ability to stimulate transcription of a reporter gene (Liu, H. Y. et al. (1997) EMBO J. 16: 5289-5298). The assay entails the use of a well characterized reporter gene construct, LexAOp-LacZ, that consists of LexA DNA transcriptional control elements (LexAP) fused to sequences encoding the E. coli LacZ enzyme. The methods for constructing and expressing fusion genes, introducing them into cells, and measuring LacZ enzyme activity, are well known to those skilled in the art. Sequences encoding REMAP are cloned into a plasmid that directs the synthesis of a fusion protein, LexA-REMAP, consisting of

REMAP and a DNA-binding domain derived from the LexA transcription factor. The resulting plasmid, encoding a LexA-REMAP fusion protein, is introduced into yeast cells along with a plasmid containing the LexAp-LacZ reporter gene. The amount of LacZ enzyme activity associated with LexA-REMAP transfected cells, relative to control cells, is proportional to the amount of transcription stimulated by the REMAP.

XIX. Identification of REMAP Ligands REMAP is expressed in a eukaryotic cell line such as CHO (Chinese Hamster Ovary) or HEK (Human Embryonic Kidney) 293 which have a good history of GPCR expression and which contain a wide range of G-proteins allowing for functional coupling of the expressed REMAP to downstream effectors. The transformed cells are assayed for activation of the expressed receptors in the presence of candidate ligands. Activity is measured by changes in intracellular second messengers, such as cyclic AMP or Ca2+. These may be measured directly using standard methods well known in the art, or by the use of reporter gene assays in which a luminescent protein (e. g. firefly luciferase or green fluorescent protein) is under the transcriptional control of a promoter responsive to the stimulation of protein kinase C by the activated receptor (Milligan, G. et al. (1996) Trends Pharmacol. Sci. 17: 235- 237). Assay technologies are available for both of these second messenger systems to allow high throughput readout in multi-well plate format, such as the adenylyl cyclase activation FlashPlate Assay (NEN Life Sciences Products), or fluorescent Ca2+ indicators such as Fluo-4 AM (Molecular Probes) in combination with the FLIPR fluorimetric plate reading system (Molecular Devices). In cases where the physiologically relevant second messenger pathway is not known, REMAP may be coexpressed with the G-proteins G/ig which have been demonstrated to couple to a wide range of G- proteins (Offermanns, S. and M. I. Simon (1995) J. Biol. Chem. 270: 15175-15180), in order to funnel the signal transduction of the REMAP through a pathway involving phospholipase C and Ca2+ mobilization. Alternatively, REMAP may be expressed in engineered yeast systems which lack endogenous GPCRs, thus providing the advantage of a null background for REMAP activation screening. These yeast systems substitute a human GPCR and G protein for the corresponding components of the endogenous yeast pheromone receptor pathway. Downstream signaling pathways are also modified so that the normal yeast response to the signal is converted to positive growth on selective media or to reporter gene expression (Broach, J. R. and J. Thorner (1996) Nature 384 (supp.): 14-16). The receptors are screened against putative ligands including known GPCR ligands and other naturally occurring bioactive molecules. Biological extracts from tissues, biological fluids and cell supernatants are also screened.

Various modifications and variations of the described methods and systems of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with certain embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.

Table 1 Incyte Project ID Polypeptide Incyte Polynucleotide Incyte SEQ ID NO: Polypeptide ID SEQ ID NO: Polynucleotide ID Incyte Full Length Clones 3015053 1 3015053CD1 27 3015053CB1 7481761 2 7481761CD1 28 7481761CB1 942253 3 942253CD1 29 942253CB1 2118277CA2, 4157745CA2, 491408CA2, 6558412CA2 1506342 4 1506342CD1 30 1506342CB1 6301177 5 6301177CD1 31 6301177CB 1996319CA2, 2786725CA2 257833 6 257833CD1 32 257833CB1 7580043 7 7580043CD1 33 7580043CB1 8120340 8 8120340CD1 34 8120340CB1 7475307 9 7475307CD1 35 7475307CB1 90149569CA2, 9014593CA2 7475243 10 7475243CD1 36 7475243CB1 7490257 11 7490257CD1 37 7490257CB1 793400 12 793400CD1 38 793400CB1 1488208CA2 8210895 13 8210895CD1 39 8210895CB1 90089080CA2 55069745 14 55069745CD1 40 55069745CB1 90175531CA2 3212783 15 3212783CD1 41 3212783CB1 6986529 16 6986529CD1 42 6986529CB1 7474928 17 7474928CD1 43 7474928CB1 3736039 18 3736039CD1 44 3736039CB1 1284008CA2, 1619905CA2, 1703355CA2, 1986001CA2, 2211376CA2, 2924753CA2, 2992663CA2, 3277054CA2, 3641126CA2, 3736039CA2, 3960914CA2, 4073743CA2, 4672702CA2, 4821333CA2, 4947639CA2, 5557612CA2, 789492CA2, 7981643CA2 1798572 19 1798572CD1 45 1798572CB1 1500870CA2, 1579942CA2, 6495534CA2 3038391 20 3038391CD1 46 303839CB1 5822287 21 5822287CD1 47 5822287CB1 7231805 22 7231805CD1 48 7231805CB1 4032542 23 4032542CD1 49 4032542CB1 1593941 24 1593941CD1 50 1593941CB1 Table 1 Incyte Project ID Polypeptide Incyte Polynucleotide Incyte SEQ ID NO: Polypeptide ID SEQ ID NO: Polynucleotide ID Incyte Full Length Clones 3471414 25 3471414CD1 51 3471414CB1 3741414CA2 7504960 26 7504960CD1 52 7504960CB1 9008896CA2 Table 2 Polypeptide SEQ Incyte genBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID N: 1 3015053CD1 g12007720 0.0 [Mus musculus] VPS10 domain receptor protein SorCS2 Rezgaoui, M., et al. (2001) Identification of SorCS2, a novel member of the VPS10 domain containing receptor family, prominently expressed in the developing mouse brain. Mech. Dev. 100, 335-338 2 7481761CD1 g11225481 5.9e-27 [Rattus norvegicus] ETL protein Nechiporuk, T., et al. (2001) ETL, a novel seven- tranmembrane receptor that is developmentally regulated in the heart. ETL is a member of the secretin family and belongs to the epidermal growth factor-seven- transmembrane subfamily. J. Biol. Chem. 276, 4150-4157 6 257833CD1 g1326155 0.0 [Homo sapiens] melatonin-related receptor Reppert, S.M., et al. (1996) FEBS Lett. 386:219-224 Cloning of a melatonin-related receptor from human pituitary 7 7580043CD1 g459832 2.1e-219 [Homo sapiens] opioid receptor Mestek, A., et al. (1995) J. Neurosci. 15:2396- 2406 The human mu opioid receptor: modulation of functional desensitization by calcium/ calmodulin-dependent protein kinase and protein kinase C 8 8120340CD1 g339677 2.4e-51 [Homo sapiens] thrombin receptor Vu, T.H. et al. (1991) Cell 64:1057-1068 Molecular cloning of a funcional thrombin receptor reveals a novel proteolytic mechanism of receptor activaiton 9 7475307CD1 g18480666 1e-142 [Mus musculus] olfactory receptor MOR240-3 zhang, X. and Firestein, S. (2002) The olfactory receptor gene superfamily of the mouse. Nat. Neurosci. 5:124-133 10 7475243CD1 g3831958 2.8e-112 [Homo sapiens] olfactory receptor Buettner, J.A. et al. (1998) Genomics 53:56-68 Organization and envolution of olfactory receptor genes on human chromosome 11 Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 11 7490257CD1 g2792018 1.3e-116 [Homo sapiens] olfactory receptor Vanderhaeghen, P. et al. (1997) Bichem. Biophys. Res. Commun. 237:283-287 Molecular cloning and chromosomal mapping of olfactory receptor genes expressed in the male germ line: evidence for their wide distribvution in the human genome 12 793400CD1 g12597759 4.2e-42 [Arabidopsis thaliana] pheromone receptor, putative (AR401) 13 8210895CD1 g13543724 7.3e-89 [Mus musculus] progesterone receptor membrane component 14 55069745CD1 g11908216 1.1e-170 [Homo sapiens] HOR5'Beta7 Bulger, M. et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97:14560-14565 Comparatives structural and fuctional analysis of the olfactory receptor genes flanking the human and mouse betaglobin gene clusters 15 3212783CD1 g14701768 0.0 [Homo sapiens] Vam6/Vps39-like protein Caplan, S. et al. (2001) Human Vam6p promotes lysosome clustering and fusion in vivo. J. Cell Biol. 154:109-122 16 6986529CD1 g2826476 1.3e-18 [Homo sapiens] IL-17 receptor Yao, Z. (1997) Cytokine 9:794-800 Molecular characterization of the human interleukin (@@)-17 receptor 17 7474928CD1 g12407653 1.2e-81 [Homo sapiens] Nogo receptor Fournier, A.E. (2001) Nature 409:341-346 Identitification of a receptor mediating Nogo-66 inhibition of axonal regeneration 18 3736039CD1 g2071989 1.5e-24 [Mus musculus] D9 splice variant 2 Scott, L.M. (1996) Blood 88 (7), 2517-2530 E3, a hematopoietic-specific transcript directly regulated by the retinoic acid receptor alpha 19 1798572CD1 g6274473 1.6e-166 [Homo sapiens] pregnancy-induced growth inhibitor Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 20 3038391CD1 g12840671 1e-115 [Mus musculus] BECLIN 1 (COILED-COIL MYOSIN-LIKE BCL2- INTERACTING PROTEIN)#data source:SPTR, source key:O88597, evidence:ISS#putative Carninnci, P. and Hayashizaki, Y. (1999) High-efficiency full-length cDNA cloning. Meth. Enzymol. 303:19-44 Carninci, P.et al. (2000) Normalization and subtraction of cap-trapper-selected cDNAs to prepare full-length cDNA libraries for rapid discovery of new genes. Genome Res. 10:1617-1630 Shibata, K.et al. (2000) RIKEN integrated sequence analysis (RISA) system--384- format sequencing pipeline with 384 multicapillary sequencer. Genome Res. 10:1757-1771 22 7231805CD1 g18071668 0.0 [Caenorhabditis elegans] HID-1 23 4032542CD1 g16755530 0.0 [Homo sapiens] androgen receptor-associated coregulator 267-a Wang, X. et al. (2001) Identitification and Characterization of a Novel Androgen Receptor Coregulator ARA267-alpha in Prostate Caner Cells. J. Biol. Chem. 276:40417-40423 24 1593941CD1 g4530437 0.0 [Homo sapiens] thyroid hormone receptor-associated protein complex component TRAP240. Ito, M. et al. (1999) Identity between TRAP and SMCC complexes indicates novel pathways for the function of nuclear receptors and diverse mammalian activators. Mol. Cell 3:361-370. 25 3471414CD1 g1869971 3.4e-180 [Mus musculus] nuclear orphan receptor/transcription factor. Matysiak-Scholze, U. and Nehls, M. (1997) The structural integrity of POR alpha isoforms is mutated in stggerer mice: cerebellar coexpression of ROR alphal and ROR alpha4. Genomics 43:78-84. 26 7504960CD1 g2062022 7.6e-65 [Homo sapiens] putative progesterone binding protein Gerdes, D. et al. (1998) Cloning and tissue expression of two putative steroid membrane receptors. Biol. Chem. 379:907-911 Table 2 Polypeptide SEQ incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 26 568476#PGRMC1 6.6e-66 [Homo sapiens][Receptor (signalling); Small molecule-binding protein] [Plasma membrane] Transmembrane protein that binds progesterone, expressed primarily in liver and kidney Gerdes, D. et al. (1998) cloning and tissue expression of two putative steroid membrane receptors. Biol. Chem. 379:907-11 323894#Pgrmc1 3.8e-63 [Mus musculus][Receptor (signalling); Small molecule-binding protein] Protein with strong similarity to human HPR6.6, which is a transmembrane protein that binds progesterone, expressed primarily in liver and kidney Gerdes, D. et al. (1998) Cloning and tissue expression of two putative steroid membrane receptors. Biol Chem 379:907-911 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 1 3015053CD1 1159 S30 S223 S225 S234 S299 N158 N328 N362 signal_cleavage:M1-A48 SPSCAN S330 S369 S393 S433 N600 N830 N891 S589 S629 S632 S655 N902 N1140 S674 S680 S735 S851 S1031 S1034 S1128 T16 T202 T212 T283 T348 T553 T681 T695 T722 T813 T925 T1013 T1027 T1069 Y479 Y512 Y654 BNR repeat: Y273-L284, K587-T598, F232-K243, HMMER_PFAM Y545-O556, W182-K193, L468-Y479 Transmembrane domains: P31-G59 G1077-K1105 TMAP N-terminus is cytosolic GLYCOPROTEIN PROTEIN PRECURSOR BLAST_PRODOM SIGNAL TRANSMEMBRANE LR11 PUTATIVE MEMBRANE VACUOLAR RECEPTOR D007682:W634-P776 PD004576:L526-V608 YIL173W; MEMBRANE; DM0179#P32319#1110- BLAST_DOMO 1233: H518-G626 C-type lectin domain signature: C684-C708 MOTIFS Table 3 SEQ incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites NO: ID 2 7481761CD1 722 S11 S173 S212 S229 S255 N87 N112 N193 signal_cleavage:M1-G29 SPSCAN S318 S330 S341 S417 N253 N308 N348 S476 S481 S511 S546 N367 N371 N402 S565 S647 S689 T41 N407 N439 N447 T124 T175 T181 T282 N470 N498 N503 T286 T293 T449 Y426 N542 N563 N645 Signal Peptide: M1-G29 HMMER Sushi domain (SCR repeat): C179-C234, C239-C294 HMMER_PFAM Transmembrane domain: P10-A28 TMAP N-terminus is non-cytosolic GLYCOPROTEIN EGF-LIKE DOMAIN BLAST_PRODOM VACUOLAR SORTING PROTEIN RECEPTOR RECEPTOR-LIKE HOMOLOG CODED PD101665: C42-Y156 (P-value = 9.4e-09) EGF-LIKE DOMAIN DM00864#I55476#159-241: BLAST_DOMO C91-Y156 (P-value = 5.53-09) EGF-like domain signature 2: C57-C71 MOTIFS Calcium-binding EGF-like domain pattern signature: MOTIFS D73-C100 D125-C150 Aspartic acid and asparagine hydroxylation site: C91- MOTIFS C102 C141-C152 3 942253CD1 107 T28 T30 signal_cleavage: M1-G18 SPSCAN BolA-like protein: T30-R107 HMMER_PFAM do BOLA; RPON; H1082; 9.2; BLAST_DOMO DM01731#P53082#38-119: T30-H96 (P-value=9.3e- 08) Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 4 1506342CD1 5208 S10 S43 S45 S69 S88 N37 N38 N100 Zinc finger, C3HC4 type (RING finger): C3998- HMMER_PFAM S226 S232 S411 S440 N346 N896 N1073 C4036 S653 S654 S671 S752 N1192 N1380 S858 S883 S980 S1018 N1523 N1602 S1128 S1187 S1188 N1735 N2327 S1205 S1258 S1264 N2491 N2611 S1282 S1422 S1451 N2675 N2907 S1512 S1573 S1581 N2987 N3552 S1596 S1644 S1686 N3701 N3705 S1773 S1785 S1815 N3828 N3889 S1991 S2105 S2122 N4388 N4906 N5123 S2148 S2211 S2293 S2532 S2549 S2607 S2638 S2703 S2774 S2799 S2853 S2905 S2914 S2971 S2976 S2989 S3027 S3112 S3187 S3214 S3296 S3394 S3513 S3529 S3642 S3713 S3780 S3842 S4143 S4182 S4191 S4245 S4285 S4286 S4412 S4422 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signautre Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 4 S4438 S4513 S4635 S4658 S4659 S4670 S4711 S4743 S4809 S4973 S5018 S5027 S5103 S5158 T14 T84 T156 T186 T200 T293 T317 T554 T830 T910 T966 T1053 T1196 T1299 T1315 T1392 T1521 T1604 T1692 T1737 T1858 T1869 Transmembrane domains: T372-F393 S618-H464 TMAP T1870 T2022 T2131 L757-R783 A869-V885 L1081-I1109 G2065-R2089 T2193 T2243 T2337 L2244-T2264 A2748-L2768 L3665-G3689 G4442- T2387 T2441 T2445 Q4459 T4570-L4593 T2630 T267 T2822 N-terminus is non-cytosolic T3059 T3097 T3136 T3179 T3327 T3339 T3370 T3380 T3450 T3496 T3533 T3620 T3654 T3731 T3776 T3781 T3796 T3875 T3978 T4443 T4483 T4749 T4846 T4859 T4865 T4948 T5125 T5147 Y589 Y689 Y1504 Y1700 Y2305 Y3693 Y4897 Y4909 Leucine zipper pattern: L2565-L2586 L2572-L2593 MOTIFS L4863-L4884 L4870-L4891 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 4 ATP/GTP-binding site motif A (P-loop): G2420- MOTIFS T2427 Cytochrome c family heme-binding site signature: MOTIFS C4526-A4531 Eukaryotic putative RNA-binding region RNP-1 MOTIFS signature: R3426-F3433 5 6301177CD1 222 S156 S196 S217 T172 signal_cleavage: M1-R27 SPSCAN T192 Signal Peptide: M1-G18 HMMER Transmembrane domains: M1-H28 T41-L69 L73- TMAP L101 I141-G158 N-terminus is cytosolic 6 257833CD1 673 S62 S68 S131 S201 S323 7 transmembrane receptor (rhodopsin family): S123- HMMER_PFAM S567 S633 S652 T110 Y354, G45-A63 T572 T588 T639 Transmembrane domain: L27-K55, E122-S150, TMAP Q160-Y187, R198-I226, L243-I271, V293-A321, Y336-L356, Y365-D385 N-terminus is non-cytosolic G-protein coupled recept BL00237: W154-S193, BLIMPS_BLOCKS F257-Y268, L290-L316, N346-R362 G-protein coupled receptors signature: G165-T212 PROFILESCAN Rhodopsin-like GPCR superfamily signature BLIMSP_PRINTS PR00237:F30-T54, S123-L144, T168-I190, N204- Y225, T249-W272, N295-L319, Y336-R362 Melatonin receptor signaure PR00857: A328-Y340, BLIMPS_PRINTS S193-Y208, N223-C238, C238-H256, Q289-F303 Table 3 SEQ Incyte Amio Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 6 MELATONIN RELATED RECEPTOR GPROTEIN BLAST_PRODOM COUPLED TRANSMEMBRANE H9 PD03916: G355-I662 PD027031:M1-K57 RECEPTOR COUPLED GPROTEIN BLAST_PRODOM TRANSMEMBRANE GLYCOPROTEIN PHOSPHORYLATION LIPOPROTEIN PALMITATE FAMILY PD000009: R111-G227 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO P49288#18-307: N124-Y365, M35-S62 P49219#26- 315: N124-Y365, M35-S62 P49285#23-313: N124- Y365, P21-S62 P48040#37-327: N124-Y365, P19- S62 Cytochrome c family heme-binding site signature: MOTIFS C188-S193 G-protein coupled receptors signature: G174-I190 MOTIFS Serine proteases, trypsin family, histidine active site: MOTIFS L595-C600 7 7580043CD1 462 S17 S46 S61 S97 S325 N71 N74 N95 N102 Signal_cleavage: M1-C23 SPSCAN S420 S427 T42 T244 N110 N426 T271 T343 T371 7 transmembrane receptor (rhodopsin family): G149- HMMER_PFAM Y400 Transmembrane domain: L138-V158, T165-L185, TMAP Y192-Y212, P245-K273, N294-R322, D336-Y363 N-terminus is non-cytosolic G-protein coupled receptor BL00237:R337-Y363, BLIMPS_BLOCKS N392-K408, W197-P236, F305-Y316 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 7 G-protein coupled receptors signature: S209-V253 PROFILESCAN Rhodopsin-liek GPCR superfamily siganture BLIMPS_PRINTS PR00237:T134-V158, T167-Q188, D211-V233, N247-F268, K297-I320, I342-I366, W382-K408 Opioid receptor signautre PR00384: R159-F172, BLIMPS_PRINTS D211-C223, L240-K249, L329-R341, F384-T391, F402-F411 Mu opioid receptor signature PR00537: M267-D280, BLIMPS_PRINTS K367-F384, I416-T428, H437-L451, C75-P88, G91- C107, C107-C121 RECEPTOR COUPLED GPROTEIN BLAST_PRODOM TRANSMEMBRANE GLYCOPROTEIN PD000009: K162-P265 PD012409: A401-L451 PD028780: R340-R412 PD014949: M63-L99 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO P35372#64-354: S126-I416 I48665#62-352: S126-I416 P41143#43-333: S128-C415 P41144#53-345: S126- C415 G-protein coupled receptors signature: T217-V233 MOTIFS 8 8120340CD1 463 S66 S130 S131 S426 T7 N5 N11 N64 7 transmembrane receptor (rhodopsin family): G40- HMMER_PFAM T174 T340 Y293 Transmembrane domain: L17-L45, S58-Y81, H88- TMAP Y108, T115-R135, A142-D162, H168-A188, L194- T214, V236-R264 N-terminus is non-cytosolic G-protein coupled receptor BL00237: W89-P128, BLIMPS_BLOCKS F202-Y213, E230-V256, N285-Q301 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 8 G-protein coupled receptors signature: T100-T147 PROFILESCAN Rhodopsin-like GPCR superfamily signature BLIMPS_PRINTS PR00237:A25-C49, S58-Q79, F103-V125, Y139- R160, L194-I217, A235-A259, Y275-Q301 RECEPTOR COUPLED GRPOTEIN BLAST_PRODOM TRANSMEMBRANE PD000009: R50-L158 PD155551:F195-A334 PUTATIVE G PROTEINCOUPLED RECEPTOR BLAST_PRODOM GPR43 GPROTEIN COUPLED TRANSMEMBRANE PD066158:I173-S297 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO P47749#95-381: L26-L308 P55085#69-360:V24- Q301 Q00991#98-388: L17-L304 I48705#71-362: V24- L308 G-protein coupled receptors signature: S109-V125 MOTIFS 9 7475307CD1 310 S78 S193 S229 T288 N5 Signal_cleavage: M1-T38 SPSCAN 7 transmembrane receptor (rhodopsin family): G41- HMMER_PFAM Y287 Transmembrane domain: Q24-D52, Q95-Y123, T138- TMAP L166, E196-M224, G230-Y258 N-terminus is cytosolic G-protein coupled receptor BL00237: K90-P129, BLIMPS-BLOCKS K231-I257, T279-K295 G-protein coupled receptors signature: F102-A147 PROFILESCAN Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 9 Rhodopsin-like GPCR superfamily signature BLIMPS_PRINTS PR00237:F26-T50, M59-K80, F104-I126, L140- A161, M199-L222, A236-W260, K269-K295 Olfactory receptor signature PR00245: M59-K80, BLIMPS-PRINTS F177-D191, A237-I252, V271-L282, T288-G302 RECEPTOR OLFACTORY GPROTEIN COUPLED BLAST_PRODOM TRANSMEMBRANE GLYCOPROTEIN MULTIGENE FAMILY PD000921:L166-I245 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO S29710#15-301: S18-L301 P23266#E17-306: S18-L301 P23275#17-306:S18-L301 P30955#18-305: Q24-L301 G-protein coupled receptors signature: G110-I126 MOTIFS 10 7475243CD1 333 S11 S12 S86 S156 S207 N24 7 transmembrane receptor (rhodopsin family): G60- HMMER_PFAM S214 S310 T97 T223 Y309 T319 Transmembrane domain: F36-M64, H75-H92, I110- TMAP A138, I218-K246, F257-C279, S285-N313 G-protein coupled recept BL00237: R109-P148, I301- BLIMPS_BLOCKS K317 G-protein coupled receptors signature: F121-S171 PROFILESCAN Olfactory receptor signature PR00245: M78-K99, BLIMPS_PRINTS F196-D210, F257-G272, A293-L304, S310-V324 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID RECEPTOR OLFACTORY PROTEIN GPROTEIN BLAST_PRODOM COUPLED TRANSMEMBRANE GLYCOPROTEIN MULTIGENE FAMILY PD000921:L185-L264 PD149621:T265-V324 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO S51356#18-307: P40-I325 P37067#17-306: P40-L323 S29709#11-299: P40-K322 P23266#17-306: S37-V324 G-protein coupled receptors signature: T129-V145 MOTIFS 11 7490257CD1 313 S66 S74 S273 S289 N5 N64 7 transmembrane receptor (rhodopsin family): G40- HMMER_PFAM Y288 Transmembrane domain: R25-I45, I49-D69, Q99- TMAP F122, M135-L163, T191-L213 N-terminus is non-cytosolic G-protein coupled receptor BL00237: K89-P128, BLIMPS_BLOCKS T280-K296 G-protein coupled receptors signature: Y101-V146 PROFILESCAN Rodopsin-like GPCR superfamily signature BLIMSP_PRINTS PR00237: R25-I49, M58-K79, F103-I125, L139- L160, A270-K296 Olfactory receptor signature PR00245: A272-L283, BLIMPS_PRINTS S289-L303, M58-K79, F176-D190, F236-C251 OLFACTORY RECEPTOR GPROTEIN COUPLED BLAST_PRODOM TRANSMEMBRANE GLYCOPROTEIN MULTIGENE FAMILY PD149621: S244-K306 PD000921:L165-L243 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 11 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO P23265#17-306:F17-L303 P23268#18-307:F17-L303 P23274#18-306:F26-L299 S29707#18-306:F26-K300 G-protein coupled receptors signature: L109-I125 MOTIFS 12 793400CD1 236 S3 S17 S21 S82 S120 T48 N104 N139 N193 Transmembrane domain: K74-F102 TMAP T106 T195 T215 Y42 N-terminus is non-cytosolic FGGY family of carbohydrate kinases signature 2: MOTIFS S103-E123 13 8210895CD1 182 S14 S57 S141 S168 S177 Signal_cleavage:M1-G48 SPSCAN T9 T139 T148 Y167 Transmembrane domain: L21-R47 TMAP PROTEIN BINDING PUTATIVE BLAST_PRODOM PROGESTERONE CHROMOSOME MEMBRANE STEROID ASSOCIATED RECEPTOR COMPONENT PD006731:D59-S177 PUTATIVE PROGESTERONE BINDING BLAST_PRODOM PROTEIN MEMBRANE ASSOCIATED RECEPTOR COMPONENT 25DX STEROID PD012960: M1-D59 Cell attachment sequence: R47-D49 MOTIFS 14 55069745CD1 326 S120 S242 S305 S325 N16 7 transmembrane receptor (rhodopsin family): G53- HMMER_PFAM Y304 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID Transmembrane domain: E33-I61, A79-F99, H117- TMAP A137, Q151-P179, F206-I233, F250-K278, P281- I303 N-terminus is non-cytosolic G-protein coupled recept BL00237: H102-P141, BLIMPS_BLOCKS R247-V273, P296-H312 Olfactory receptor signature PR00245: M288-L299, BLIMPS_PRINTS S305-L319, M71-T92, S189-D203, F250-V265 Melanocortin receptor family signature PR00534: T63- BLIMPS_PRINTS L75, I138-T149 PUTATIVE GPROTEIN COUPLED RECEPTOR BLAST_PRODOM RA1C PD170483: V260-L318 RECEPTOR OLFACTORY GPROTEIN COUPLED BLAST_PRODOM TRANSMEMBRANE GLYCOPROTEIN MULTIGENE FAMILY PD000921: Y180-L257 G-PROTEIN COUPLED RECEPTORS DM00013 BLAST_DOMO G45774#18-309: I39-L319 H45774#28-318: G31-L319 P30954#29-316: I39-G320 P23269#15-304: K36-L319 15 3212783CD1 886 S69 S74 S91 S162 S195 N72 N242 N432 Domain found in NIK1-like kinase PF00780I: N242- BLIMPS_PFAM S337 S339 S405 S452 E271 PF00780E: K71-T109 PF00780G: E174-K217 S526 S672 S780 T129 T202 T236 T421 T449 T784 CHROMOSOME I VACULOAR ASSEMBLY BLAST_PRODOM VPS39 MORPHOGENESIS VAM6 PD018571: Q544-S878, V309-D564, I18-Q329 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 15 Leucine zipper pattern: L754-L775, L761-L782 MOTIFS 16 6986529CD1 595 S24 S120 S187 S219 S300 N27 N62 N133 Transmembrane domain: A151-K179 TMAP S351 S385 S394 S474 N-terminus is non-cytosolic S510 S547 S575 S584 T102 T134 T135 T505 Y186 Y291 RECEPTOR IL17 INTERLEUKIN PD044054: E191-BLAST_PRODOM G519 17 7474928CD1 401 S263 T43 T5 T82 T271 N31 N74 N161 Signal_cleavage:M1-C27 SPSCAN T347 N217 N306 Signal Peptide: M1-C18 HMMER leucine Rich Repeat: A88-E112, R209-P232, R113- HMMER_PFAM V136, N64-Q87, S137-A160, N161-G184, S185- S208, S42-S63 Leucine rich repeat C-terminal domain: N242-P292 HMMER_PFAM Leucine-rich repeat signature PR00019:L138-L151, BLIMPS_PRINTS L159-L172 Leucine zipper pattern: L141L162 MOTIFS 18 3736039CD1 81 Signal_cleavage: M1-Q56 SPSCAN Transmembrane domain: K27-R55 TMAP N-terminus is non-cytosolic Tropomyosins signature: D26-L74 PROFILESCAN D9 SPLICE VARIANT PD019594:E13-F81 BLAST_PRODOM D9 SPLICE VARIANT A B PD027075: M1-K29, BLAST_PRODOM E48-L74 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 19 1798572CD1 477 S3 S4 S12 S64 S270 S296 Transmembrane domain: A280-A308, G382-L402 TMAP S353 S398 S421 S436 T42 N-terminus is non-cytosolic T58 T108 T171 T199 T437 T473 CODED FOR BY C. ELEGANS CDNA CM11H1 BLAST_PRODOM PD135944:G100-I349 20 3038391CD1 247 S23 S52 S82 S164 S206 N60 Cyclin: T75-Y112 HMMER_PFAM T38 T75 T105 T128 Transmembrane domain: S164-H186, Q191-L215 TMAP N-terminus is non-cytosolic 21 5822287CD1 981 S69 S200 S336 S415 S724 N379 N452 N811 Signal Peptide: M1-G27 HMMER S798 S800 S854 S874 S902 S911 S924 T158 T217 T243 T288 T363 T414 T424 Transmembrane domain: S4-F30, Y476-L504, G533- TMAP G561, L568-L590, V596-L624, P635-Y662, S672- A700 PROTEIN REPEAT MICROTUBULE- BLAST_PRODOM ASSOCIATED MICROTUBULES PHOSPHORYLATION BASSOON ALTERNATIVE SPLICING LARGE PROLINE-RICH PD005493: T243-P457 22 7231805CD1 788 S42 S87 S162 S166 S170 N96 N360 Transmembrane domain: H137-R165, L386-D408, TMAP S361 S444 S559 S648 G422-P442, P452-S472, T516-I540 S670 S687 T18 T167 N-terminus is cytosolic T311 T362 T457 T584 T597 T612 T627 T637 Tabl3 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 23 4032542CD1 2696 S96 S166 S179 S209 S213 N38 N88 N155 PHD-finger: W1709-G1751, V1545-G1589, E2120- HMMER-PFAM S259 S277 S316 S356 N190 N264 N275 C2167 S373 S419 S446 S451 N729 N756 N1650 S466 S486 S503 S510 N1706 N2076 S585 S647 S656 S661 N2148 S707 S752 S789 S811 S830 S850 PWWP domain: K1753-G1829, K320-R401 HMMER-PFAM S868 S871 S935 S965 S981 S1000 S1008 S1011 S1027 S1077 S1086 S1123 S1143 S1212 S1243 S1269 S1278 S1309 SET domain: C1935-G2064 HMMER-PFAM S1310 S1337 S1382 S1402 S1432 S1491 S1498 S1510 S1528 S1599 S1782 S1821 S1937 S2096 S2123 S2132 PWWP domain proteins: PF00855: D325-I341 BLIMPS-PFAM S2173 S2194 S2201 S2251 S2265 S2312 S2330 S2344 S2369 S2397 S2403 S2415 S2516 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 23 T1098 T1338 T1409 SET domain proteins: PF00856: G1953-Q1989, BLIMPS-PFAM T1461 T1514 T1694 I2007-E2028 T1833 T1958 T2029 T2109 T2150 T2203 T2407 T2462 Y409 Y1834 S2556 S2599 S2631 PROTEIN TRANSCRIPTION REPRESSOR: BLIMPS-PRODOM S2687 T145 T157 T175 PD02411:P2025-Y2058 T197 T235 T266 T303 T317 T577 T640 T641 T881 T1017 T1050 NSD1 PROTEIN: PD185955: V413-N1544, M1- BLAST-PRODOM G324, R2346-S2532, A2247-D2358, L1838-N1903, K1874-T1923, P2230-D2358, L2354-Q2416, C1699- S1724, T2395-L2439 PD185332: C2062-K2696 PD178526: A1750-L1848 PD173847: G1632-S1708 SET DOMAIN: DM01286#P46995#8-258: Q1883- BLAST-DOMO L2081 DM01286#P38827#814-1078: P1869-L2081 DM01286#P20659#3548-3757: K1938-L2081 DM01286#P42124#508-759: S1890-Y2058 Leucine_Zipper: L2418-L2439 MOTIFS RGD: R941-D943 MOTIFS Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 24 1593941CD1 2210 S13 S23 S253 S288 S314 N158 N217 N387 Transmembrane Domain: P266-F284 TMAP S361 S365 S391 S464 N535 N745 N1495 N-terminus is cytosolic S492 S515 S521 S522 N1551 N1567 S593 S624 S629 S635 N1862 S708 S741 S806 S817 S831 S923 S1006 S1071 S1152 S1153 S1319 S1489 S1652 S1820 S1842 S1864 S1870 S1925 S1938 S2076 S2150 T179 ATP/GTP-binding site motif A (P-loop): G1463- MOTIFS T232 T329 T400 T418 T1470 T536 T594 T717 T737 T779 T857 T1043 T1050 T1053 T1056 T1059 T1088 T1215 T1343 T1654 T1663 T1735 T1783 T1815 T1987 T2103 T2161 Y241 Y261 Y1148 Y1271 25 3471414CD1 336 S84 S96 S326 T65 T229 N49 N92 N189 Ligand-binding domain of nuclear hormone: I138- HMMER-PFAM T289 T298 Y103 Y146 C294 Zinc-finger, C4 type (two domains): I16-D86 HMMER-PFAM Transmembrane Domain: L228-L249 TMAP Nuclear hormones receptors DNA-binding region BLIMPS-BLOCKS proteins: BL00031: C18-A50, Y52-M83 Nuclear hormones receptors DNA-binding region PROFILESCAN signature (steroid_finger.prf): A8-R64 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 25 C4-type steroid receptor zinc-finger signature: BLIMPS-PRINTS PR00047:L75-M83, C18-T34, T34-N49, R67-L75 Vitamin D receptor signature: PR00350: C18-T34, BLIMPS-PRINTS C35-C54, D195-F215, A234-V253, I279-M302 Steroid hormone receptor signature: PR00398: C18- BLIMPS-PRINTS C35, K39-P55, C60-C78, A143-Q162, Q165-F185 PROTEIN RECEPTOR NUCLEAR BLAST-PRODOM TRANSCRIPTION REGULATION DNA-BINDING ZINC-FINGER HORMONE FAMILY MULTIGENE: PD000035: C18-S84 RECEPTOR NUCLEAR TRANSCRIPTION BLAST-PRODOM REGULATION DNA-BINDING PROTEIN ZINC- FINGER ACTIVATOR ALTERNATIVE SPLICING: PD009370: C294-E329 PD005318: M83-I140 NUCLEAR HORMONES RECEPTORS DNA- BLAST-DOMO BINDING REGION: DM00047#P35397#63-439: V81- V253 DM00047#P51449#21-436: E87-K252 DM00047#P31396#41-405: A8-E99 DM00047#B56856#1-378: V81-E251 Nuclear Receptor: C18-R44 MOTIFS 26 7504960CD1 171 S14 S130 S157 S166 T9 signal_cleavage: M1-K44 SPSCAN T50 T128 T137 Y156 Steroid binding domain: D48-L147 HMMER_PFAM Cytosolic domain: Y43-D171 TMHMMER Transmembrane domain: G20-L42 Non-cytosolic domain: M1-G19 Table 3 SEQ Incyte Amino Acid Potential Phosphorylation Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Sites Glycosylation Sites and Databases NO: ID 26 PROTEIN BINDING PUTATIVE BLAST_PRODOM PROGESTERONE CHROMOSOME MEMBRANE STEROID ASSOCIATED RECEPTOR COMPONENT PD006731:R47-S166 PUTATIVE PROGESTERONE BINDING BLAST_PRODOM PROTEIN MEMBRANE ASSOCIATED RECEPTOR COMPONENT 25DX STEROID PD012960:M1-A52 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 27/3015053CB1/6073 1-237, 130-500, 130-526, 152-789, 161-6000, 461-1305, 657-1388, 737-946, 737-1009, 737-1095, 737-1162, 737- 1170, 737-1264, 737-1290, 737-1308, 737-1397, 737-1408, 737-1496, 737-1545, 764-1436, 830-1402, 833-1496, 838-1453, 862-1652, 865-1406, 880-1164, 917-1421, 953-1452, 972-1376, 1014-1467, 1027-1573, 1029-1457, 1091- 1525, 1098-1595, 1106-1463, 1116-1605, 1127-1311, 1141-1398, 1176-1598, 1181-1784, 1199-1608, 1213-1995, 1271-1784, 1414-1940, 1414-2054, 1438-1995, 1449-1595, 1496-1888, 1522-2119, 1522-2192, 1875-1908, 1974- 2631, 1975-2676, 2026-2296, 2069-2607, 2148-2631, 2161-2518, 2163-2516, 2163-2518, 2173-2518, 2474-2501, 2586-2658, 3087-3344, 3087-3629, 3089-3338, 3089-3341, 3143-3369, 3143-3370, 3162-3278, 3162-3413, 3225- 3370, 3233-3843, 3515-3780, 3515-3784, 3515-4021, 3681-3939, 3701-3900, 3709-4287, 3740-4151, 3766-4275, 3808-4340, 3825-4397, 3827-4397, 3860-4141, 3867-4504, 3873-4408, 3908-4517, 3935-4561, 3943-4193, 4002-4360, 4006-4223, 4011-4614, 4013-4584, 4019-4494, 4026-4671, 4088-4531, 4091-4723, 4101-4688, 4107- 4672, 4111-4575, 4115-4732, 4122-4564, 4140-4656, 4153-4758, 4153-4789, 4172-4708, 4220-4507, 4230-4859, 4233-4904, 4269-4772, 4308-4973, 4311-4915, 4363-5032, 4386-4574, 4388-5030, 4392-4886, 4406-4898, 4425- 4648, 4434-5092, 4441-4981, 4444-5049, 4453-4999, 4459-4921, 4464-5006, 4474-5035, 4476-5049, 4477-4942, 4497-4715, 4499-5056, 4502-4941, 4510-4791, 4510-5148, 4510-5183, 4517-5106, 4519-5071, 4523-5184, 4528- 5032, 4532-5052, 4532-5078, 4538-4995, 4576-5143, 4603-5148, 4625-4948, 4640-4882, 4651-5175, 4665-4937, 4675-5290, 4678-5203, 4693-4840, 4695-5284, 4738-5214, 4738-5230, 4756-5298, 4768-4808, 4768-4810, 4768- 4830, 4768-4832, 4768-4836, 4768-4843, 4768-4849, 4768-4865, 4768-4869, 4768-4870, 4768-4880, 4768-4901, 4768-4902, 4768-4908, 4769-5431, 4773-4835, 4773-4836, 4773-4956, 4774-4908, 4792-4907, 4795-4908, 4805-5284, 4805-5458, 4833-5451, 4834-5464, 4840-4894, 4840-4912, 4840-4941, 4840-4952, 4840-4972, 4840- 4974, 4840-4980, 4841-4980, 4845-4978, 4845-4980, 4864-5282, 4867-5410, 4877-4980, 4888-4980, 4905-4980, 4906-4980, 4912-4966, 4912-4974, 4912-4978, 4912-4980, 4917-4974, 4917-4980, 4936-4980, 4939-4980, 4955- 4980, 4972-5582, 4992-5534, 5023-5588, 5027-5313, 5027-5548, 5027-5553, 5028-5591, 5028-5592, 5044-5343, 5055-5312, 5066-5676, 5071-5410, 5074-5609, 5075-5344, 5081-5565, 5109-5682, 5132-5505, 5237-5420, 5261- 5893, 5300-5599, 5300-5970, 5332-5568, 5336-5994, 5338-5990, 5345-5647, 5356-5581, 5357-5654, 5363-6035, 5372-5990, 5392-5985, 5397-5983, 5411-6025, 5412-6015, 5416-6033, 5452-6026, 5456-5962, 5463-6000, 5475- 5737, 5497-5780, 5529-5791, 5536-6060, 5536-6073, 5537-6029, 5537-6073, 5539-5851, 5540-5821, 5544-6061, 5569-6073, 5582-6041, 5583-6041, 5586-6041, 5588-6058, 5598-6063, 5621-6073, 5623-6041, 5624-6064, 5638- 6041, 5645-6041, 5681-6064, 5722-6041, 5725-5967, 5729-5983, 5788-6041, 5861-6045, Table 4 Polynucleotide Sequence Fragments SEQ Id NO:/ Incyte ID/ Sequence Length 28/7481761CB1/2735 1-610, 1-2511, 244-911, 310-596, 381-500, 381-996, 381-1165, 412-529, 444-684, 614-1116, 881-1273, 1024-1405, 1141-1370, 1233-1830, 1543-1773, 1546-1884, 1580-2116, 1648-1785, 1654-1912, 1679-1964, 1712-2366, 1721- 1974, 1721-2210, 1721-2238, 1783-1957, 1806-2021, 1870-2139, 1945-2172, 2025-2291, 2032-2345, 2043-2274, 2059-2735, 2107-2337, 2113-2532 29/942253CB1/534 1-71, 1-232, 1-251, 1-270 1-274, 1-348, 1-406, 1-491, 1-534 3-475, 4-270, 4-296, 4-311, 7-350, 10-531, 12-277, 14- 268, 15-300, 18-333, 22-305, 23-290, 23-302, 23-471, 23-534, 24-259, 24-282, 24-486, 28-303, 30-534, 35-297, 42- 393, 43-307, 50-534, 52-234, 69-478, 74-526, 76-438, 78-525, 78-526, 79-456, 81-525, 81-532, 87-526, 89-527, 95- 526, 100-525, 102-525, 117-514, 120-525, 125-529, 150-367, 151-525, 172-427, 172-437, 181-526, 183-526, 196- 525, 206-525, 211-525, 215-526, 215-529, 232-508, 238-531, 242-525, 244-512, 244-534, 284-456, 321-518, 393- 527 30/1506342CB1/16159 1-571, 63-748, 116-407, 196-388, 196-520, 196-564, 198-512, 201-813, 292-935, 293-747, 313-834, 315-529, 594- 1172, 605-1174, 646-1309, 723-1183, 785-1380, 884-1277, 1114-1237, 1236-1382, 1236-1947, 1236-1968, 1236- 1969, 1236-1990, 1236-2005, 1236-2069, 1312-1943, 1463-2159, 1484-2036, 1615-2192, 1775-2042, 1775-2058, 1871-2241, 1901-2460, 1902-2172, 2059-2315, 2059-2492, 2059-2516, 2100-2766, 2102-2516, 2147-2513, 2173- 2220, 2173-2921, 2173-2952, 2173-2953, 2173-2964, 2179-2982, 2185-3032, 2201-3032, 2234-3032, 2239-2513, 2247-3032, 2249-3032, 2250-3032, 2292-3032, 2343-3104, 2423-2958, 2441-2718, 2441-2900, 2511-3020, 2593- 2886, 2700-3246, 2748-3069, 2748-3264, 2864-3319, 2933-3630, 2954-3327, 3085-3327, 3088-3285, 3119-3614, 3128-3258, 3128-3275, 3129-3306, 3131-3327, 3279-3509, 3438-4041, 3452-4089, 3456-3719, 3456-4068, 3499- 4089, 3572-3818, 3572-4089, 3572-4091, 3572-4102, 3572-4106, 3572-4122, 3572-4147, 3572-4162, 3572-4199, 3572-4205, 3572-4225, 3572-4232, 3572-4241, 3572-4265, 3572-4275, 3572-4278, 3621-3866, 3621-4103, 3624- 3833, 3674-3933, 3674-4278, 3688-4245, 3728-4459, 3750-4447, 3823-4313, 3858-4538, 3933-4381, 3965-4588, 3993-4476, 4017-4043, 4072-4446, 4087-4338, 4116-4830, 4129-4381, 4138-4315, 4138-4363, 4138-4663, 4149- 4701, 4187-4476, 4242-4830, 4254-4567, 4301-4830, 4335-5030, 4373-5215, 4382-4830, 4386-4830, 4435-4830, 4491-4830, 4502-5130, 4519-4830, 4568-4830, 4592-5142, 4599-4830, 4603-4830, 4831-5052, 4836-5465, 4845- 5478, 4931-5412, 4938-5183, 4938-5529, 5137-5270, 5142-5695, 5203-5858, 5256-5811, 5534-6164, 5554-6023, 5861-6313, 5863-6305, 5870-6328, 5881-6328, 6001-6611, 6134-6646, 6293-6602, 6420-6583, 6421-6972, 6421- 7087, 6454-7116, 6455-6928, 6633-7204, 6651-7342, 6681-7377, 6685-7209, 6730-7446, 7180-7497, 7238-7902, 7240-7794, 7268-7499, 7708-7834, 7708-8345, 7759-8294, 7782-8502, 7791-8330, 7793-8377, 7810-8456, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 30 7838-8265, 7870-8598, 7913-8214, 7913-8343, 7913-8346, 7913-8351, 7913-8402, 7914-8340, 7924-8339, 7956- 8196, 7956-8489, 7960-8356, 7991-8267, 7993-8671, 8021-8411, 8058-8745, 8214-8887, 8252-8576, 8270-8797, 8278-8924, 8290-8883, 8331-8745, 8333-8775, 8333-9024, 8338-9020, 8423-9008, 8439-9055, 8439-9062, 8476- 8890, 8479-8890, 8522-8968, 8661-9303, 8661-9447, 8831-9450, 8945-9304, 9046-9513, 9050-9798, 9077-9798, 9501-10275, 9526-10193, 9633-10228, 9680-10463, 9791-10129, 9791-10421, 9804-10421, 9916-10174, 10133- 10588, 10211-10518, 10258-10394, 10324-10876, 10340-10922, 10390-10696, 10390-10948, 10457-10704, 10602- 10895, 10804-11406, 10840-11417, 10899-11335, 10938-11524, 11136-11380, 11136-11506, 11185-11792, 11212- 11837, 11250-11506, 11289-11566, 11303-11800, 11436-12014, 11449-12029, 11514-12106, 11558-12126, 11612- 12287, 11849-12486, 11934-12627, 11944-12067, 11944-12600, 11985-12382, 12178-12555, 12236-12860, 12396-12711, 12484-12820, 12540-12786, 12551-12763, 12551-12923, 12551-13001, 12551-13183, 12511-13192, 12555-13189, 12582-13210, 12615-12943, 12678-12750, 12684-13218, 12771-13200, 12771-13380, 12788-13380, 12809-13097, 12809-13463, 12810-13382, 12884-13231, 12905-13380, 12913-13224, 12926-13380, 12955-13504, 12995-13606, 12996-13278, 12997-13177, 12998-13241, 13043-13412, 13048-13719, 13054-13594, 13106-13704, 13191-13875, 13249-13468, 13302-13916, 13317-13380, 13330-14135, 13332-13380, 13589-14225, 13623-14327, 13756-14207, 13809-14446, 13889-14101, 13909-14133, 13915-14191, 13975-14626, 14092-14672, 14093-14561, 14168-14725, 14285-14575, 14285-14807, 14287-14907, 14292-14739, 14309-14801, 14326-14973, 14358-14859, 14387-15236, 14402-15019, 14402-15051, 14458-15019, 14471-14704, 14484-14749, 14493-14949, 14504-15084, 14511-14801, 14540-14843, 14540-15139, 14541-15114, 14549-14807, 14579-14843, 14580-15152, 14583-15210, 14622-15387, 14645-15218, 14656-15285, 14675-15470, 14681-15340, 14688-15433, 14761-15210, 14814-15393, 14840-15116, 14881-15143, 14897-15147, 14928-15216, 14932-15216, 14932-15306, 14948-15358, 14981-15261, 15008-15611, 15009-15306, 15026-15387, 15050-15601, 15087-15346, 15087-15348, 15110-15696, 15142-15717, 15142-15730, 15142-15758, 15142-15786, 15142-15799, 15163-15711, 15177-15850, 15206-15769, 15239-15908, 15267-15903, 15288-15864, 15304-15570, 15304-15672,1 5306-15599, 15315-16021, 15319-15739, 15335-16095, 15342-15594, 15342-15998, 15409-16034, 15414-15712, 15425-16067, 15441-15651, 15448-15612, 15450-15657, 15452-15841, 15455-15958, 15455-15964, 15474-16087, 15478-16159, 15485-15724, 15485-15871, 15512-16106, 15515-16130, 15515-16159, 15519-15776, 15519-15787, 15519-15905, 15519-16159, 15520-15784, 15520-16076, 15526-15686, 15553-16101, 15560-16159, 15568-15672 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 31/6301177CB1/1125 1-502, 108-549, 137-179, 140-179, 158-179, 317-369, 317-400, 317-492, 317-520, 317-1018, 318-490, 318-494, 318- 499, 318-503, 318-506, 318-510, 318-513, 318-517, 318-522, 318-525, 318-533, 318-536, 318-542, 318-549, 318- 567, 318-571, 318-576, 318-580, 318-636, 318-640, 318-726, 318-735, 318-787, 318-797, 318-812, 318-849, 318- 857, 318-885, 318-894, 318-902, 318-943, 318-968, 318-975, 318-978, 318-982, 318-998, 318-999, 318-1000, 318- 1009, 318-1017, 318-1018, 318-1023, 320-521, 322-608, 328-562, 335-741, 340-1006, 354-600, 369-1110, 379-637, 435-1095, 453-744, 464-1017, 471-1101, 493-775, 502-705, 503-1119, 504-1017, 508-1019, 511-991, 518-697, 562- 711, 562-1078, 564-904, 564-1019, 568-1019, 582-1111, 597-1084, 616-859, 677-898, 707-893, 707-898, 769-1125, 901-1017, 928-1019, 930-1019, 1015-1065, 1015-1120, 1015-1124, 1015-1125 32/257833CB1/2119 1-2119, 70-256 33/7580043CB1/2279 1-2150, 1026-1555, 1026-1716, 1026-1722, 1757-2279 34/8120340CB1/2690 1-282, 10-225, 19-225, 35-454, 39-251, 44-868, 147-441, 147-566, 147-655, 153-752, 153-806, 251-883, 256-677, 599-990, 763-1459, 836-1007, 853-1007, 854-1007, 856-1007, 1181-1785, 1181-1992, 1238-1455, 1238-1501, 1238- 1680, 1238-1787, 1271-1895, 1289-1565, 1289-1880, 1324-1815, 1376-2044, 1391-1895, 1417-1694, 1430-1474, 1430-1475, 1461-1909, 1479-1826, 1480-1506, 1480-1525, 1510-2293, 1525-2306, 1629-2460, 1750-2690, 1777- 2408, 1779-2045, 1779-2267, 1804-1988 35/7475307CB1/933 1-933, 47-915 36/7475243CB1/1002 1-1002 37/7490257CB1/1132 1-219, 184-1101, 1032-1132 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 38/793400CB1/3386 1-630, 21-522, 21-579, 21-790, 44-632, 64-529, 342-642, 617-848, 617-995, 617-1007, 911-1469, 1018-1288, 1087- 1642, 1248-1543, 1426-1998, 1426-2030, 1442-1763, 1516-1988, 1516-1994, 1532-2172, 1634-1883, 1665-2023, 1683-1993, 1684-1947, 1715-2310, 1726-1968, 1727-1975, 1727-1979, 1727-2202, 1741-2008, 1748-2314, 1760- 2023, 1827-2294, 1946-2159, 2119-2377, 2198-2492, 2323-2592, 2343-2516, 2421-2678, 2447-2692, 2447-2906, 2516-2809, 2528-3171, 2635-2903, 2648-3129, 2650-3303, 2666-2930, 2671-2922, 2671-3269, 2775-3061, 2865- 3309, 2879-3386, 2924-3121, 2926-3302, 2930-3298, 3045-3302 39/8210895CB1/1165 1-86, 1-182, 1-256, 3-119, 8-679, 205-240, 385-961, 385-1014, 392-633, 397-850, 453-722, 524-744, 531-767, 541- 1165, 547-814, 550-1069 40/55069745CB1/1101 1-1101, 297-501, 297-502, 309-767, 309-1079 41/3212783CB1/3156 1-269, 1-694, 22-269, 183-269, 303-757, 303-772, 303-931, 303-961, 303-1225, 419-703, 419-864, 419-881, 453- 1099, 553-1061, 561-881, 561-1138, 594-2938, 670-881, 672-881, 672-1162, 675-867, 689-1152, 703-881, 705- 1189, 774-1134, 992-1487, 994-1594, 998-1452, 1129-1311, 1309-1751, 1309-1789, 1309-1804, 1309-1889, 1353- 1580, 1353-1817, 1357-1645, 1365-1759, 1394-1676, 1420-1465, 1427-1660, 1812-1843, 1843-2012, 1843-2459, 1902-2208, 1903-2061, 1942-2159, 1967-2270, 2117-2378, 2117-2789, 2181-2446, 2210-2437, 2228-2497, 2290- 2635, 2290-2791, 2291-2562, 2324-2590, 2338-2563, 2345-2613, 2347-2617, 2411-2633, 2480-3156, 2482-2731, 2578-2831, 2578-2862, 2600-2850, 2612-2851, 2615-2872, 2630-2870, 2654-2769, 2662-3156, 2704-2939, 2738- 3016, 2804-3156, 2862-3123 42/6986529CB1/4450 1-444, 1-542, 406-700, 406-949, 406-958, 406-1039, 440-1182, 460-1182, 605-4450, 765-1518, 843-1197, 1111- 1689, 1210-1832, 1445-2090, 1665-2292, 1847-2237, 2061-2554, 2092-2467, 2166-2855, 2209-2602, 2457-2824, 2522-2933, 2678-3308 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 43/7474928CB1/1865 1-444, 1-492, 1-497, 1-518, 1-535, 1-539, 1-540, 1-547, 1-564, 1-565, 1-574, 1-580, 1-615, 1-618, 1-645, 1-647, 1- 655, 1-657, 1-682, 1-704, 1-787, 6-787, 138-425, 138-426, 138-781, 143-781, 146-781, 159-781, 164-413, 169-781, 170-781, 187-457, 210-781, 216-781, 271-781, 272-781, 278-781, 279-681, 285-741, 293-789, 306-781, 313-629, 313-781, 313-784, 313-785, 313-787, 314-790, 340-781, 353-764, 355-790, 359-790, 386-649, 387-764, 406-784, 406-789, 490-1024, 516-761, 552-781, 557-787, 784-1295, 909-1122, 981-1792, 1086-1338, 1104-1805, 1127-1358, 1141-1822, 1188-1409, 1188-1687, 1188-1758, 1276-1539, 1374-1611, 1384-1636, 1384-1829, 1386-1852, 1416- 1865, 1426-1865, 1446-1863, 1459-1842, 1462-1742, 1516-1865, 1520-1862, 1528-1779, 1528-1793, 1557-1823, 1557-1824, 1557-1865, 1626-1865, 1690-1865, 1754-1865 44/3736039CB1/798 1-231, 1-491, 17-42, 40-245, 40-254, 43-338, 45-312, 47-331, 51-295, 51-328, 51-355, 53-286, 53-343, 54-146, 54- 277, 55-264, 56-169, 56-224, 56-305, 56-322, 56-329, 56-481, 57-343, 58-282, 58-324, 58-371, 58-376, 60-584, 61- 303, 62-209, 62-381, 62-616, 63-345, 64-300, 65-146, 68-392, 69-376, 70-487, 71-229, 84-341, 84-507, 88-335, 88- 339, 92-290, 95-798, 98-528, 114-390, 114-752, 131-200, 141-382, 146-436, 153-399, 161-427, 200-257, 200-385, 200-610, 209-532, 228-433, 309-604, 325-582, 415-757, 415-794, 415-797 45/1798572CB1/1785 1-599, 15-212, 15-778, 21-281, 21-283, 21-656, 24-603, 24-625, 25-163, 25-522, 25-544, 25-585, 25-596, 25-632, 25-697, 25-703, 25-715, 25-738, 25-783, 25-789, 25-808, 25-844, 26-619, 33-1785, 35-697, 39-654, 41-714, 48-636, 48-652, 48-665, 48-669, 48-759, 255-442, 255-719, 333-688, 368-575, 587-866, 649-947, 716-976, 719-841, 730- 974, 840-1151, 854-1334, 901-993, 938-1202, 938-1218, 966-1222, 977-1238, 1002-1441, 1054-1326, 1064-1610, 1075-1337, 1078-1127, 1103-1534, 1103-1547, 1170-1396, 1200-1407, 1241-1783, 1254-1496, 1289-1785, 1302- 1785, 1303-1785, 1305-1784, 1306-1785, 1309-1785, 1310-1785, 1315-1557, 1316-1776, 1319-1785, 1345-1438, 1347-1775, 1360-1785, 1400-1785, 1403-1677, 1403-1785, 1415-1776, 1433-1785, 1466-1785, 1551-1785, 1560- 1783, 1560-1785, 1564-1783, 1564-1785, 1570-1785, 46/3038391CB1/1167 1-257, 1-371, 133-961, 134-432, 316-1151, 544-784, 690-1166, 704-1167, 713-1165, 731-1162, 733-1162, 851- 1166, 947-1143 47/5822287CB1/3431 1-604, 1-3388,1 5-603, 15-658, 36-313, 36-368, 36-380, 37-380, 49-286, 76-357, 76-358, 76-359, 76-360, 177-504, 338-605, 355-1047, 707-859, 767-1319, 791-942, 1412-1657, 1412-2121, 1414-1770, 1483-2058,1 567-1851, 1737- 2136, 1777-1933, 1781-2039, 1860-2073, 2079-2550, 2116-2533, 2142-2240, 2170-2551, 2319-2543, 2416-2543, 2417-2543, 2720-3220, 2747-3199, 2747-3220, 2747-3314, 2766-3107, 2807-3066, 2809-3220, 2858-3431, 2882- 3179, 2908-3181, 2908-3220, 2909-3033, 2972-3168 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 48/7231805CB1/2947 1-472, 9-496, 9-2947, 15-233, 20-591, 22-313, 22-502, 24-247, 24-638, 25-473, 28-319, 28-624, 28-827, 30-625, 32- 652, 34-654, 39-702, 41-545, 51-204, 89-752, 90-750, 92-374, 100-473, 100-665, 100-966, 167-473, 395-997, 464- 1084, 505-1162, 508-1124, 571-1077, 573-1162, 578-1122, 617-1279, 665-1053, 683-1210, 684-1210, 702-1326, 751-1062, 780-1457, 782-1394, 858-1127, 941-1482, 953-1435, 968-1164, 974-1563, 1004-1061, 1121-1618, 1121- 1638, 1121-1659, 1121-1697, 1121-1721, 1121-1749, 1121-1762, 1121-1769, 1121-1775, 1121-1827, 1121-1994, 1124-1757, 1124-1817, 1128-1620, 1149-1450, 1149-1898, 1150-1347, 1162-1957, 1165-1798, 1174-1655, 1174- 1841, 1183-1927, 1199-1557, 1218-1917, 1223-2020, 1262-1444, 1283-1937, 1283-1955, 1306-1594, 1307-1917, 1311-2002, 1320-1864, 1320-1870, 1327-1780, 1327-1848, 1329-1779, 1333-1998, 1353-2056, 1356-1938, 1378-2030, 1382-1757, 1383-2092, 1391- 1557, 1397-1663, 1401-1520, 1401-1897, 1401-1962, 1401-1983, 1401-1989, 1402-1885, 1402-1913, 1427-1668, 1431-2069, 1433-2244, 1436-2057, 1440-2119, 1440-2152, 1457-2163, 1477-2063, 1485-2087, 1488-2291, 1521- 2229, 1523-2066, 1526-1734, 1539-2124, 1546-2139, 1552-2146, 1553-2271, 1573-1941, 1579-1983, 1584-1875, 1593-2138, 1600-2210, 1604-1972, 1604-2266, 1611-1865, 1613-1874, 1620-2265, 1622-1858, 1622-2268, 1628- 2205, 1636-2124, 1636-2362, 1655-2332, 1662-1876, 1667-2308, 1680-1772, 1688-2033, 1704-2265, 1705-2329, 1734-2106, 1793-2006, 1804-2410, 1871-2573, 1960-2594, 1989-2666, 2049-2718, 2243-2947 49/4032542CB1/8531 1-617, 1-660, 43-723, 94-699, 183-830, 481-767, 655-937, 692-986, 896-1630, 1029-1810, 1059-1580, 1087-1638, 1087-1639, 1231-1799, 1232-1799, 1535-2150, 1535-2353, 1535-2368, 2535-2371, 1535-2404, 1535-2415, 1539- 2389, 1543-2344, 1595-2470, 1634-2087, 1640-2470, 1679-2470, 1798-2472, 1843-2471, 1851-2470, 1866-2534, 1871-2470, 2078-2353, 2144-2413, 2164-2654, 2179-2435, 2213-2460, 2213-2853, 2375-2669, 2414-2822, 2600- 2853, 2606-3012, 2676-3441, 2685-3171, 2732-2997, 2808-3094, 2808-3212, 2808-3221, 2808-3252, 2808-3259, 2808-3315, 2808-3350, 2808-3381, 2808-3409, 2808-3410, 2810-3385, 2814-3394, 2843-3457, 2958-3477, 2963-3522, 2997-3469, 3008-3598, 3035-3323, 3104- 3497, 3540-3948, 3561-4138, 3679-3925, 3874-4523, 3898-4252, 3909-4432, 3945-4420, 4255-4883, 4307-4945, 4469-4974, 4469-5063, 4510-4758, 4557-5046, 4563-5196, 4565-4967, 4596-4934, 4723-5204, 4792-5393, 5024- 5158, 5036-5699, 5156-5610, 5195-5494, 5195-5835, 5214-5834, 5349-5622, 5365-5905, 5394-5645, 5502-6111, 5506-6208, 5517-6182, 5535-5932, 5544-6246, 5567-5882, 5588-6088, 5595-6277, 5614-6247, 5825-6081, 5861- 6562, 5877-6550, 5911-6403, 5953-6202, 6049-6286, 6072-6483, 6077-6358, 6158-6690, 6248-6525, 6248-6653, 6256-6501, 6465-7000, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 49 6481-6856, 6513-7216, 6541-6953, 6610-7289, 6662-7176, 6675-7404, 6747-7023, 6954-7218, 6981-7269, 6992- 7242, 6992-7544, 7072-7510, 7204-7708, 7205-7474, 7209-7456, 7257-7695, 7258-7498, 7259-7822, 7273-7823, 7305-7849, 7314-7878, 7367-7930, 7367-8088, 7385-7683, 7396-8136, 7401-7599, 7413-7787, 7426-7824, 7493- 7746, 7510-8202, 7537-8206, 7546-8061, 7552-8150, 7555-8254, 7578-8288, 7586-7845, 7586-7934, 7586-8125, 7614-8130, 7637-7873, 7674-7934, 7688-8398, 7709-8140, 7709-8435, 7712-8142, 7717-8376, 7718-8427, 7727- 7982, 7727-8384, 7736-7993, 7756-8297, 7789-8085, 7790-8399, 7808-8035, 7812-8436, 7815-8421, 7820-8273, 7825-8437, 7827-8448, 7829-8419, 7833-8396, 7841-8369, 7843-8447, 7880-8304, 7880-8440, 7893-8380, 7895- 8448, 7903-8148, 7904-8531, 7918-8429, 7928-7956, 7928-8448, 7949-8419, 7963-8205, 7963-8229, 7977-8268, 7982-8449, 7985-8010, 7992-8448, 7994-8432, 8029-8448, 8037-8445, 8069-8447, 8072-8448, 8159-8447, 8205- 8447 50/1593941CB1/9375 1-281, 49-141, 74-357, 101-590, 113-327, 163-442, 164-430, 298-707, 345-843, 375-848, 389-875, 402-780, 414- 929, 594-831, 596-1006, 746-1046, 781-1003, 781-1326, 803-1437, 913-1362, 968-1608, 1041-1450, 1103-1787, 1138-1692, 1141-1676, 1215-1912, 1350-1631, 1453-2102, 1574-2199, 1585-1946, 1608-2212, 1726-2218, 1728- 2001, 1850-2328, 1850-2550, 1926-2196, 1926-2239, 1946-2487, 1947-2621, 2049-2306, 2049-2508, 2049-2582, 2050-2666, 2051-2169, 2093-2623, 2233-2637, 2286-2826, 2309-2701, 2322-3015, 2337-2822, 2411-2949, 2423- 2686, 2448-2996, 2508-2996, 2530-2996, 2534-2996, 2550-2991, 2559-2996, 2573-2972, 2576-2996, 2582-2996, 2588-2996, 2595-2996, 2596-2991, 2602-3056, 2617-2996, 2622-3126, 2628-2996, 2633-3355, 2738-2996, 2768-2996, 2776-2996, 2816-2996, 3181-3744, 3181- 3745, 3194-3650, 3246-3794, 3295-3862, 3312-4066, 3495-4058, 3735-4299, 3907-4491, 3940-4385, 4058-4708, 4089-4708, 4095-4799, 4130-4670, 4411-5075, 4422-4902, 4531-5080, 4602-5128, 4602-5214, 4602-5234, 4602- 5241, 4602-5258, 4602-5267, 4602-5277, 4602-5286, 4602-5298, 4602-5343, 4602-5354, 4602-5370, 4602-5393, 4603-5192, 4603-5251, 4606-5290, 4630-5412, 4704-5302, 4706-5364, 4716-5152, 4716-5318, 4717-5150, 4742- 5586, 4743-5211, 4784-5630, 4785-5406, 4830-5588, 4831-5447, 4872-5624, 4872-5630, 4933-5480, 4934-5447, 4947-5586, 4954-5686, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 50 4981-5499, 5077-5630, 514-5737, 5220-5755, 5410-6113, 5529-6268, 5554-6203, 5571-6232, 5633-6195, 5707- 6185, 5735-6420, 5737-6574, 5748-6267, 5765-6234, 5837-6372, 5842-6324, 5846-6733, 5929-6548, 5948-6611, 6018-6529, 6039-6591, 6045-6584, 6048-6655, 6071-6584, 6094-6639, 6142-6640, 6192-6690, 6209-6684, 6221- 6844, 6244-6684, 6244-6905, 6251-6684, 6313-6906, 6313-7097, 6328-6905, 6358-6885, 6360-6906, 6399-6921, 6400-6984, 6430-6989, 6477-7025, 6576-7075, 6582-7058, 6645-7354, 6697-7302, 6700-7346, 6702-7345, 6703- 7252, 6766-7247, 6900-7522, 6910-7475, 6910-7563, 6941-7583, 6956-7542, 6988-7620, 7065-7506, 7103-7685, 7147-7681, 7150-7727, 7153-7827, 7217-7814, 7244-7826, 7255-7852, 7258-7910, 7260-7771, 7260-7906, 7260- 8043, 7260-8089, 7260-8098, 7260-8168, 7261-7820, 7261-8092, 7273-8093, 7295-7853, 7324-7875, 7331-7999, 7335-7997, 7335-8193, 7335-8318, 7351-7928, 7354-8026, 7374-7988, 7405-7929, 7443-8026, 7475-8080, 7477-7931, 7544-8074, 7554-8110, 7564- 8097, 7573-8181, 7585-8042, 7585-8068, 7585-8129, 7599-8354, 7605-8093, 7608-8097, 7611-8096, 7626-8251, 7631-8093, 7632-8100, 7650-8097, 7652-8098, 7662-8097, 7667-8097, 7705-8341, 7755-8265, 7761-8435, 7761- 8537, 7777-8522, 7778-8606, 7779-8522, 7782-8268, 7808-8550, 7816-8383, 7829-8303, 7839-8297, 7848-8405, 7895-8455, 7900-8463, 7940-8517, 7957-8592, 7968-8549, 7970-8612, 7976-8616, 7980-8618, 8019-8663, 8032- 8558, 8038-8544, 8039-8533, 8049-8622, 8072-8622, 8073-8559, 8076-8637, 8097-8601, 8117-8747, 8132-9004, 8136-8605, 8142-8607, 8144-8607, 8157-8605, 8160-8605, 8173-8717, 8191-8788, 8307-8916, 8431-9307, 8450- 9078, 8474-8983, 8475-8905, 8648-9358, 8668-9234, 8686-9289, 8709-9361, 8752-9330, 8843-9361, 8896-9361, 8904-9361, 8913-9358, 8935-9375, 8939-9368 51/3471414cb1/1146 1-252, 1-495, 1-514, 1-579, 1-607, 50-533, 90-703, 93-810, 110-641, 132-450, 141-711, 218-891, 290-555, 310-814, 360-1104, 393-917, 412-1146, 493-1072, 523-1146, 557-1057, 625-1102, 650-1060, 774-1083, 792-1124 52/7504960CB1/790 1-232, 1-750, 66-350, 67-514, 67-528, 67-529, 67-546, 67-561, 67-590, 67-641, 67-665, 67-677, 67-687, 67-693, 91- 714, 181-679, 237-466, 252-688, 257-527, 259-734, 262-744, 264-400, 264-665, 274-790, 277-539, 284-740, 289- 531, 289-744, 293-623, 309-543, 309-559, 333-582, 334-586, 335-658, 339-557, 339-721, 340-627, 388-633, 389- 644, 393-663, 398-639, 402-729, 403-645, 405-682, 408-682, 430-701, 448-706, 451-683, 459-729, 462-707, 483- 724, 637-717 Table 5 Polynucleotide SEQ Incyte Project ID: Representative Library ID NO: 27 3015053CB1 BRAITUT26 28 7481761CB1 SPLNDIC01 29 942253CB1 BRSTTUT02 30 1506342CB1 LUNGNON07 31 6301177CB1 BRSTTUT03 33 7580043CB1 BRAIFEC01 34 8120340CB1 THYRNOT03 38 793400CB1 BRAINOT03 39 8210895CB1 LUNGTUT06 40 55069745CB1 GPCRDRV20 41 3212783CB1 ESOGTUT02 42 6986529CB1 BRAIFER05 43 7474928CB1 LIVRTUN04 44 3736039CB1 PROSNOT06 45 1798572CB1 THYRNOT02 46 3038391CB1 TESTNOT11 47 5822287CB1 PROSTUS23 48 7231805CB1 LIVRTUE01 49 4032542CB1 LUNGNOT10 50 1593941CB1 TONSDIE01 51 3471414CB1 LEUKNOT02 52 7504960CB1 LUNGNOT02 Table 6 Library Verctor Library Cescription BRAIFEC01 pINCY This large size-fractionated library was constructed using RNA isolated from brain tissue removed from a Caucasian male fetus who was stillborn with a hypoplastic left heart at 23 weeks' gestation. BRAIFER05 pINCY Library was constructed using RNA isolated from brain tissue removed from a Caucasian male fetus who was stillborn with a hypoplastic left heart at 23 weeks' gestation. BRAINOT03 PSPORT1 Library was constructed using RNA isolated from brain tissue removed from a 26-year-old Caucasian male during cranioplasty and excision of a cerebral meningeal lesion. Pathology for the associated tumor tissue indicated a grade 4 oligoastrocytoma in the right fronto-parietal part of the brain. BRAITUT26 pINCY Library was constructed using RNA isolated from brain tumor tissue removed from the right posterior fossa, occipital convexity of a 70-year-old Caucasian male during cerebral meninges lesion excision. pathology indicated meningioma. Patient history included a benign colon neoplasm and unspecified personality disorder. Family history included chronic proliferative nephritis, acute myocardial infarction, atherosclerotic coronary artery disease, and chronic proliferative nephritis. BRSTTUT02 PSPORT1 Library was constructed using RNA isolated from breast tumor tissue removed from a 54-year-old Caucasian female during a bilateral radical mastectomy with reconstruction. Pathology indicated residual invasive grade 3 mammary ductal adenocarcinoma. The remaining breast parenchyma exhibited proliferative fibrocystic changes without atypia. One of 10 axillary lymph nodes had metastatic tumor as a microscopic intranodal focus. Patient history included kidney infection and condyloma acuminatum. Family history included benign hypertension, hyperlipidemia, and a malignant colon neoplasm. BRSTTUT03 PSPORT1 Library was constructed using RNA isolated from breast tumor tissue removed from a 58-year-old Caucasian female during a unilateral extended simple mastectomy. Pathology indicated multicentric invasive grade 4 lobular carcinoma. The mass was identified in the upper oute quadrant, and three separate nodules were found in the lower outer quadrant of the left breast. Patient history included skin cancer, rheumatic heart disease, osteoarthritis, and tuberculosis. Family history included cerebrovascular disease, coronary artery aneurysm, breast cancer, prostate cancer, atherosclerotic coronary artery disease, and type I diabetes. ESOGTUT02 pINCY Library was constructed using RNA isolated from esophageal tumor tisue obtained from a 61-year-old Caucasian male during a partial esophagectomy, proximal gastrectomy, pyloromyotomy, and rgional lymph node excision. Pathology indicated an invasive grade 3 adenocarcinoma in the esophagus. Family history included atherosclerotic coronary artery disease, type II diabetes, chronic liver disease, primary cardiomyopathy, benign hypertension, and cerebrovascular disease.

Table 6 Library Vector Library Description GPCRDRV20 PCR2-TOPOTA Library was constructed using pooled cDNA from different donors. cDNA was generated using mRNA isolated from the following: aorta, para-aortic soft tissue, fental femur, unterated epidermal keratinocytes, neck muscle, supraglottic soft tissue, calf muscle, retroperitoneal soft tissue, sacral bone giant cell tumor, treated breast skin fibroblast cells, abdominal skin, untreated T-lymphocyte cell line (Jurkat cell line), fetal smjall intestine, fetal colon, colon tumor (grade 3 colonic adenocarcinoma) small intestine, colon, ascending colon, diseased descending colon tissue (chronic ulcerative colitis, moderate to severe), cecal tumor (grade 1 neuroendocrine carcinoma), diseased ileum tissue (Crohn's disease), diseased small intestine (focal reactive foveolar hyperplasia consistent iwth bile reflux), ascending colon, fetal stomach, diseased gallbladder (moderate chronic cholecystitis and cholelithiasis), esophagus, diseased gallblader (acute hemorrhagic cholecystitris with cholelithiasis), esophagus tumor (invasive grade 3 adenocarcinoma), stomach, diseased gallbladder (chronic cholecystitis and cholelithiasis), diseased gallbladder (acute necrotizing cholecystitis with cholelithiasis (clinically hydrops), endometrium, diseased cervix tissue (mild chronic cervicitis with focal squamous metaplasia), uterus tumor (leiomyoma), diseased ovary (polycystic ovarian disease), myometrium, uterus, endometrial tumor (grade 3 adenosquamous carcinoma) ovary, fetal penis, testis, untreated prostate epithelial cells (PrEC Cells), testicle tumor (embryonal carcinoma), seminal vesicle, diseased prostate (adenofibromatous hyperplasia), fetal spleen, spleen, thymus, diseased tonsil tissue (reactive lymphoid hyperplasia). from diseased spleen (idiopatic thrombocytopenic purpura), spleen tumor (malignant lymphoma, diffuse large cell type, B-cell phenotype with abundant reactive T-cells), thymus, diseased tonsil tissue (lymphoid hyperplasia), pelvic lymph node (matched with Hodgkin's disease, nodular sclerosing type), a treated chronic myelogenous leukemia precursor cell line (K562 Cells), axillary lymph node tumor (metastatic adenocarcinoma), fetal liver, fetal pancreas, pancreas, liver tumor (metastatic grade 2 (of 4) neuroendocrine carcinoma), fetal kidney, renal pyramid, kidney tumor 9renal cell carcinoma, clear cell type), diseased kidney tissue (chronic interstitial nephritis), ureter tumor (transitional cell carcinoma), kidney cortex, ureter tumor (invasive grade 3 (of 3) transitional cell carcinoma), pooled lung, adrenal gland, benign parotid tumor (sebaceous lymphadenoma), parotid, thyroid, diseased thyroid (adenomatous hyperplasia), diseased breast (proliferative fibrocystic changes), breast, submandibular gland, adrenal tumor (pheochromocytoma), and hyperplastic parathyroid. LEUKNOT02 pINCY Library was constructed using RNA isolated from white blood cells of a 45-year-old female with blood type O+. The donor tested positive for cytomegalovirus (CMV).

Table 6 Library Vector Library Description LIVRTUE01 PCDNA2.1 This 5' biased random primed library was constructed using RNA isolated from liver tumor tissue removed from a 72-year- old Caucasian male during partial hepatectomy. Pathology indicated metastatic grade 2 (of 4) neuroendocrine carcinoma forming a mass. The patient presented with metastatic liver cancer. Patient history included benign hypertension, type I diabetes, prostatic hyperplasia, prostate cancer, alcohol abuse in remission, and tobacco abuse in remission. Previous surgeries included destruction of a pancreatic lesion, closed prostatic biopsy, transurethral prostatectomy, removal of bilateral testes and total splenectomy. Patient medications included Eulexin, Hytrin, Procar, Ecotrin, and insulin. Family history included atherosclerotic coronary artery disease and acute myocardial infarction in the mother; atherosclerotic coronary artery disease and type II diabetes in th father. LIVRTUN04 pINCY This normalized liver tumor cell line library was constructed from 1.72 million independent clones from a hepatocyte library. Starting RNA was isolasted from an untreated C3A hepatocyte cell linr, which is a derivative of a hepatoblastoma removed from a 15-year-old Caucasian male. The library was normalized in two rounds using conditions adapted from Soares et al., PNAS (1994) 91:9228-9232 and Bonaldo et al., Genome Research (1996) 6:791, (except that a significantly longer (48 -hours/round) reannealing hybridization was used. LUNGNON07 pINCY This normalized lung tissue library was constructed from 5.1 million independent clones from a lung tissue library. Starting RNA was made from RNA isolated from lung tissue. The library was cnormalized in two rounds using conditions adapted from Soares et al., PNAS (1994) 91:9228-9232 and Bonaldo et al., Genome reseasrch (1996) 6:791, except that a significantly longer (48 hours/round) reannealing hybridization was used. LUNGNOT02 PBLUESCRIPT Library was constructed using RNA isolated from the lung tissue of a 47-year-old Caucasian male, who died of a subarachnoid hemorrhage. LUNGNOT10 pINCY Library was constructed using RNA isolated from the lung tissue of a Caucasian male fetus, who died at 23 weeks' gestation. LUNGTUT06 pINCY Library was constructed using RNA isolated from apical lung tumor tissue removed from an 80-year-old Caucasian female during a segment lung resection. Pathology lindicated a metastatic granulosa cell tumor. Patient history included pelvic soft tissue tumor and chemotherapy for one yar. Family history included tuberculosis, lung cancer, atherosclerotic coronary artery disease. PROSNOT06 PSPORT1 Library was constructed using RNA isolated from the diseased prostate tissue of a 57-year-old Caucasian male during radical prostatectomy, removal of both testes and excision of regional lymph nodes. Pathology indicated adenofibromatous hyperplasia. Pathology for the associated tumor tissue indicated adenocarcinoma (Gleason grade 3+3). Patient history included a benign neoplasm of the large bowel and type I diabetes. Family history included a malignant neoplasm of the prostate and type I diabetes.

Table 6 Library Vector Library Description PROSTUS23 pINCY This subtracted prostate tumor library was constructed using 10 million clons from a pooled prostate tumor library that was subjected to 2 rouds of subtractive hybridization with 10 million clones from a pooled prostate tissue library. The starting library for subtraction wasconstructed by pooling equal numbers of clones from 4 prostate tumor libraries using mRNA isolated from prostate tumor removed from Caucasian males at ages 58 (A), 61 (B), 66 (C), and 68 (D) during prostatectomy with lymph node excision. Pathology indicated adenocarcinoma in all donors. History included elevated PSA, induration and tobacco abuse in donor A; elevated PSA, indration, prostate hyperplasia, renal failure, osteoarthiritis, renal artery stenosis, benign HTN, thrombocytopenia, hyperlipidemia, tobacco/alcohol abuse and hepatitis C (carier) in donor B; elevated PSA, induration, and tobacco abuse in donor C; and elevated PSA, induration, hypercholesterolemia, and idney calculus in donor D. The hybridization probe for subtraction was constructed by pooling equal numbers of cDNA clones from 3 prostate tissue libraries derived from prostate tissue, prostate epithelial cells, and fibroblasts from prostate stroma from 3 different donors. Subtractive hybridization conditions were based on the methodologies of Swaroop et al., DNAR 19 (1991):1954 and Bonaldo, et al. Genome Research 6 (1996):791. SPLNDIC01 pINCY This large size-fractionated library was constructed using pooled cDNA from two differenti donors. cDNA was generated using mRNA isolated from spleen tissue reoved from an 8-year-old Black male (donor A) who died from anoxia and from diseased spleen tissue removed from a 14-year-old Asian male (donor B) during a total splenectomy. Pathology fro donor B indicated changes consistent with idiopathic thrombocytopenic purpura. Serologies were negative for donor A. Donor B presented with bruising. Patient medications included DDAVP, Versed, labetalol (donor A), and Vincristine (donor B). TESTNOT11 pINCY Library was constructed using RNA isolated from testicular tissue removed from a 16-year-old Caucasian male who died from hanging. Patient history included drug use (tobacco, marijuana, and cocaine use), and medications included Lithium, Ritalin, and Paxil. THYRNOT02 PSPORT1 Library was constructed using RNA isolated from the diseased thyroid tissue of a 6-year-old Caucasian female with Graves' disease (hyperthyrodidism). THYRNOT03 pINCY Library was constructed using rNA isolated from thyroid tissue removed from the left thyroid of a 28-year-old Caucasian female during a complete thyroidectomy. Pathology indicated a small nodule of adenomatous hyperplasia present in the left thyroid. Pathology for the associated tumor tissue indicated dominant follicular adenoma, forming a well-encapsulated mass in the left thyroid.

Table 6 Library Vector Library Description TONSDIE01 PCDNA2.1 This 5' biased random primed library was constructed using rNA isolated from diseased left tonsil tissue removed from a 6 year-old Caucasian male during adenotonsillectomy. Pathology indicated reactive lymphoid hyperplasia, bilaterally. The patient presented with sleep apnea. Patient history included a bacterial infection. Previous surgeries included myringotomy wth tube insertion. The patient was not taking any medications. Family history inlcuded benign hypertension, myocardial infarction, and atherosclerotic coronry artery disease in the grandparent(s).

Table 7<BR> Program Description Reference Parameter Threshold<BR> ABI FACTURA A program that removes vector sequences and Applied Biosystems, Foster City, CA.<BR> masks ambiguous based in nucleic acie seuences.<BR> <P>ABI/PARACEL FDF A Fast Data Finder useful in comparing and Applied biosystems, Foster City, CA; Mismatch <50%<BR> annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA.<BR> <P>ABI AutoAssembler A program that assembles nucleic acid sequences. Applied Biosystems, Foster City, CA.<BR> <P>BLAST A Basic Local Alignment Seasrch Tool usedful in Altschul, S.F. et al. (1990) J. Mol. Biol. ESTs: Probability value=1.0E-8<BR> aequence similarity search for amino acid and 215:403-410; Altschul, S.F. et al. (1997) or less<BR> nucleic acid sequences. BLAST includes five Nucleic Acids Res. 25:3389-3402. Full Length sequences; Probability<BR> functions: blastp, blastn, blastx, tblastn, and tblastx. value = 1.0E-10 or less<BR> FASTA A Pearson and Lidpdman algorithm that searches for Pearson, W.R. and D.J. Lipman (1988) Proc. ESTs: fasta E value=1.06E-6<BR> similarity between a query sequence and a group fo Natl. Acad Sci. USA 85:2444-2448; Pearson, Assembled ESTs: fasta Identity=<BR> sequences of the same type. FAST$A comprises as W.R. (1990) Methods Enzymol. 183:63-98; 95% or greater and<BR> least five functions: fasta, tfasta, fastx, tfastx, and and Smith, T.F. and M. S. waterman (1981) Match length=200 based or greater;<BR> ssearch. Adv. Appl. Math. 2:482-489. fastx E value=1.0E-8 or less<BR> Full Length sequences:<BR> fastx score=100 or greaster<BR> BLIMPS A BLocks IMProved Searcher that matches a Henikoff, S. and J.G. Henikoff (1991) Nucleic Probability value=1.0E-3 or less<BR> sequence against those in BLOCKS, PRINTS, Acids Res. 19:6565-6572; Henikoff, J.G. and<BR> DOMO, PRODOM, and PFAM databases to search S. Henikoff (1996) Methods Enzymol.<BR> for gene families, sequence homology, and structural 266:88-105; and Attwood, T.K. et al. (1997) J.<BR> fingerprint regions. Chem., Inf. Comput. Sci. 37:417-424.<BR> <P>HMMER An algorithm for searching a query sequence against Krogh, A. et al. (1994) J. Mol. Biol. PFAM, INCY, SMART, or TIGRFAM<BR> hidden markov model (HMM)-based databases of 235;1501-1531; Sonnhammer, E.L.L. et al. hits: Probability value=1.0E-3 or less<BR> protein family consensus sequences, such as dPFAM, (1988) Nucleic Acids Res. 26:320-322; Signal peptide hits: Score= 0 or<BR> INCY, SMART, and TIGRFAM. Durbin, R. et al. (1998) Our World View, in agreater<BR> Nutshell, Cambridge Univ. Press, pp. 1-350.

Table 7 (cont.)<BR> Program Descritpion Reference Parameter Threshold<BR> ProfileScan an algorithm that searches for structural and sequence Gribskov, M. et al. (1988) CABOS 4:61-66; Normalized quality score#GCG-<BR> motifs in protein sequences that msatch sequence patterns gribskov, M. et al.d (1989) Metods Enzuymol. specified "HIGH" vlaue for that<BR> defined in Prosite. 183:146-159; Bairochm, A. et al. (1997) prticular Prosite motif.<BR> <P>Nucleic Acids Res. 25:217-221. Generally, score=1.4-2.1.<BR> dPhred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res.<BR> sequencesr traces with high sensitivity and probability. 8:175-185; Ewing, B. and P. Green<BR> (1998) Genome Res. 8:186-194.<BR> <P>Phrap A Phils Revised Assembly Program including SWAT and Smith, T.F. and M.S. Wasterman (1981) Adv. Score=12-0 or greaster;<BR> CrossMatch, programs based on efficient implementation Appl. Math. 2:482-489; Smith, t.F. and M.S. Match length=56 or greater<BR> of the Smith-Waterman algorithm, useful in searching Waterman (1981) J. Mol. Biol. 147:195-197;<BR> sequence homology and assembling DNA sequences. and Green, P., University of Washington,<BR> Seattle, WA.<BR> <P>Consed A graphical tool for viewing and editing Phrap assemblies. Phrap assemblies. Gordon, D. et al. (1998) Genome Res. 8:195-202.<BR> <P>SPscan A weight matrix analysis program that scans protein Nielson, H. et al. (1997) Protein Engineering Score=3.5 or greaster<BR> sequences for the presence of secretory signal peptides.d 10:1-6; Claverie, J.M. and S. Audic (1997)<BR> CABIOS 12:431-439.<BR> <P>TMAP A program that uses weight matrices to delineate Persson, B. and P. Argos (1994) J. Mol. Biol.<BR> transmembrane segments on protein sequences and 237:182-192; Persson, B. and P,. Argos (1996)<BR> determine orientation. Protein Sci. 5:363-371.<BR> <P>TMHMMER A program that uses a hidden Markov model (HMM) to Sodnnhammer, E.L. et al. (1998) Proc. Sixth Intl.<BR> delineaste transmembrane segments on protein sequences Conf. on Intelligents Systems for Mol. Biol.,<BR> and determine orientation. Glasgow et al., eds., The Am. Assoc. for Artificial<BR> Intelligence Press, Menlo Park, CA, pp. 175-182.<BR> <P>Motifs A program that seasrches amino acid sequences for patterns Bairohc, A. et al. (1997) Nucleic Acids Res. 25:217-221;<BR> that matched those defined in Prosite. Wisconsin Package Program Manual, version 9, page<BR> M51-59, Genetics Computer Group, Madison, WI.