Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PROTEINS ASSOCIATED WITH CELL GROWTH, DIFFERENTIATION, AND DEATH
Document Type and Number:
WIPO Patent Application WO/2003/095622
Kind Code:
A2
Abstract:
Various embodiments of the invention provide human proteins associated with cell growth, differentiation, and death (CGDD) and polynucleotides which identify and encode CGDD. Embodiments of the invention also provide expression vectors, host cells, antibodies, agonists, and antagonists. Other embodiments provide methods for diagnosing, treating, or preventing disorders associated with aberrant expression of CGDD.

Inventors:
KABLE AMY E (US)
ELLIOTT VICKI S (US)
KHARE REENA (US)
BULLOCH SEAN A (US)
LEE SOO YEUN (US)
BECHA SHANYA D (US)
CHAWLA NARINDER K (US)
TANG Y TOM (US)
YUE HENRY (US)
BAUGHN MARIAH R (US)
TRAN UYEN K (US)
KAREHT STEPHANIE K (US)
EMERLING BROOKE M (US)
GRIFFIN JENNIFER A (US)
SWARNAKAR ANITA (US)
CHANG HSIN-RU (US)
RAMKUMAR JAYALAXMI (US)
RICHARDSON THOMAS W (US)
MARQUIS JOSEPH P (US)
Application Number:
PCT/US2003/014742
Publication Date:
November 20, 2003
Filing Date:
May 08, 2003
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INCYTE CORP (US)
KABLE AMY E (US)
ELLIOTT VICKI S (US)
KHARE REENA (US)
BULLOCH SEAN A (US)
LEE SOO YEUN (US)
BECHA SHANYA D (US)
CHAWLA NARINDER K (US)
TANG Y TOM (US)
YUE HENRY (US)
BAUGHN MARIAH R (US)
TRAN UYEN K (US)
KAREHT STEPHANIE K (US)
EMERLING BROOKE M (US)
GRIFFIN JENNIFER A (US)
SWARNAKAR ANITA (US)
CHANG HSIN-RU (US)
RAMKUMAR JAYALAXMI (US)
RICHARDSON THOMAS W (US)
MARQUIS JOSEPH P (US)
International Classes:
C07K14/47; (IPC1-7): C12N/
Domestic Patent References:
WO1999024836A11999-05-20
Attorney, Agent or Firm:
FOLEY & LARDNER LLP (3000 K Street N.W.,Suite 50, Washington D.C., 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: 1-33, 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 : 2, SEQ ID N0 : 5, SEQ ID N0 : 9, SEQ ID NO: 12-14, SEQ ID NO: 16, SEQ ID NO : 19-20, SEQ ID N0 : 24, and SEQ ID N0 : 28-32, c) a polypeptide comprising a naturally occurring amino acid sequence at least 96% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 4 and SEQ ID NO: 17, d) a polypeptide comprising a naturally occurring amino acid sequence at least 91% identical to the amino acid sequence of SEQ ID N0 : 6, e) a polypeptide comprising a naturally occurring amino acid sequence at least 92% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 10 and SEQ ID N0 : 26-27, f) a polypeptide comprising a naturally occurring amino acid sequence at least 94% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 11 and SEQ ID N0 : 22, g) a polypeptide consisting essentially of a naturally occurring amino acid sequence at least 90% identical to an amino acid sequence selected from the group consisting of SEQ ID N0 : 1, SEQ ID N0 : 3, SEQ ID N0 : 7-8, SEQ ID N0 : 15, SEQ ID NO : 18, SEQ ID NO : 23, SEQ ID N0 : 25, and SEQ ID NO : 33, h) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, and i) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33.
2. An isolated polypeptide of claim 1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33.
3. An isolated polynucleotide encoding a polypeptide of claim 1.
4. An isolated polynucleotide encoding a polypeptide of claim 2.

5. An isolated polynucleotide of claim 4 comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66.
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: 1-33.
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 : 34-66, 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 : 45-47, SEQ ID NO : 52-53, and SEQ ID NO : 62-64, c) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 97% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 36, SEQ ID NO : 39, and SEQ ID NO : 57, d) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 98% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34 and SEQ ID NO : 49, e) a polynucleotide comprising a naturally occurring polynucleotide sequence at least

95% identical to the polynucleotide sequence of SEQ ID NO : 37, f) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 99% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 38, SEQ ID NO : 40, and SEQ ID NO : 43-44, g) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 92% identical to the polynucleotide sequence of SEQ ID NO : 65, h) a polynucleotide consisting essentially of a naturally occurring polynucleotide sequence at least 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 35, SEQ ID NO : 41-42, SEQ ID NO : 48, SEQ ID NO : 50-51, SEQ ID NO : 54, SEQ ID NO : 55-56, SEQ ID NO : 58, SEQ ID NO : 59-61, and SEQ ID NO : 66, i) a polynucleotide complementary to a polynucleotide of a), j) a polynucleotide complementary to a polynucleotide of b), k) a polynucleotide complementary to a polynucleotide of c), 1) a polynucleotide complementary to a polynucleotide of d), m) a polynucleotide complementary to a polynucleotide of e), n) a polynucleotide complementary to a polynucleotide of f), o) a polynucleotide complementary to a polynucleotide of g), p) a polynucleotide complementary to a polynucleotide of h), and q) an RNA equivalent of a)-p).

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: 1-33.
19. A method for treating a disease or condition associated with decreased expression of functional CGDD, 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 CGDD, 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 CGDD, 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 method for a diagnostic test for a condition or disease associated with the expression of CGDD 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 CGDD in a subject, comprising administering to said subject an effective amount of the composition of claim 32.
34. A composition of claim 32, further comprising a label.
35. A method of diagnosing a condition or disease associated with the expression of CGDD

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 : 1-33, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibodies from the 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 : 1-33.
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 : 1-33, or an immunogenic fragment thereof, under conditions to elicit an antibody response, b) isolating antibody producing cells from the animal, c) fusing the antibody producing cells with immortalized cells to form monoclonal antibody-producing hybridoma cells, d) culturing the hybridoma cells, and e) isolating from the culture monoclonal antibody which specifically binds to a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33.
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: 1-33 in a sample, the method comprising: a) incubating the antibody of claim 11 with the 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: 1-33 in the sample.
45. A method of purifying a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33 from a sample, the method comprising: a) incubating the antibody of claim 11 with the 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 : 1-33.
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 NO : 2.
58. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 3.
59. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 4.
60. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 5.

61. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 6.
62. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 7.
63. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 8.
64. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 9.
65. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 10.
66. A polypeptide of claim 1, comprising the amino acid sequence of SEQ BD NO: 11.
67. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 12.
68. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 13.
69. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 14.
70. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 15.
71. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 16.
72. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 17.
73. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 18.
74. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO: 19.
75. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 20.
76. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 21.
77. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 22.
78. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID NO : 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 polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 27.
83. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 28.
84. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 29.
85. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 30.
86. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 31.
87. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 32.
88. A polypeptide of claim 1, comprising the amino acid sequence of SEQ ID N0 : 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.
108. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 53.
109. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 54.
110. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 55.
111. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 56.
112. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 57.
113. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 58.
114. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 59.
115. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 60.
116. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 61.
117. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 62.

118. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 63.
119. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 64.
120. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 65.
121. A polynucleotide of claim 12, comprising the polynucleotide sequence of SEQ ID NO : 66.
Description:

PROTEINS ASSOCIATED WITH CELL GROWTH, DIFFERENTIATION, AND DEATH TECHNICAL FIELD The invention relates to novel nucleic acids, proteins associated with cell growth, differentiation, and death encoded by these nucleic acids, and to the use of these nucleic acids and proteins in the diagnosis, treatment, and prevention of cell proliferative disorders including cancer, developmental disorders, neurological disorders, autoimmune/inflammatory disorders, reproductive disorders, and disorders of the placenta. The invention also relates to the assessment of the effects of exogenous compounds on the expression of nucleic acids and proteins associated with cell growth, differentiation, and death.

BACKGROUND OF THE INVENTION Human growth and development requires the spatial and temporal regulation of cell differentiation, cell proliferation, and apoptosis. These processes coordinately control reproduction, aging, embryogenesis, morphogenesis, organogenesis, and tissue repair and maintenance. At the cellular level, growth and development is governed by the cell's decision to enter into or exit from the cell division cycle and by the cell's commitment to a terminally differentiated state. These decisions are made by the cell in response to extracellular signals and other environmental cues it receives. The following discussion focuses on the molecular mechanisms of cell division, embryogenesis, cell differentiation and proliferation, and apoptosis, as well as disease states such as cancer which can result from disruption of these mechanisms.

Cell Cycle Cell division is the fundamental process by which all living things grow and reproduce. In unicellular organisms such as yeast and bacteria, each cell division doubles the number of organisms.

In multicellular species many rounds of cell division are required to replace cells lost by wear or by programmed cell death, and for cell differentiation to produce a new tissue or organ. Progression through the cell cycle is governed by the intricate interactions of protein complexes. This regulation depends upon the appropriate expression of proteins which control cell cycle progression in response to extracellular signals, such as growth factors and other mitogens, and intracellular cues, such as DNA damage or nutrient starvation. Molecules which directly or indirectly modulate cell cycle progression fall into several categories, including cyclins, cyclin-dependent protein kinases, growth factors and their receptors, second messenger and signal transduction proteins, oncogene products, and tumor-suppressor proteins.

Details of the cell division cycle may vary, but the basic process consists of three principle events. The first event, interphase, involves preparations for cell division, replication of the DNA,

and production of essential proteins. In the second event, mitosis, the nuclear material is divided and separates to opposite sides of the cell. The final event, cytokinesis, is division and fission of the cell cytoplasm. The sequence and timing of cell cycle transitions is under the control of the cell cycle regulation system which controls the process by positive or negative regulatory circuits at various check points.

Mitosis marks the end of interphase and concludes with the onset of cytokinesis. There are four stages in mitosis, occurring in the following order: prophase, metaphase, anaphase and telophase. Prophase includes the formation obi-polar mitotic spindles, composed of microtubules and associated proteins such as dynein, which originate from polar mitotic centers. During metaphase, the nuclear material condenses and develops kinetochore fibers which aid in its physical attachment to the mitotic spindles. The ensuing movement of the nuclear material to opposite poles along the mitotic spindles occurs during anaphase. Telophase includes the disappearance of the mitotic spindles and kinetochore fibers from the nuclear material. Mitosis depends on the interaction of numerous proteins. For example, centromere-associated proteins such as CENP-A, -B, and-C, play structural roles in kinetochore formation and assembly (Saffery, R. et al. (2000) Human Mol.

Gen. 9: 175-185).

During the M phase of eukaryotic cell cycling, structural rearrangements occur ensuring appropriate distribution of cellular components between daughter cells. Breakdown of interphase structures into smaller subunits is common. The nuclear envelope breaks into vesicles, and nuclear lamins are disassembled. Subsequent phosphorylation of these lamins occurs and is maintained until telophase, at which time the nuclear lamina structure is reformed. cDNAs responsible for encoding M phase phosphorylation (MPPs) are components of U3 small nucleolar ribonucleoprotein (snoRNP), and relocalize to the nucleolus once mitosis is complete (Westendorf, J. M. et al. (1998) J. Biol.

Chem. 9: 437-449). U3 snoRNPs are essential mediators of RNA processing events.

Proteins involved in the regulation of cellular processes such as mitosis include the Ser/Thr- protein phosphatases type 1 (PP-1). PP-ls act by dephosphorylation of key proteins involved in the metaphase-anaphase transition. The gene PP1R7 encodes the regulatory polypeptide sds22, having at least six splice variants (Ceulemans, H. et al. (1999) Eur. J. Biochem. 262: 36-42). Sds22 modulates the activity of the catalytic subunit of PP-ls, and enhances the PP-1-dependent dephosphorylation of mitotic substrates.

Cell cycle regulatory proteins play an important role in cell proliferation and cancer. For example, failures in the proper execution and timing of cell cycle events can lead to chromosome segregation defects resulting in aneuploidy or polyploidy. This genomic instability is characteristic of transformed cells (Luca, F. C. and M. Winey (1998) Mol. Biol. Cell. 9: 29-46). A recently identified protein, mMOB 1, is the mammalian homolog of yeast MOB 1, an essential yeast gene

required for completion of mitosis and maintenance of ploidy. The mammalian mMOBI is a member of protein complexes including protein phosphatase 2A (PP2A), and its phosphorylation appears to be regulated by PP2A (Moreno, C. S. et al. (2001) J. Biol. Chem. 276: 24253-24260). PP2A has been implicated in the development of human cancers, including lung and colon cancers and leukemias.

Cell cycle regulation involves numerous proteins interacting in a sequential manner. The eukaryotic cell cycle consists of several highly controlled events whose precise order ensures successful DNA replication and cell division. Cells maintain the order of these events by making later events dependent on the successful completion of earlier events. This dependency is enforced by cellular mechanisms called checkpoints. Examples of additional cell cycle regulatory proteins include the histone deacetylases (HDACs). HDACs are involved in cell cycle regulation, and modulate chromatin structure. Human HDAC1 has been found to interact in vitro with the human Husl gene product, whose Schizosaccharo77lyces ponzbe homolog has been implicated in G2/M checkpoint control (Cai, R. L. et al. (2000) J. Biol. Chem. 275: 27909-27916).

DNA damage (G2) and DNA replication (S-phase) checkpoints arrest eukaryotic cells at the G2/M transition. This arrest provides time for DNA repair or DNA replication to occur before entry into mitosis. Thus, the G2/M checkpoint ensures that mitosis only occurs upon completion of DNA replication and in the absence of chromosomal damage. The Hus 1 gene of Schizosaccharomyces pombe is a cell cycle checkpoint gene, as are the rad family of genes (e. g., radl and rad9) (Volkmer, E. and L. M. Karnitz (1999) J. Biol. Chem. 274: 567-570; Kostrub C. F. et al. (1998) EMBO J.

17: 2055-2066). These genes are involved in the mitotic checkpoint, and are induced by either DNA damage or blockage of replication. Induction of DNA damage or replication block leads to loss of function of the Hus 1 gene and subsequent cell death. Human homologs have been identified for most of the rad genes, including ATM and ATR, the human homologs of rad3p. Mutations in the ATM gene are correlated with the severe congenital disease ataxia-telagiectasia (Savitsky, K. et al. (1995) Science 268: 1749-1753). The human Husl protein has been shown to act in a complex with radl protein which interacts with rad9, making them central components of a DNA damage-responsive protein complex of human cells (Volkmer and Karnitz, supra).

The entry and exit of a cell from mitosis is regulated by the synthesis and destruction of a family of activating proteins called cyclins. Cyclins act by binding to and activating a group of cyclin-dependent protein kinases (Cdks) which then phosphorylate and activate selected proteins involved in the mitotic process. Cyclins are characterized by a large region of shared homology that is approximately 180 amino acids in length and referred to as the"cyclin box" (Chapman, D. L. and D. J. Wolgemuth (1993) Development 118: 229-240). In addition, cyclins contain a conserved 9 amino acid sequence in the N-terminal region of the molecule called the"destruction box. "This sequence is believed to be a recognition code that triggers ubiquitin-mediated degradation of cyclin B (Hunt, T.

(1991) Nature 349: 100-101). Several types of cyclins exist (Ciechanover, A. (1994) Cell 79: 13-21).

Progression through G1 and S phase is driven by the Gl cyclins and their catalytic subunits, including Cdk2-cyclin A, Cdk2-cyclin E, Cdk4-cyclin D and Cdk6-cyclin D. Progression through the G2-M transition is driven by the activation of mitotic CDK-cyclin complexes such as Cdc2-cyclin A, Cdc2-cyclin B1 and Cdc2-cyclin B2 complexes (reviewed in Yang, J. and S. Kornbluth (1999) Trends Cell Biol. 9: 207-210).

Cyclins are degraded through the ubiquitin conjugation system (UCS), a major pathway for the degradation of cellular proteins in eukaroytic cells and in some bacteria. The UCS mediates the elimination of abnormal proteins and regulates the half-lives of important regulatory proteins that control cellular processes such as gene transcription and cell cycle progression. The UCS is implicated in the degradation of mitotic cyclin kinases, oncoproteins, tumor suppressor genes such as p53, viral proteins, cell surface receptors associated with signal transduction, transcriptional regulators, and mutated or damaged proteins (Ciechanover, supra).

The process of ubiquitin conjugation and protein degradation occurs in five principle steps (Jentsch, S. (1992) Annu. Rev. Genet. 26: 179-207). First ubiquitin (Ub), a small, heat stable protein is activated by a ubiquitin-activating enzyme (El) in an ATP dependent reaction which binds the C- terminus of Ub to the thiol group of an internal cysteine residue in E1. Second, activated Ub is transferred to one of several Ub-conjugating enzymes (E2). Different ubiquitin-dependent proteolytic pathways employ structurally similar, but distinct ubiquitin-conjugating enzymes that are associated with recognition subunits which direct them to proteins carrying a particular degradation signal. Third, E2 transfers the Ub molecule through its C-terminal glycine to a member of the ubiquitin-protein ligase family, E3. Fourth, E3 transfers the Ub molecule to the target protein.

Additional Ub molecules may be added to the target protein forming a multi-Ub chain structure.

Fifth, the ubiquinated protein is then recognized and degraded by the proteasome, a large, multisubunit proteolytic enzyme complex, and Ub is released for re-utilization.

Prior to activation, Ub is usually expressed as a fusion protein composed of an N-terminal ubiquitin and a C-terminal extension protein (CEP) or as a polyubiquitin protein with Ub monomers attached head to tail. CEPs have characteristics of a variety of regulatory proteins; most are highly basic, contain up to 30% lysine and arginine residues, and have nucleic acid-binding domains (Monia, B. P. et al. (1989) J. Biol. Chem. 264: 4093-4103). The fusion protein is an important intermediate which appears to mediate co-regulation of the cell's translational and protein degradation activities, as well as localization of the inactive enzyme to specific cellular sites. Once delivered, C-terminal hydrolases cleave the fusion protein to release a functional Ub (Monia et al., supra).

Ub-conjugating enzymes (E2s) are important for substrate specificity in different UCS

pathways. All E2s have a conserved domain of approximately 16 kDa called the UBC domain that is at least 35% identical in all E2s and contains a centrally located cysteine residue required for ubiquitin-enzyme thiolester formation (Jentsch, supra). A well conserved proline-rich element is located N-terminal to the active cysteine residue. Structural variations beyond this conserved domain are used to classify the E2 enzymes. Class I E2s consist almost exclusively of the conserved UBC domain. Class II E2s have various unrelated C-terminal extensions that contribute to substrate specificity and cellular localization. Class III E2s have unique N-terminal extensions which are believed to be involved in enzyme regulation or substrate specificity.

A mitotic cyclin-specific E2 (E2-C) is characterized by the conserved UBC domain, an N- terminal extension of 30 amino acids not found in other E2s, and a 7 amino acid unique sequence adjacent to this extension. These characteristics together with the high affinity of E2-C for cyclin identify it as a new class of E2 (Aristarkhov, A. et al. (1996) Proc. Natl. Acad. Sci. 93: 4294-99).

Ubiquitin-protein ligases (E3s) catalyze the last step in the ubiquitin conjugation process, covalent attachment of ubiquitin to the substrate. E3 plays a key role in determining the specificity of the process. Only a few E3s have been identified so far. One type of E3 ligases is the HECT (homologous to E6-AP C-terminus) domain protein family. One member of the family, E6-AP (E6-associated protein) is required, along with the human papillomavirus (HPV) E6 oncoprotein, for the ubiquitination and degradation of p53 (Scheffner, M. et al. (1993) Cell 75: 495-505). The C-terminal domain of HECT proteins contains the highly conserved ubiquitin-binding cysteine residue. The N-terminal region of the various HECT proteins is variable and is believed to be involved in specific substrate recognition (Huibregtse, J. M. et al. (1997) Proc. Natl Acad. Sci. USA 94: 3656-3661). The SCF (Skpl-Cdc53/Cullin-F box receptor) family of proteins comprise another group of ubiquitin ligases (Deshaies, R. (1999) Annu. Rev. Dev. Biol. 15: 435-467). Multiple proteins are recruited into the SCF complex, including Skpl, cullin, and an F box domain containing protein.

The F box protein binds the substrate for the ubiquitination reaction and may play roles in determining substrate specificity and orienting the substrate for reaction. Skpl interacts with both the F box protein and cullin and may be involved in positioning the F box protein and cullin in the complex for transfer of ubiquitin from the E2 enzyme to the protein substrate. Substrates of SCF ligases include proteins involved in regulation of CDK activity, activation of transcription, signal transduction, assembly of kinetochores, and DNA replication.

Sgtl was identified in a screen for genes in yeast that suppress defects in kinetochore function caused by mutations in Skpl (Kitagawa, K. et al. (1999) Mol. Cell 4: 21-33). Sgtl interacts with Skpl and associates with SCF ubiquitin ligase. Defects in Sgtl cause arrest of cells at either Gl or G2 stages of the cell cycle. A yeast Sgtl null mutant can be rescued by human Sgtl, an indication of the conservation of Sgtl function across species. Sgtl is required for assembly of kinetochore

complexes in yeast.

Abnormal activities of the UCS are implicated in a number of diseases and disorders. These include, e. g. , cachexia (Llovera, M. et al. (1995) Int. J. Cancer 61 : 138-141), degradation of the tumor-suppressor protein, p53 (Ciechanover, supra), and neurodegeneration such as observed in Alzheimer's disease (Gregori, L. et al. (1994) Biochem. Biophys. Res. Commun. 203: 1731-1738).

Since ubiquitin conjugation is a rate-limiting step in antigen presentation, the ubiquitin degradation pathway may also have a critical role in the immune response (Grant, E. P. et al. (1995) J. Immunol.

155: 3750-3758).

Certain cell proliferation disorders can be identified by changes in the protein complexes that normally control progression through the cell cycle. A primary treatment strategy involves reestablishing control over cell cycle progression by manipulation of the proteins involved in cell cycle regulation (Nigg, E. A. (1995) BioEssays 17: 471-480).

Embryogenesis Mammalian embryogenesis is a process which encompasses the first few weeks of development following conception. During this period, embryogenesis proceeds from a single fertilized egg to the formation of the three embryonic tissues, then to an embryo which has most of its internal organs and all of its external features.

The normal course of mammalian embryogenesis depends on the correct temporal and spatial regulation of a large number of genes and tissues. These regulation processes have been intensely studied in mouse. An essential process that is still poorly understood is the activation of the embryonic genome after fertilization. As mouse oocytes grow, they accumulate transcripts that are either translated directly into proteins or stored for later activation by regulated polyadenylation.

During subsequent meiotic maturation and ovulation, the maternal genome is transcriptionally inert, and most maternal transcripts are deadenylated and/or degraded prior to, or together with, the activation of the zygotic genes at the two-cell stage (Stutz, A. et al. (1998) Genes Dev. 12 : 2535- 2548). The maternal to embryonic transition involves the degradation of oocyte, but not zygotic transcripts, the activation of the embryonic genome, and the induction of cell cycle progression to accommodate early development.

MATER (Maternal Antigen That Embryos Require) was initially identified as a target of antibodies from mice with ovarian immunity (Tong, Z-B. and L. M. Nelson (1999) Endocrinology 140: 3720-3726). Expression of the gene encoding MATER is restricted to the oocyte, making it one of a limited number of known maternal-effect genes in mammals (Tong, Z-B. et al. (2000) Mamm.

Genome 11: 281-287). The MATER protein is required for embryonic development beyond two cells, based upon preliminary results from mice in which this gene has been inactivated. The 1111-amino acid MATER protein contains a hydrophilic repeat region in the amino terminus, and a region

containing 14 leucine-rich repeats in the carboxyl terminus. These repeats resemble the sequence found in porcine ribonuclease inhibitor that is critical for protein-protein interactions.

The degradation of maternal transcripts during meiotic maturation and ovulation may involve the activation of a ribonuclease just prior to ovulation. Thus the function of MATER may be to bind to the maternal ribonuclease and prevent degradation of zygotic transcripts (Tong et al., supra). In addition to its role in oocyte development and embryogenesis, MATER may also be relevant to the pathogenesis of ovarian immunity, as it is a target of autoantibodies in mice with autoimmune oophoritis (Tong and Nelson, supra).

The maternal mRNA D7 is a moderately abundant transcript in Xerzopus laevis whose expression is highest in, and perhaps restricted to, oogenesis and early embryogenesis. The D7 protein is absent from oocytes and first begins to accumulate during oocyte maturation. Its levels are highest during the first day of embryonic development and then they decrease. The loss of D7 protein affects the maturation process itself, significantly delaying the time course of germinal vesicle breakdown. Thus, D7 is a newly described protein involved in oocyte maturation (Smith, R. C. et al.

(1988) Genes Dev. 2 (10): 1296-306. ) Many other genes are involved in subsequent stages of embryogenesis. After fertilization, the oocyte is guided by fimbria at the distal end of each fallopian tube into and through the fallopian tube and thence into the uterus. Changes in the uterine endometrium prepare the tissue to support the implantation and embryonic development of a fertilized ovum. Several stages of division have occurred before the dividing ovum, now a blastocyst with about 100 cells, enters the uterus. Upon reaching the uterus, the developing blastocyst usually remains in the uterine cavity an additional two to four days before implanting in the endometrium, the inner lining of the uterus. Implantation results from the action of trophoblast cells that develop over the surface of the blastocyst. These cells secrete proteolytic enzymes that digest and liquefy the cells of the endometrium. The invasive process is reviewed in Fisher, S. J. and C. H. Damsky (1993; Semin Cell Biol 4: 183-188) and Graham, C. H. and P. K. Lala (1992; Biochem Cell Biol 70: 867-874). Once implantation has taken place, the trophoblast and other sublying cells proliferate rapidly, forming the placenta and the various membranes of pregnancy. (See Guyton, A. C. (1991) Textbook of Medical Physiology, 8'b ed. W. B.

Saunders Company, Philadelphia PA, pp. 915-919.) The placenta has an essential role in protecting and nourishing the developing fetus. In most species the syncytiotrophoblast layer is present on the outside of the placenta at the fetal-maternal interface. This is a continuous structure, one cell deep, formed by the fusion of the constituent trophoblast cells. The syncytiotrophoblast cells play important roles in maternal-fetal exchange, in tissue remodeling during fetal development, and in protecting the developing fetus from the maternal immune response (Stoye, J. P. and J. M. Coffin (2000) Nature 403: 715-717).

A gene called syncytin is the envelope gene of a human endogenous defective provirus.

Syncytin is expressed in high levels in placenta, and more weakly in testis, but is not detected in any other tissues (Mi, S. et al. (2000) Nature 403: 785-789). Syncytin expression in the placenta is restricted to the syncytiotrophoblasts. Since retroviral env proteins are often involved in promoting cell fusion events, it was thought that syncytin might be involved in regulating the fusion of trophoblast cells into the syncytiotrophoblast layer. Experiments demonstrated that syncytin can mediate cell fusion in vitro, and that anti-syncytin antibodies can inhibit the fusion of placental cytotrophoblasts (Mi et al., supra). In addition, a conserved immunosuppressive domain present in retroviral envelope proteins, and found in syncytin at amino acid residues 373-397, might be involved in preventing maternal immune responses against the developing embryo.

Syncytin may also be involved in regulating trophoblast invasiveness by inducing trophoblast fusion and terminal differentiation (Mi et al., supra). Insufficient trophoblast infiltration of the uterine wall is associated with placental disorders such as preeclampsia, or pregnancy induced hypertension, while uncontrolled trophoblast invasion is observed in choriocarcinoma and other gestational trophoblastic diseases. Thus syncytin function may be involved in these diseases.

Cell Differentiation Multicellular organisms are comprised of diverse cell types that differ dramatically both in structure and function, despite the fact that each cell is like the others in its hereditary endowment.

Cell differentiation is the process by which cells come to differ in their structure and physiological function. The cells of a multicellular organism all arise from mitotic divisions of a single-celled zygote. The zygote is totipotent, meaning that it has the ability to give rise to every type of cell in the adult body. During development the cellular descendants of the zygote lose their totipotency and become determined. Once its prospective fate is achieved, a cell is said to have differentiated. All descendants of this cell will be of the same type.

Human growth and development requires the spatial and temporal regulation of cell differentiation, along with cell proliferation and regulated cell death. These processes coordinate to control reproduction, aging, embryogenesis, morphogenesis, organogenesis, and tissue repair and maintenance. The processes involved in cell differentiation are also relevant to disease states such as cancer, in which case the factors regulating normal cell differentiation have been altered, allowing the cancerous cells to proliferate in an anaplastic, or undifferentiated, state.

The mechanisms of differentiation involve cell-specific regulation of transcription and translation, so that different genes are selectively expressed at different times in different cells.

Genetic experiments using the fruit fly Drosophila nzelanogaster have identified regulated cascades of transcription factors which control pattern formation during development and differentiation.

These include the homeotic genes, which encode transcription factors containing homeobox motifs.

The products of homeotic genes determine how the insect's imaginal discs develop from masses of undifferentiated cells to specific segments containing complex organs. Many genes found to be involved in cell differentiation and development in Drosophila have homologs in mammals. Some human genes have equivalent developmental roles to their Drosophila homologs. The human homolog of the Drosophila eyes absent gene (eya) underlies branchio-oto-renal syndrome, a developmental disorder affecting the ears and kidneys (Abdelhak, S. et al. (1997) Nat. Genet. 15: 157- 164). The Drosophila slit gene encodes a secreted leucine-rich repeat containing protein expressed by the midline glial cells and required for normal neural development.

At the cellular level, growth and development are governed by the cell's decision to enter into or exit from the cell cycle and by the cell's commitment to a terminally differentiated state.

Differential gene expression within cells is triggered in response to extracellular signals and other environmental cues. Such signals include growth factors and other mitogens such as retinoic acid; cell-cell and cell-matrix contacts; and environmental factors such as nutritional signals, toxic substances, and heat shock. Candidate genes that may play a role in differentiation can be identified by altered expression patterns upon induction of cell differentiation in vitro.

The final step in cell differentiation results in a specialization that is characterized by the production of particular proteins, such as contractile proteins in muscle cells, serum proteins in liver cells and globins in red blood cell precursors. The expression of these specialized proteins depends at least in part on cell-specific transcription factors. For example, the homeobox-containing transcription factor PAX-6 is essential for early eye determination, specification of ocular tissues, and normal eye development in vertebrates.

In the case of epidermal differentiation, the induction of differentiation-specific genes occurs either together with or following growth arrest and is believed to be linked to the molecular events that control irreversible growth arrest. Irreversible growth arrest is an early event which occurs when cells transit from the basal to the innermost suprabasal layer of the skin and begin expressing squamous-specific genes. These genes include those involved in the formation of the cross-linked envelope, such as transglutaminase I and III, involucrin, loricin, and small proline-rich repeat (SPRR) proteins. The SPRR proteins are 8-10 kDa in molecular mass, rich in proline, glutamin, and cysteine, and contain similar repeating sequence elements. The SPRR proteins may be structural proteins with a strong secondary structure or metal-binding proteins such as metallothioneins.

(Jetten, A. M. and B. L. Harvat (1997) J. Dermatol. 24: 711-725; PRINTS Entry PR00021 PRORICH Small proline-rich protein signature.) The Wnt gene family of secreted signaling molecules is highly conserved throughout eukaryotic cells. Members of the Wnt family are involved in regulating chondrocyte differentiation within the cartilage template. Wnt-5a, Wnt-5b and Wnt-4 genes are expressed in chondrogenic

regions of the chicken limb, Wnt-5a being expressed in the perichondrium (mesenchymal cells immediately surrounding the early cartilage template). Wnt-5a misexpression delays the maturation of chondrocytes and the onset of bone collar formation in chicken limb (Hartmann, C. and C. J. Tabin (2000) Development 127: 3141-3159).

Glypicans are a family of cell surface heparan sulfate proteoglycans that play an important role in cellular growth control and differentiation. Cerebroglycan, a heparan sulfate proteoglycan expressed in the nervous system, is involved with the motile behavior of developing neurons (Stipp, C. S. et al. (1994) J. Cell Biol. 124 : 149-160).

Notch plays an active role in the differentiation of glial cells, and influences the length and organization of neuronal processes (for a review, see Frisen, J. and U. Lendahl (2001) Bioessays 23: 3-7). The Notch receptor signaling pathway is important for morphogenesis and development of many organs and tissues in multicellular species. Drosophila fringe proteins modulate the activation of the Notch signal transduction pathway at the dorsal-ventral boundary of the wing imaginal disc.

Mammalian fringe-related family members participate in boundary determination during segmentation (Johnston, S. H. et al. (1997) Development 124: 2245-2254).

Recently a number of proteins have been found to contain a conserved cysteine-rich domain of about 60 amino-acid residues called the LIM domain (for Lin-11 Isl-1 Mec-3) (Freyd, G. et al.

(1990) Nature 344 : 876-879; Baltz, R. et al. (1992) Plant Cell 4: 1465-1466). In the LIM domain, there are seven conserved cysteine residues and a histidine. The LIM domain binds two zinc ions (Michelsen, J. W. et al. (1993) Proc. Natl. Acad. Sci. U. S. A. 90 : 4404-4408). LIM does not bind DNA; rather, it seems to act as an interface for protein-protein interaction.

Apoptosis Apoptosis is the genetically controlled process by which unneeded or defective cells undergo programmed cell death. Selective elimination of cells is as important for morphogenesis and tissue remodeling as is cell proliferation and differentiation. Lack of apoptosis may result in hyperplasia and other disorders associated with increased cell proliferation. Apoptosis is also a critical component of the immune response. Immune cells such as cytotoxic T-cells and natural killer cells prevent the spread of disease by inducing apoptosis in tumor cells and virus-infected cells. In addition, immune cells that fail to distinguish self molecules from foreign molecules must be eliminated by apoptosis to avoid an autoimmune response.

Apoptotic cells undergo distinct morphological changes. Hallmarks of apoptosis include cell shrinkage, nuclear and cytoplasmic condensation, and alterations in plasma membrane topology.

Biochemically, apoptotic cells are characterized by increased intracellular calcium concentration, fragmentation of chromosomal DNA, and expression of novel cell surface components.

The molecular mechanisms of apoptosis are highly conserved, and many of the key protein

regulators and effectors of apoptosis have been identified. Apoptosis generally proceeds in response to a signal which is transduced intracellularly and results in altered patterns of gene expression and protein activity. Signaling molecules such as hormones and cytokines are known both to stimulate and to inhibit apoptosis through interactions with cell surface receptors. Transcription factors also play an important role in the onset of apoptosis. A number of downstream effector molecules, especially proteases, have been implicated in the degradation of cellular components and the proteolytic activation of other apoptotic effectors.

The Bcl-2 family of proteins, as well as other cytoplasmic proteins, are key regulators of apoptosis. There are at least 15 Bcl-2 family members within 3 subfamilies. These proteins have been identified in mammalian cells and in viruses, and each possesses at least one of four Bcl-2 homology domains (BH1 to BH4), which are highly conserved. Bcl-2 family proteins contain the BH1 and BH2 domains, which are found in members of the pro-survival subfamily, while those proteins which are most similar to Bcl-2 have all four conserved domains, enabling inhibition of apoptosis following encounters with a variety of cytotoxic challenges. Members of the pro-survival subfamily include Bcl-2, Bcl-xL, Bcl-w, Mcl-1, and Al in mammals; NF-13 (chicken); CED-9 (Caenorhabditis elegans) ; and viral proteins BHRF1, LMW5-HL, ORF16, KS-Bcl-2, and E1B-19K.

The BH3 domain is essential for the function of pro-apoptosis subfamily proteins. The two pro- apoptosis subfamilies, Bax and BH3, include Bax, Bak, and Bok (also called Mtd); and Bik, Blk, Hrk, BNIP3, BimL, Bad, Bid, and Egl-1 (C. elegant) ; respectively. Members of the Bax subfamily contain the BH1, BH2, and BH3 domains, and resemble Bcl-2 rather closely. In contrast, members of the BH3 subfamily have only the 9-16 residue BH3 domain, being otherwise unrelated to any known protein, and only Bik and Blk share sequence similarity. The proteins of the two pro-apoptosis subfamilies may be the antagonists of pro-survival subfamily proteins. This is illustrated in C. elegans where Egl-1, which is required for apoptosis, binds to and acts via CED-9 (for review, see Adams, J. M. and S. Cory (1998) Science 281: 1322-1326).

Heterodimerization between pro-apoptosis and anti-apoptosis subfamily proteins seems to have a titrating effect on the functions of these protein subfamilies, which suggests that relative concentrations of the members of each subfamily may act to regulate apoptosis. Heterodimerization is not required for a pro-survival protein; however, it is essential in the BH3 subfamily, and less so in the Bax subfamily.

The Bcl-2 protein has 2 isoforms, alpha and beta, which are formed by alternative splicing. It forms homodimers and heterodimers with Bax and Bak proteins and the Bcl-X isoform Bcl-xs.

Heterodimerization with Bax requires intact BH1 and BH2 domains, and is necessary for pro-survival activity. The BH4 domain seems to be involved in pro-survival activity as well. Bcl-2 is located within the inner and outer mitochondrial membranes, as well as within the nuclear envelope and

endoplasmic reticulum, and is expressed in a variety of tissues. Its involvement in follicular lymphoma (type II chronic lymphatic leukemia) is seen in a chromosomal translocation T (14; 18) (q32; q21) and involves immunoglobulin gene regions.

The Bcl-x protein is a dominant regulator of apoptotic cell death. Alternative splicing results in three isoforms, Bcl-xB, a long isoform, and a short isoform. The long isoform exhibits cell death repressor activity, while the short isoform promotes apoptosis. Bcl-xL forms heterodimers with Bax and Bak, although heterodimerization with Bax does not seem to be necessary for pro-survival (anti- apoptosis) activity. Bcl-xS forms heterodimers with Bcl-2. Bcl-x is found in mitochondrial membranes and the perinuclear envelope. Bcl-xS is expressed at high levels in developing lymphocytes and other cells undergoing a high rate of turnover. Bcl-xL is found in adult brain and in other tissues'long-lived post-mitotic cells. As with Bcl-2, the BH1, BH2, and BH4 domains are involved in pro-survival activity.

The Bcl-w protein is found within the cytoplasm of almost all myeloid cell lines and in numerous tissues, with the highest levels of expression in brain, colon, and salivary gland. This protein is expressed in low levels in testis, liver, heart, stomach, skeletal muscle, and placenta, and a few lymphoid cell lines. Bcl-w contains the BH1, BH2, and BH4 domains, all of which are needed for its cell survival promotion activity. Although mice in which Bcl-w gene function was disrupted by homologous recombination were viable, healthy, and normal in appearance, and adult females had normal reproductive function, the adult males were infertile. In these males, the initial, prepuberty stage of spermatogenesis was largely unaffected and the testes developed normally. However, the seminiferous tubules were disorganized, contained numerous apoptotic cells, and were incapable of producing mature sperm. This mouse model may be applicable to some cases of human male sterility and suggests that alteration of programmed cell death in the testes may be useful in modulating fertility (Print, C. G. et al. (1998) Proc. Natl. Acad. Sci. USA 95: 12424-12431).

Studies in rat ischemic brain found Bcl-w to be overexpressed relative to its normal low constitutive level of expression in nonischemic brain. Furthermore, ifs vitro studies to examine the mechanism of action of Bcl-w revealed that isolated rat brain mitochondria were unable to respond to an addition of recombinant Bax or high concentrations of calcium when Bcl-w was also present. The normal response would be the release of cytochrome c from the mitochondria. Additionally, recombinant Bcl-w protein was found to inhibit calcium-induced loss of mitochondrial transmembrane potential, which is indicative of permeability transition. Together these findings suggest that Bcl-w may be a neuro-protectant against ischemic neuronal death and may achieve this protection via the mitochondrial death-regulatory pathway (Yan, C. et al. (2000) J. Cereb. Blood Flow Metab. 20: 620-630).

The bfl-1 gene is an additional member of the Bcl-2 family, and is also a suppressor of

apoptosis. The Bfl-1 protein has 175 amino acids, and contains the BH1, BH2, and BH3 conserved domains found in Bcl-2 family members. It also contains a Gln-rich NH2-terminal region and lacks an NH domain 1, unlike other Bcl-2 family members. The mouse Al protein shares high sequence homology with Bfl-l and has the 3 conserved domains found in Bfl-1. Apoptosis induced by the p53 tumor suppressor protein is suppressed by Bfl-1, similar to the action of Bcl-2, Bcl-xL, and EBV- BHRF1 (D'Sa-Eipper, C. et al. (1996) Cancer Res. 56: 3879-3882). Bfl-1 is found intracellularly, with the highest expression in the hematopoietic compartment, i. e. blood, spleen, and bone marrow; moderate expression in lung, small intestine, and testis; and minimal expression in other tissues. It is also found in vascular smooth muscle cells and hematopoietic malignancies. A correlation has been noted between the expression level of bfl-l and the development of stomach cancer, suggesting that the Bfl-1 protein is involved in the development of stomach cancer, either in the promotion of cancerous cell survival or in cancer (Choi, S. S. et al. (1995) Oncogene 11: 1693-1698).

Cancers are characterized by continuous or uncontrolled cell proliferation. Some cancers are associated with suppression of normal apoptotic cell death. Strategies for treatment may involve either reestablishing control over cell cycle progression, or selectively stimulating apoptosis in cancerous cells (Nigg, E. A. (1995) BioEssays 17: 471-480). Immunological defenses against cancer include induction of apoptosis in mutant cells by tumor suppressors, and the recognition of tumor antigens by T lymphocytes. Response to mitogenic stresses is frequently controlled at the level of transcription and is coordinated by various transcription factors. For example, the Rel/NF-kappa B family of vertebrate transcription factors plays a pivotal role in inflammatory and immune responses to radiation. The NF-kappa B family includes p50, p52, RelA, ReIB, cRel, and other DNA-binding proteins. The p52 protein induces apoptosis, upregulates the transcription factor c-Jun, and activates c-Jun N-terminal kinase 1 (JNK1) (Sun, L. et al. (1998) Gene 208: 157-166). Most NF-kappa B proteins form DNA-binding homodimers or heterodimers. Dimerization of many transcription factors is mediated by a conserved sequence known as the bZIP domain, characterized by a basic region followed by a leucine zipper.

The Fas/Apo-1 receptor (FAS) is a member of the tumor necrosis factor (TNF) receptor family. Upon binding its ligand (Fas ligand), the membrane-spanning FAS induces apoptosis by recruiting several cytoplasmic proteins that transmit the death signal. One such protein, termed FAS- associated protein factor 1 (FAF1), was isolated from mice, and it was demonstrated that expression of FAF1 in L cells potentiated FAS-induced apoptosis (Chu, K. et al. (1995) Proc. Natl. Acad. Sci.

USA 92: 11894-11898). Subsequently, FAS-associated factors have been isolated from numerous other species, including fruit fly and quail (Frohlich, T. et al. (1998) J. Cell Sci. 111: 2353-2363).

Another cytoplasmic protein that functions in the transmittal of the death signal from Fas is the Fas- associated death domain protein, also known as FADD. FADD transmits the death signal in both

FAS-mediated and TNF receptor-mediated apoptotic pathways by activating caspase-8 (Bang, S. et al.

(2000) J. Biol. Chem. 275: 36217-36222).

Fragmentation of chromosomal DNA is one of the hallmarks of apoptosis. DNA fragmentation factor (DFF) is a protein composed of two subunits, a 40-kDa caspase-activated nuclease termed DFF40/CAD, and its 45-kDa inhibitor DFF45/ICAD. Two mouse homologs of DFF45/ICAD, termed CIDE-A and CIDE-B, have recently been described (Inohara, N. et al. (1998) EMBO J. 17: 2526-2533). CIDE-A and CIDE-B expression in mammalian cells activated apoptosis, while expression of CIDE-A alone induced DNA fragmentation. In addition, FAS-mediated apoptosis was enhanced by CIDE-A and CIDE-B, further implicating these proteins as effectors that mediate apoptosis.

Transcription factors play an important role in the onset of apoptosis. A number of downstream effector molecules, particularly proteases such as the cysteine proteases called caspases, are involved in the initiation and execution phases of apoptosis. The activation of the caspases results from the competitive action of the pro-survival and pro-apoptosis Bcl-2-related proteins (Print, C. G. et al. (1998) Proc. Natl. Acad. Sci. USA 95: 12424-12431). A pro-apoptotic signal can activate initiator caspases that trigger a proteolytic caspase cascade, leading to the hydrolysis of target proteins and the classic apoptotic death of the cell. Two active site residues, a cysteine and a histidine, have been implicated in the catalytic mechanism. Caspases are among the most specific endopeptidases, cleaving after aspartate residues.

Caspases are synthesized as inactive zymogens consisting of one large (p20) and one small (plO) subunit separated by a small spacer region, and a variable N-terminal prodomain. This prodomain interacts with cofactors that can positively or negatively affect apoptosis. An activating signal causes autoproteolytic cleavage of a specific aspartate residue (D297 in the caspase-1 numbering convention) and removal of the spacer and prodomain, leaving a p10/p20 heterodimer.

Two of these heterodimers interact via their small subunits to form the catalytically active tetramer.

The long prodomains of some caspase family members have been shown to promote dimerization and auto-processing of procaspases. Some caspases contain a"death effector domain"in their prodomain by which they can be recruited into self-activating complexes with other caspases and FADD protein- associated death receptors or the TNF receptor complex. In addition, two dimers from different caspase family members can associate, changing the substrate specificity of the resultant tetramer.

Tumor necrosis factor (TNF) and related cytokines induce apoptosis in lymphoid cells.

(Reviewed in Nagata, S. (1997) Cell 88: 355-365. ) Binding of TNF to its receptor triggers a signal transduction pathway that results in the activation of a proteolytic caspase cascade. One such caspase, ICE (Interleukin-1p converting enzyme), is a cysteine protease comprised of two large and two small subunits generated by ICE auto-cleavage (Dinarello, C. A. (1994) FASEB J. 8: 1314-1325).

ICE is expressed primarily in monocytes. ICE processes the cytokine precursor, interleukin-lp, into its active form, which plays a central role in acute and chronic inflammation, bone resorption, myelogenous leukemia, and other pathological processes. ICE and related caspases cause apoptosis when overexpressed in transfected cell lines.

A caspase recruitment domain (CARD) is found within the prodomain of several apical caspases and is conserved in several apoptosis regulatory molecules such as Apaf-2, RAIDD, and cellular inhibitors of apoptosis proteins (IAPs) (Hofmann, K. et al. (1997) Trends Biochem. Sci.

22: 155-157). The regulatory role of CARD in apoptosis may be to allow proteins such as Paf 1 to associate with caspase-9 (Li, P. et al. (1997) Cell 91: 479-489). A human cDNA encoding an apoptosis repressor with a CARD (ARC) which is expressed in both skeletal and cardiac muscle has been identified and characterized. ARC functions as an inhibitor of apoptosis and interacts selectively with caspases (Koseki, T. et al. (1998) Proc. Natl. Acad. Sci. USA 95: 5156-5160). All of these interactions have clear effects on the control of apoptosis (reviewed in Chan S. L. and M. P.

Mattson (1999) J. Neurosci. Res. 58: 167-190; Salveson, G. S. and V. M. Dixit (1999) Proc. Natl.

Acad. Sci. USA 96: 10964-10967).

ES18 was identified as a potential regulator of apoptosis in mouse T-cells (Park, E. J. et al.

(1999) Nuc. Acid. Res. 27: 1524-1530). ES18 is 428 amino acids in length, contains an N-terminal proline-rich region, an acidic glutamic acid-rich domain, and a putative LXXLL nuclear receptor binding motif. The protein is preferentially expressed in lymph nodes and thymus. The level of ES18 expression increases in T-cell thymoma S49. 1 in response to treatment with dexamethasone, staurosporine, or C2-ceramide, which induce apoptosis. ES18 may play a role in stimulating apoptotic cell death in T-cells.

The rat ventral prostate (RVP) is a model system for the study of hormone-regulated apoptosis. RVP epithelial cells undergo apoptosis in response to androgen deprivation. Messenger RNA (mRNA) transcripts that are up-regulated in the apoptotic RVP have been identified (Briehl, M.

M. and R. L. Miesfeld (1991) Mol. Endocrinol. 5: 1381-1388). One such transcript encodes RVP. l, the precise role of which in apoptosis has not been determined. The human homolog of RVP. 1, hRVPl, is 89% identical to the rat protein (Katahira, J. et al. (1997) J. Biol. Chem. 272: 26652- 26658). hRVPl is 220 amino acids in length and contains four transmembrane domains. hRVPl is highly expressed in the lung, intestine, and liver. Interestingly, hRVPl functions as a low affinity receptor for the Clostridium perfringens enterotoxin, a causative agent of diarrhea in humans and other animals.

Cytokine-mediated apoptosis plays an important role in hematopoiesis and the immune response. Myeloid cells, which are the stem cell progenitors of macrophages, neutrophils, erythrocytes, and other blood cells, proliferate in response to specific cytokines such as

granulocyte/macrophage-colony stimulating factor (GM-CSF) and interleukin-3 (IL-3). When deprived of GM-CSF or IL-3, myeloid cells undergo apoptosis. The murine requiem (req) gene encodes a putative transcription factor required for this apoptotic response in the myeloid cell line FDCP-1 (Gabig, T. G. et al. (1994) J. Biol. Chem. 269: 29515-29519). The Req protein is 371 amino acids in length and contains a nuclear localization signal, a single Kruppel-type zinc finger, an acidic domain, and a cluster of four unique zinc-finger motifs enriched in cysteine and histidine residues involved in metal binding. Expression of req is not myeloid-or apoptosis-specific, suggesting that additional factors regulate Req activity in myeloid cell apoptosis.

Dysregulation of apoptosis has recently been recognized as a significant factor in the pathogenesis of many human diseases. For example, excessive cell survival caused by decreased apoptosis can contribute to disorders related to cell proliferation and the immune response. Such disorders include cancer, autoimmune diseases, viral infections, and inflammation. In contrast, excessive cell death caused by increased apoptosis can lead to degenerative and immunodeficiency disorders such as AIDS, neurodegenerative diseases, and myelodysplastic syndromes. (Thompson, C. B. (1995) Science 267: 1456-1462. ) Impaired regulation of apoptosis is also associated with loss of neurons in Alzheimer's disease. Alzheimer's disease is a progressive neurodegenerative disorder that is characterized by the formation of senile plaques and neurofibrillary tangles containing amyloid beta peptide. These plaques are found in limbic and association cortices of the brain, including hippocampus, temporal cortices, cingulate cortex, amygdala, nucleus basalis and locus caeruleus. B-amyloid peptide participates in signaling pathways that induce apoptosis and lead to the death of neurons (Kajkowski, C. et al. (2001) J. Biol. Chem. 276: 18748-18756). Early in Alzheimer's pathology, physiological changes are visible in the cingulate cortex (Minoshima, S. et al. (1997) Annals of Neurology 42 : 85- 94). In subjects with advanced Alzheimer's disease, accumulating plaques damage the neuronal architecture in limbic areas and eventually cripple the memory process.

Cancer Cancers are characterized by continuous or uncontrolled cell proliferation. Some cancers are associated with suppression of normal apoptotic cell death. Understanding of the neoplastic process can be aided by the identification of molecular markers of prognostic and diagnostic importance.

Cancers are associated with oncoproteins which are capable of transforming normal cells into malignant cells. Some oncoproteins are mutant isoforms of the normal protein while others are abnormally expressed with respect to location or level of expression. Normal cell proliferation begins with binding of a growth factor to its receptor on the cell membrane, resulting in activation of a signal system that induces and activates nuclear regulatory factors to initiate DNA transcription, subsequently leading to cell division. Classes of oncoproteins known to affect the cell cycle controls

include growth factors, growth factor receptors, intracellular signal transducers, nuclear transcription factors, and cell-cycle control proteins. Several types of cancer-specific genetic markers, such as tumor antigens and tumor suppressors, have also been identified.

Oncogenes Oncoproteins are encoded by genes, called oncogenes, that are derived from genes that normally control cell growth and development. Many oncogenes have been identified and characterized. These include growth factors such as sis, receptors such as erbA, erbB, neu, and ros, intracellular receptors such as src, yes, fps, abl, and met, protein-serine/threonine kinases such as znos and raf, nuclear transcription factors such as jun, fqs, myc, N-711 : yc, ntyb, ski and rel, cell cycle control proteins such as RB and p53, mutated tumor-suppressor genes such as mdm2, Cipl, pl6, and cyclin D, ras, set, can, sec, and gag RIO.

Viral oncogenes are integrated into the human genome after infection of human cells by certain viruses. Examples of viral oncogenes include v-src, v-abl, and v-fps. Transformation of normal genes to oncogenes may also occur by chromosomal translocation. The Philadelphia chromosome, characteristic of chronic myeloid leukemia and a subset of acute lymphoblastic leukemias, results from a reciprocal translocation between chromosomes 9 and 22 that moves a truncated portion of the proto-oncogene c-abl to the breakpoint cluster region (bcr) on chromosome 22. The hybrid c-abl-bcr gene encodes a chimeric protein that has tyrosine kinase activity. In chronic myeloid leukemia, the chimeric protein has a molecular weight of 210 kd, whereas in acute leukemias a more active 180 kd tyrosine kinase is formed (Robbins, S. L. et al. (1994) Pathologic Basis of Disease, W. B. Saunders Co. , Philadelphia PA).

The Ras superfamily of small GTPases is involved in the regulation of a wide range of cellular signaling pathways. Ras family proteins are membrane-associated proteins acting as molecular switches that bind GTP and GDP, hydrolyzing GTP to GDP. In the active GTP-bound state Ras family proteins interact with a variety of cellular targets to activate downstream signaling pathways. For example, members of the Ras subfamily are essential in transducing signals from receptor tyrosine kinases (RTKs) to a series of serine/threonine kinases which control cell growth and differentiation. Activated Ras genes were initially found in human cancers and subsequent studies confirmed that Ras function is critical in the determination of whether cells continue to grow or become terminally differentiated (Barbacid, M. (1987) Annu. Rev. Biochem. 56: 779-827; Treisman, R. (1994) Curr. Opin. Genet. Dev. 4: 96-98). Mutant Ras proteins, which bind but can not hydrolyze GTP, are permanently activated, and cause continuous cell proliferation or cancer.

Activation of Ras family proteins is catalyzed by guanine nucleotide exchange factors (GEFs) which catalyze the dissociation of bound GDP and subsequent binding of GTP. A recently discovered RaIGEF-like protein, RGL3, interacts with both Ras and the related protein Rit.

Constitutively active Rit, like Ras, can induce oncogenic transformation, although since Rit fails to interact with most known Ras effector proteins, novel cellular targets may be involved in Rit transforming activity. RGL3 interacts with both Ras and Rit, and thus may act as a downstream effector for these proteins (Shao, H. and D. A. Andres (2000) J. Biol. Chem. 275: 26914-26924).

Tumor antigens Tumor antigens are cell surface molecules that are differentially expressed in tumor cells relative to non-tumor tissues. Tumor antigens make tumor cells immunologically distinct from normal cells and are potential diagnostics for human cancers. Several monoclonal antibodies have been identified which react specifically with cancerous cells such as T-cell acute lymphoblastic leukemia and neuroblastoma (Minegishi, M. et al. (1989) Leukemia Res. 13: 43-51; Takagi, S. et al.

(1995) Int. J. Cancer 61: 706-715). In addition, the discovery of high level expression of the HER2 gene in breast tumors has led to the development of therapeutic treatments (Liu, E. et al. (1992) Oncogene 7: 1027-1032; Kern, J. A. (1993) Am. J. Respir. Cell Mol. Biol. 9: 448-454). Tumor antigens are found on the cell surface and have been characterized either as membrane proteins or glycoproteins. For example, MAGE genes encode a family of tumor antigens recognized on melanoma cell surfaces by autologous cytolytic T lymphocytes. Among the 12 human MAGE genes isolated, half are differentially expressed in tumors of various histological types (De Plaen, E. et al.

(1994) Immunogenetics 40: 360-369). None of the 12 MAGE genes, however, is expressed in healthy tissues except testis and placenta.

Tumor suppressors Tumor suppressor genes are generally defined as genetic elements whose loss or inactivation contributes to the deregulation of cell proliferation and the pathogenesis and progression of cancer.

Tumor suppressor genes normally function to control or inhibit cell growth in response to stress and to limit the proliferative life span of the cell. Several tumor suppressor genes have been identified including the genes encoding the retinoblastoma (Rb) protein, p53, and the breast cancer 1 and 2 proteins (BRCA1 and BRCA2). Mutations in these genes are associated with acquired and inherited genetic predisposition to the development of certain cancers.

The role of p53 in the pathogenesis of cancer has been extensively studied. (Reviewed in Aggarwal, M. L. et al. (1998) J. Biol. Chem. 273: 1-4; Levine, A. (1997) Cell 88: 323-331. ) About 50% of all human cancers contain mutations in the p53 gene. These mutations result in either the absence of functional p53 or, more commonly, a defective form of p53 which is overexpressed. p53 is a transcription factor that contains a central core domain required for DNA binding. Most cancer- associated mutations in p53 localize to this domain. In normal proliferating cells, p53 is expressed at low levels and is rapidly degraded. p53 expression and activity is induced in response to DNA damage, abortive mitosis, and other stressful stimuli. In these instances, p53 induces apoptosis or

arrests cell growth until the stress is removed. Downstream effectors of p53 activity include apoptosis-specific proteins and cell cycle regulatory proteins, including Rb, oncogene products, cyclins, and cell cycle-dependent kinases.

The metastasis-suppressor gene KAI1 (CD82) has been reported to be related to the tumor suppressor gene p53. KAI1 is involved in the progression of human prostatic cancer and possibly lung and breast cancers when expression is decreased. KAI1 encodes a member of a structurally distinct family of leukocyte surface glycoproteins. The family is known as either the tetraspan transmembrane protein family or transmembrane 4 superfamily (TM4SF) as the members of this family span the plasma membrane four times. The family is composed of integral membrane proteins having a N-terminal membrane-anchoring domain which functions as both a membrane anchor and a translocation signal during protein biosynthesis. The N-terminal membrane-anchoring domain is not cleaved during biosynthesis. TM4SF proteins have three additional transmembrane regions, seven or more conserved cysteine residues, are similar in size (218 to 284 residues), and all have a large extracellular hydrophilic domain with three potential N-glycosylation sites. The promoter region contains many putative binding motifs for various transcription factors, including five AP2 sites and nine SpI sites. Gene structure comparisons of KAI1 and seven other members of the TM4SF indicate that the splicing sites relative to the different structural domains of the predicted proteins are conserved. This suggests that these genes are related evolutionarily and arose through gene duplication and divergent evolution (Levy, S. et al. (1991) J. Biol. Chem. 266: 14597-14602; Dong, J. T. et al. (1995) Science 268: 884-886; Dong, J. T. et al. , (1997) Genomics 41: 25-32).

The Leucine-rich gene-Glioma Inactivated (LGI1) protein shares homology with a number of transmembrane and extracellular proteins which function as receptors and adhesion proteins. LGI1 is encoded by an LLR (leucine-rich, repeat-containing) gene and maps to 10q24. LGI1 has four LLRs which are flanked by cysteine-rich regions and one transmembrane domain (Somerville, R. P. et al.

(2000) Mamm. Genome 11: 622-627). LGI1 expression is seen predominantly in neural tissues, especially brain. The loss of tumor suppressor activity is seen in the inactivation of the LGI1 protein which occurs during the transition from low to high-grade tumors in malignant gliomas. The reduction of LGI1 expression in low grade brain tumors and its significant reduction or absence of expression in malignant gliomas suggests that it could be used for diagnosis of glial tumor progression (Chernova, O. B. et al. (1998) Oncogene 17: 2873-2881).

The ST13 tumor suppressor was identified in a screen for factors related to colorectal carcinomas by subtractive hybridization between cDNA of normal mucosal tissues and mRNA of colorectal carcinoma tissues (Cao, J. et al. (1997) J. Cancer Res. Clin. Oncol. 123: 447-451). ST13 is down-regulated in human colorectal carcinomas.

Mutations in the von Hippel-Lindau (VHL) tumor suppressor gene are associated with retinal

and central nervous system hemangioblastomas, clear cell renal carcinomas, and pheochromocytomas (Hoffman, M. et al. (2001) Hum. Mol. Genet. 10: 1019-1027; Kamada, M. (2001) Cancer Res.

61: 4184-4189). Tumor progression is linked to defects or inactivation of the VHL gene. VHL regulates the expression of transforming growth factor-a, the GLUT-1 glucose transporter and vascular endothelial growth factor. The VHL protein associates with elongin B, elongin C, Cul2 and Rbxl to form a complex that regulates the transcriptional activator hypoxia-inducible factor (HIF).

HIF induces genes involved in angiogenesis such as vascular endothelial growth factor and platelet- derived growth factor B. Loss of control of HIF caused by defects in VHL results in the excessive production of angiogenic peptides. VHL may play roles in inhibition of angiogenesis, cell cycle control, fibronectin matrix assembly, cell adhesion, and proteolysis.

Mutations in tumor suppressor genes are a common feature of many cancers and often appear to affect a critical step in the pathogenesis and progression of tumors. Accordingly, Chang, F. et al.

(1995; J. Clin. Oncol. 13: 1009-1022) suggest that it may be possible to use either the gene or an antibody to the expressed protein 1) to screen patients at increased risk for cancer, 2) to aid in diagnosis made by traditional methods, and 3) to assess the prognosis of individual cancer patients.

In addition, Hamada, K. et al. (1996; Cancer Res. 56: 3047-3054) are investigating the introduction of p53 into cervical cancer cells via an adenoviral vector as an experimental therapy for cervical cancer.

The PR-domain genes were recently recognized as playing a role in human tumorigenesis.

PR-domain genes normally produce two protein products: the PR-plus product, which contains the PR domain, and the PR-minus product which lacks this domain. In cancer cells, PR-plus is disrupted or overexpressed, while PR-minus is present or overexpressed. The imbalance in the amount of these two proteins appears to be an important cause of malignancy (Jiang, G. L. and S. Huang (2000) Histol.

Histopathol. 15: 109-117).

Many neoplastic disorders in humans can be attributed to inappropriate gene transcription.

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. An important class of transcriptional regulators are the zinc finger proteins. The zinc finger motif, which binds zinc ions, generally contains tandem repeats of about 30 amino acids consisting of periodically spaced cysteine and histidine residues. Examples of this sequence pattern include the C2H2-type, C4-type, and C3HC4- type zinc fingers, and the PHD domain (Lewin, B. (1990) Genes IV, Oxford University Press, New York, NY, and Cell Press, Cambridge, MA, pp. 554-570; Aasland, R. , et al. (1995) Trends Biochem.

Sci. 20: 56-59). One clinically relevant zinc-finger protein is WT1, a tumor-suppressor protein that is inactivated in children with Wilm's tumor. The oncogene bcl-6, which plays an important role in

large-cell lymphoma, is also a zinc-finger protein (Papavassiliou, A. G. (1995) N. Engl. J. Med.

332: 45-47).

Tumor responsive proteins Cancers, also called neoplasias, are characterized by continuous and uncontrolled cell proliferation. They can be divided into three categories: carcinomas, sarcomas, and leukemias.

Carcinomas are malignant growths of soft epithelial cells that may infiltrate surrounding tissues and give rise to metastatic tumors. Sarcomas may be of epithelial origin or arise from connective tissue.

Leukemias are progressive malignancies of blood-forming tissue characterized by proliferation of leukocytes and their precursors, and may be classified as myelogenous (granulocyte-or monocyte- derived) or lymphocytic (lymphocyte-derived). Tumorigenesis refers to the progression of a tumor's growth from its inception. Malignant cells may be quite similar to normal cells within the tissue of origin or may be undifferentiated (anaplastic). Tumor cells may possess few nuclei or one large polymorphic nucleus. Anaplastic cells may grow in a disorganized mass that is poorly vascularized and as a result contains large areas of ischemic necrosis. Differentiated neoplastic cells may secrete the same proteins as the tissue of origin. Cancers grow, infiltrate, invade, and destroy the surrounding tissue through direct seeding of body cavities or surfaces, through lymphatic spread, or through hematogenous spread. Cancer remains a major public health concern and current preventative measures and treatments do not match the needs of most patients. Understanding of the neoplastic process of tumorigenesis can be aided by the identification of molecular markers of prognostic and diagnostic importance.

Current forms of cancer treatment include the use of immunosuppressive drugs (Morisaki, T. et al. (2000) Anticancer Res. 20: 3363-3373; Geoerger, B. et al. (2001) Cancer Res. 61: 1527-1532).

The identification of proteins involved in cell signaling, and specifically proteins that act as receptors for immunosuppressant drugs, may facilitate the development of anti-tumor agents. For example, immunophilins are a family of conserved proteins found in both prokaryotes and eukaryotes that bind to immunosuppressive drugs with varying degrees of specificity. One such group of immunophilic proteins is the peptidyl-prolyl cis-trans isomerase (EC 5.2. 1. 8) family (PPIase, rotamase). These enzymes, first isolated from porcine kidney cortex, accelerate protein folding by catalyzing the cis- trans isomerization of proline imidic peptide bonds in oligopeptides (Fischer, G. and F. X. Schmid (1990) Biochemistry 29: 2205-2212). Included within the immunophilin family are the cyclophilins (e. g. , peptidyl-prolyl isomerase A or PPIA) and FK-binding protein (e. g., FKBP) subfamilies.

Cyclophilins are multifunctional receptor proteins which participate in signal transduction activities, including those mediated by cyclosporin (or cyclosporine). The PPIase domain of each family is highly conserved between species. Although structurally distinct, these multifunctional receptor proteins are involved in numerous signal transduction pathways, and have been implicated in folding

and trafficking events.

The immunophilin protein cyclophilin binds to the immunosuppressant drug cyclosporin A.

FKBP, another immunophilin, binds to FK506 (or rapamycin). Rapamycin is an immunosuppressant agent that arrests cells in the Gl phase of growth, inducing apoptosis. Like cyclophilin, this macrolide antibiotic (produced by Streptomyces tsukubaensis) acts by binding to ubiquitous, predominantly cytosolic immunophilin receptors. These immunophilin/immunosuppressant complexes (e. g., cyclophilin A/cyclosporin A (CypA/CsA) and FKBP12/FK506) achieve their therapeutic results through inhibition of the phosphatase calcineurin, a calcium/calmodulin-dependent protein kinase that participates in T-cell activation (Hamilton, G. S. and J. P. Steiner (1998) J. Med. Chem. 41: 5119- 5143). The murine fkbp51 gene is abundantly expressed in immunological tissues, including the thymus and T lymphocytes (Baughman, G. et al. (1995) Molec. Cell. Biol. 15: 4395-4402).

FKBP12/rapamycin-directed immunosuppression occurs through binding to TOR (yeast) or FRAP (FKBP12-rapamycin-associated protein, in mammalian cells), the kinase target of rapamycin essential for maintaining normal cellular growth patterns. Dysfunctional TOR signaling has been linked to various human disorders including cancer (Metcalfe, S. M. et al. (1997) Oncogene 15: 1635-1642; Emami, S. et al. (2001) FASEB J. 15: 351-361), and autoimmunity (Damoiseaux, J. G. et al. (1996) Transplantation 62: 994-1001).

Several cyclophilin isozymes have been identified, including cyclophilin B, cyclophilin C, mitochondrial matrix cyclophilin, bacterial cytosolic and periplasmic PPIases, and natural-killer cell cyclophilin-related protein possessing a cyclophilin-type PPIase domain, a putative tumor-recognition complex involved in the function of natural killer (NK) cells. These cells participate in the innate cellular immune response by lysing virally-infected cells or transformed cells. NK cells specifically target cells that have lost their expression of major histocompatibility complex (MHC) class I genes (common during tumorigenesis), endowing them with the potential for attenuating tumor growth. A 150-kDa molecule has been identified on the surface of human NK cells that possesses a domain which is highly homologous to cyclophilin/peptidyl-prolyl cis-trans isomerase. This cyclophilin-type protein may be a component of a putative tumor-recognition complex, a NK tumor recognition sequence (NK-TR) (Anderson, S. K. et al. (1993) Proc. Natl. Acad. Sci. USA 90: 542-546). The NKTR tumor recognition sequence mediates recognition between tumor cells and large granular lymphocytes (LGLs), a subpopulation of white blood cells (comprised of activated cytotoxic T cells and natural killer cells) capable of destroying tumor targets. The protein product of the NKTR gene presents on the surface of LGLs and facilitates binding to tumor targets. More recently, a mouse Nktr gene and promoter region have been located on chromosome 9. The gene encodes a NK-cell-specific 150-kDa protein (NK-TR) that is homologous to cyclophilin and other tumor-responsive proteins (Simons-Evelyn, M. et al. (1997) Genomics 40: 94-100).

Other proteins that interact with tumorigenic tissue include cytokines such as tumor necrosis factor (TNF). The TNF family of cytokines are produced by lymphocytes and macrophages, and can cause the lysis of transformed (tumor) endothelial cells. Endothelial protein 1 (Edpl) has been identified as a human gene activated transcriptionally by TNF-alpha in endothelial cells, and a TNF- alpha inducible Edpl gene has been identified in the mouse (Swift, S. et al. (1998) Biochim. Biophys.

Acta 1442: 394-398).

Expression profiling Microarrays are analytical tools used in bioanalysis. A microarray has a plurality of molecules spatially distributed over, and stably associated with, the surface of a solid support.

Microarrays of polypeptides, polynucleotides, and/or antibodies have been developed and find use in a variety of applications, such as gene sequencing, monitoring gene expression, gene mapping, bacterial identification, drug discovery, and combinatorial chemistry.

One area in particular in which microarrays find use is in gene expression analysis. 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.

Osteosarcoma is a malignant primary neoplasm of bone composed of a malignant connective tissue stroma with evidence of malignant, osteoid, bone, or cartilage formation. Classical osteosarcoma is a poorly differentiated tumor affecting mainly young adults, most often involving the long bones, and is classified as osteoblastic, chondroblastic, or fibroblastic according to which histologic component predominates.

Breast cancer is a genetic disease commonly caused by mutations in breast epithelial cells.

Mutations in two genes, BRCA1 and BRCA2, are known to greatly predispose a woman to breast cancer and may be passed on from parents to children (Gish, K. (1999) AWIS Magazine 28: 7-10).

However, this type of hereditary breast cancer accounts for only about 5% to 9% of breast cancers, while the vast majority of breast cancer is due to noninherited mutations that occur in breast epithelial cells.

A good deal is already known about the expression of specific genes associated with breast cancer. For example, the relationship between expression of epidermal growth factor (EGF) and its receptor, EGFR, to human mammary carcinoma has been particularly well studied. (See Khazaie, K. et al. (1993) Cancer and Metastasis Rev. 12 : 255-274), and references cited therein for a review of this

area. ) Overexpression of EGFR, particularly coupled with down-regulation of the estrogen receptor, is a marker of poor prognosis in breast cancer patients. In addition, EGFR expression in breast tumor metastases is frequently elevated relative to the primary tumor, suggesting that EGFR is involved in tumor progression and metastasis. This is supported by accumulating evidence that EGF has effects on cell functions related to metastatic potential, such as cell motility, chemotaxis, secretion and differentiation. Changes in expression of other members of the erbB receptor family, of which EGFR is one, have also been implicated in breast cancer. The abundance of erbB receptors, such as HER- 2/neu, HER-3, and HER-4, and their ligands in breast cancer points to their functional importance in the pathogenesis of the disease, and may therefore provide targets for therapy of the disease (Bacus, S. S. et al. (1994) Am. J. Clin. Pathol. 102: S13-S24). Other known markers of breast cancer include a human secreted frizzled protein mRNA that is downregulated in breast tumors; the matrix Gla protein which is overexpressed is human breast carcinoma cells; Drgl or RTP, a gene whose expression is diminished in colon, breast, and prostate tumors; maspin, a tumor suppressor gene downregulated in invasive breast carcinomas; and CaN19, a member of the S100 protein family, all of which are down regulated in mammary carcinoma cells relative to normal mammary epithelial cells (Zhou Z. et al. (1998) Int. J. Cancer 78: 95-99; Chen, L. et al. (1990) Oncogene 5: 1391-1395; Ulrix W. et al (1999) FEBS Lett. 455: 23-26; Sager, R. et al. (1996) Curr. Top. Microbiol. Immunol. 213: 51-64; and Lee, S. W. et al. (1992) Proc. Natl. Acad. Sci. USA 89: 2504-2508).

Prostate cancer is a common malignancy in men over the age of 50, and the incidence increases with age. In the US, there are approximately 132,000 newly diagnosed cases of prostate cancer and more than 33,000 deaths from the disorder each year.

Once cancer cells arise in the prostate, they are stimulated by testosterone to a more rapid growth. Thus, removal of the testes can indirectly reduce both rapid growth and metastasis of the cancer. Over 95 percent of prostatic cancers are adenocarcinomas which originate in the prostatic acini. The remaining 5 percent are divided between squamous cell and transitional cell carcinomas, both of which arise in the prostatic ducts or other parts of the prostate gland.

As with most cancers, prostate cancer develops through a multistage progression ultimately resulting in an aggressive, metastatic phenotype. The initial step in tumor progression involves the hyperproliferation of normal luminal and/or basal epithelial cells that become hyperplastic and evolve into early-stage tumors. The early-stage tumors are localized in the prostate but eventually may metastasize, particularly to the bone, brain or lung. About 80% of these tumors remain responsive to androgen treatment, an important hormone controlling the growth of prostate epithelial cells.

However, in its most advanced state, cancer growth becomes androgen-independent and there is currently no known treatment for this condition.

A primary diagnostic marker for prostate cancer is prostate specific antigen (PSA). PSA is a

tissue-specific serine protease almost exclusively produced by prostatic epithelial cells. The quantity of PSA correlates with the number and volume of the prostatic epithelial cells, and consequently, the levels of PSA are an excellent indicator of abnormal prostate growth. Men with prostate cancer exhibit an early linear increase in PSA levels followed by an exponential increase prior to diagnosis.

However, since PSA levels are also influenced by factors such as inflammation, androgen and other growth factors, some scientists maintain that changes in PSA levels are not useful in detecting individual cases of prostate cancer.

Current areas of cancer research provide additional prospects for markers as well as potential therapeutic targets for prostate cancer. Several growth factors have been shown to play a critical role in tumor development, growth, and progression. The growth factors Epidermal Growth Factor (EGF), Pibroblast Growth Factor (FGF), and Tumor Growth Factor alpha (TGFa) are important in the growth of normal as well as hyperproliferative prostate epithelial cells, particularly at early stages of tumor development and progression, and affect signaling pathways in these cells in various ways (Lin J et al. (1999) Cancer Res. 59: 2891-2897; Putz T et al. (1999) Cancer Res 59: 227-233). The TGF-p family of growth factors are generally expressed at increased levels in human cancers and the high expression levels in many cases correlates with advanced stages of malignancy and poor survival (Gold LI (1999) Crit Rev Oncog 10: 303-360). Finally, there are human cell lines representing both the androgen-dependent stage of prostate cancer (LNCap) as well as the androgen-independent, hormone refractory stage of the disease (PC3 and DU-145) that have proven useful in studying gene expression patterns associated with the progression of prostate cancer, and the effects of cell treatments on these expressed genes (Chung TD (1999) Prostate 15: 199-207).

Ovarian cancer is the leading cause of death from a gynecologic cancer. The majority of ovarian cancers are derived from epithelial cells, and 70% of patients with epithelial ovarian cancers present with late-stage disease. As a result, the long-term survival rates for this disease is very low.

Identification of early-stage markers for ovarian cancer would significantly increase the survival rate.

Genetic variations involved in ovarian cancer development include mutation of p53 and microsatellite instability. Gene expression patterns likely vary when normal ovary is compared to ovarian tumors.

Tangier disease (TD) is a genetic disorder characterized by the near absence of circulating high density lipoprotein (HDL) and the accumulation of cholesterol esters in many tissues, including tonsils, lymph nodes, liver, spleen, thymus, and intestine. Low levels of HDL represent a clear predictor of premature coronary artery disease and homozygous TD correlates with a four-to six-fold increase in cardiovascular disease compared to controls. HDL plays a cardio-protective role in reverse cholesterol transport, the flux of cholesterol from peripheral cells such as tissue macrophages through plasma lipoproteins to the liver. The HDL protein, apolipoprotein A-1, plays a major role in this process, interacting with the cell surface to remove excess cholesterol and phospholipids. This

pathway is severely impaired in TD and the defect lies in a specific gene, the ABC1 transporter. This gene is a member of the family of ATP-binding cassette transporters, which utilize ATP hydrolysis to transport a variety of substrates across membranes.

Gemfibrozil is a fibric acid antilipemic agent that lowers serum triglycerides and produces favorable changes in lipoproteins. Gemfibrozil is effective in reducing the risk of coronary heart disease in men (Frick, M. H. , et al. (1987) New Engl. J. Med; 317: 1237-1245). The compound can inhibit peripheral lipolysis and decrease hepatic extraction of free fatty acids, which decreases hepatic triglyceride production. Gemfibrozil also inhibits the synthesis and increases the clearance of apolipoprotein B, a carrier molecule for VLDL. Gemfibrozil has variable effects on LDL cholesterol.

Although it causes moderate reductions in patients with type IIa hyperlipoproteinemia, changes in patients with either type Db or type IV hyperlipoproteinemia are unpredictable. In general, the HMG- CoA reductase inhibitors are more effective than gemfibrozil in reducing LDL cholesterol. At the molecular level gemfibozil may function as a peroxisome proliferator-activated receptor (PPAR) agonist. Gemfibrozil is rapidly and completely absorbed from the GI tract and undergoes enterohepatic recirculation. Gemfibrozil is metabolized by the liver and excreted by the kidneys, mainly as metabolites, one of which possesses pharmacologic activity. Gemfibozil causes peroxisome proliferation and hepatocarcinogenesis in rats, which is a cause for concern generally for fibric acid derivative drugs. In humans, fibric acid derivatives are known to increase the risk of gall bladder disease although gemfibrozil is better tolerated than other fibrates. The relative safety of gemfibrozil in humans compared to rodent species including rats may be attributed to differences in metabolism and clearance of the compound in different species (Dix, K. J. , et al. (1999) Drug Metab.

Distrib. 27: 138-146 ; Thomas, B. F. , et al. (1999) Drug Metab. Distrib. 27: 147-157).

The human C3A cell line is a clonal derivative of HepG2/C3 (hepatoma cell line, isolated from a 15-year-old male with liver tumor), which was selected for strong contact inhibition of growth.

The use of a clonal population enhances the reproducibility of the cells. C3A cells have many characteristics of primary human hepatocytes in culture: i) expression of insulin receptor and insulin-like growth factor II receptor; ii) secretion of a high ratio of serum albumin compared with a- fetoprotein; iii) conversion of ammonia to urea and glutamin ; iv) metabolism of aromatic amino acids; and v) proliferation in glucose-free and insulin-free medium. The C3A cell line is now well established as an in vitro model of the mature human liver (Mickelson et al. (1995) Hepatology 22: 866-875 ; Nagendra et al. (1997) Am. J. Physiol. 272: G408-G416).

Colorectal cancer is the second leading cause of cancer deaths in the United States, and is thought to be a disease of aging since 90% of the total cases occur in individuals over the age of 55. A widely accepted hypothesis is that several mutations must accumulate over time in an individual who develops the disease.

While soft tissue sarcomas are relatively rare, more than 50% of new patients diagnosed with the disease will die from it. The molecular pathways leading to the development of sarcomas are relatively unknown, due to the rarity of the disease and variation in pathology. Colon cancer evolves through a multi-step process whereby pre-malignant colonocytes undergo a relatively defined sequence of events leading to tumor formation. Several factors participate in the process of tumor progression and malignant transformation including genetic factors, mutations, and selection.

To understand the nature of gene alterations in colorectal cancer, a number of studies have focused on the inherited syndromes. The first, Familial Adenomatous Polyposis (FAP), is caused by mutations in the Adenomatous Polyposis Coli gene (APC), resulting in truncated or inactive forms of the protein. This tumor suppressor gene has been mapped to chromosome 5q. The second known inherited syndrome is hereditary nonpolyposis colorectal cancer (HNPCC), which is caused by mutations in mismatch repair genes.

Although hereditary colon cancer syndromes occur in a small percentage of the population, and most colorectal cancers are considered sporadic, knowledge from studies of the hereditary syndromes can be applied broadly. For instance, somatic mutations in APC occur in at least 80% of sporadic colon tumors. APC mutations are thought to be the initiating event in disease progression.

Other mutations occur subsequently. Approximately 50% of colorectal cancers contain activating mutations in ras, while 85% contain inactivating mutations in p53. Changes in all of these genes lead to gene expression changes in colon cancer. Less is understood about downstream targets of these mutations and the role they may play in cancer development and progression.

There is a need in the art for new compositions, including nucleic acids and proteins, for the diagnosis, prevention, and treatment of cell proliferative disorders including cancer, developmental disorders, neurological disorders, autoimmune/inflammatory disorders, reproductive disorders, and disorders of the placenta.

SUMMARY OF THE INVENTION Various embodiments of the invention provide purified polypeptides, proteins associated with cell growth, differentiation, and death, referred to collectively as'CGDD'and individually as <BR> <BR> <BR> <BR> 'CGDD-1,''CGDD-2,''CGDD-3,''CGDD-4,''CGDD-5,''CGDD-6,''CGDD- 7,''CGDD-8,'<BR> <BR> <BR> <BR> <BR> <BR> <BR> CGDD-9,'CGDD-10,'CGDD-11,'CGDD-12,'CGDD-13,'CGDD-I4,'CGDD-15 ,'CGDD-<BR> <BR> <BR> <BR> <BR> <BR> 16,''CGDD-17,''CGDD-18,''CGDD-19,''CGDD-20,''CGDD-21,''CGDD- 22,''CGDD-23,'<BR> <BR> <BR> <BR> <BR> <BR> 'CGDD-24,''CGDD-25,''CGDD-26,''CGDD-27,''CGDD-28,''CGDD-29,' 'CGDD-30,''CGDD- 31,''CGDD-32,'and'CGDD-33'and methods for using these proteins and their encoding polynucleotides for the detection, diagnosis, and treatment of diseases and medical conditions.

Embodiments also provide methods for utilizing the purified proteins associated with cell growth,

differentiation, and death and/or their encoding polynucleotides for facilitating the drug discovery process, including determination of efficacy, dosage, toxicity, and pharmacology. Related embodiments provide methods for utilizing the purified proteins associated with cell growth, differentiation, and death and/or their encoding polynucleotides for investigating the pathogenesis of diseases and medical conditions.

An embodiment 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- 33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. Another embodiment provides an isolated polypeptide comprising an amino acid sequence of SEQ ID NO : 1-33.

Still another embodiment 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: 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33. In another embodiment, the polynucleotide encodes a polypeptide selected from the group consisting of SEQ ID NO: 1-33. In an alternative embodiment, the polynucleotide is selected from the group consisting of SEQ ID NO : 34-66.

Still another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. Another embodiment provides a cell transformed with the recombinant polynucleotide.

Yet another embodiment provides a transgenic organism comprising the recombinant polynucleotide.

Another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. 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.

Yet another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33.

Still yet another embodiment 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 : 34-66, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical or at least about 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, 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 other embodiments, the polynucleotide can comprise at least about 20,30, 40, 60,80, or 100 contiguous nucleotides.

Yet another embodiment provides a method for detecting a target polynucleotide in a sample, said target polynucleotide being selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical or at least about 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, 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. In a related embodiment, the method can include detecting the amount of the hybridization complex. In still other embodiments, the probe can comprise at least about 20,30, 40,60, 80, or 100 contiguous nucleotides.

Still yet another embodiment provides a method for detecting a target polynucleotide in a sample, said target polynucleotide being selected from the group consisting of a) a polynucleotide comprising a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, b) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical or at least about 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, 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. In a related embodiment, the method can include detecting the amount of the amplified target polynucleotide or fragment thereof.

Another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and a pharmaceutically acceptable excipient.

In one embodiment, the composition can comprise an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. Other embodiments provide a method of treating a disease or condition associated with decreased or abnormal expression of functional CGDD, comprising administering to a patient in need of such treatment the composition.

Yet another embodiment 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: 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence

selected from the group consisting of SEQ ID NO : 1-33. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting agonist activity in the sample. Another embodiment provides a composition comprising an agonist compound identified by the method and a pharmaceutically acceptable excipient. Yet another embodiment provides a method of treating a disease or condition associated with decreased expression of functional CGDD, comprising administering to a patient in need of such treatment the composition.

Still yet another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. The method comprises a) exposing a sample comprising the polypeptide to a compound, and b) detecting antagonist activity in the sample. Another embodiment provides a composition comprising an antagonist compound identified by the method and a pharmaceutically acceptable excipient. Yet another embodiment provides a method of treating a disease or condition associated with overexpression of functional CGDD, comprising administering to a patient in need of such treatment the composition.

Another embodiment 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: 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, c) a biologically active fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. 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.

Yet another embodiment 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 : 1-33, b) a polypeptide comprising a naturally occurring amino acid sequence at least 90% identical or at least about 90% identical to an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33, c) a biologically active

fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO : 1-33, and d) an immunogenic fragment of a polypeptide having an amino acid sequence selected from the group consisting of SEQ ID NO: 1-33. 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.

Still yet another embodiment 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 : 34-66, 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.

Another embodiment 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 : 34-66, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical or at least about 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, 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 : 34-66, ii) a polynucleotide comprising a naturally occurring polynucleotide sequence at least 90% identical or at least about 90% identical to a polynucleotide sequence selected from the group consisting of SEQ ID NO : 34-66, 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 can comprise a fragment of a polynucleotide 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 full length polynucleotide and polypeptide embodiments of the 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 polypeptide embodiments 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 embodiments, 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 embodiments, along with selected fragments of the polynucleotides.

Table 5 shows representative cDNA libraries for polynucleotide embodiments.

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 polynucleotides and polypeptides, along with applicable descriptions, references, and threshold parameters.

Table 8 shows single nucleotide polymorphisms found in polynucleotide sequences of the invention, along with allele frequencies in different human populations.

DESCRIPTION OF THE INVENTION Before the present proteins, nucleic acids, and methods are described, it is understood that embodiments of the invention are not limited to the particular machines, instruments, 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 invention.

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 various embodiments of 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 "CGDD"refers to the amino acid sequences of substantially purified CGDD 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 CGDD. Agonists may include proteins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of CGDD either by directly interacting with CGDD or by acting on components of the biological pathway in which CGDD participates.

An"allelic variant"is an alternative form of the gene encoding CGDD. 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 CGDD include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as CGDD or a polypeptide with at least one functional characteristic of CGDD. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding CGDD, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide encoding CGDD. 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 CGDD.

Deliberate amino acid substitutions may be made on the basis of one or more similarities in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of CGDD 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 glutamin ; 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"can refer to an oligopeptide, a peptide, a polypeptide, or a 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.

Amplification may be carried out using polymerase chain reaction (PCR) technologies or other nucleic acid amplification technologies well known in the art.

The term"antagonist"refers to a molecule which inhibits or attenuates the biological activity of CGDD. Antagonists may include proteins such as antibodies, anticalins, nucleic acids, carbohydrates, small molecules, or any other compound or composition which modulates the activity of CGDD either by directly interacting with CGDD or by acting on components of the biological pathway in which CGDD 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 CGDD 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 (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 polynucleotide having 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 CGDD, 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"and a"composition comprising a given polypeptide"can refer to any composition containing the given polynucleotide or polypeptide. The composition may comprise a dry formulation or an aqueous solution. Compositions comprising polynucleotides encoding CGDD or fragments of CGDD may be employed as hybridization probes.

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

"Consensus sequence"refers to a nucleic acid sequence which has been subjected to repeated DNA sequence analysis to resolve uncalled bases, extended using the XL-PCR kit (Applied Biosystems, Foster City CA) in the 5'and/or the 3'direction, and resequenced, or which has been assembled from one or more overlapping cDNA, EST, or genomic DNA fragments using a computer program for fragment assembly, such as the GELVIEW fragment assembly system (Accelrys, Burlington MA) 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 He 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 CGDD or a polynucleotide encoding CGDD which can be 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 about 5 to about 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 : 34-66 can comprise a region of unique polynucleotide sequence that specifically identifies SEQ ID N0 : 34-66, for example, as distinct from any other sequence in the

genome from which the fragment was obtained. A fragment of SEQ ID NO : 34-66 can be employed in one or more embodiments of methods of the invention, for example, in hybridization and amplification technologies and in analogous methods that distinguish SEQ ID NO : 34-66 from related polynucleotides. The precise length of a fragment of SEQ ID NO : 34-66 and the region of SEQ ID NO : 34-66 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: 1-33 is encoded by a fragment of SEQ ID NO : 34-66. A fragment of SEQ ID NO : 1-33 can comprise a region of unique amino acid sequence that specifically identifies SEQ ID NO : 1-33. For example, a fragment of SEQ ID NO : 1-33 can be used as an immunogenic peptide for the development of antibodies that specifically recognize SEQ ID NO: 1-33. The precise length of a fragment of SEQ ID NO: 1-33 and the region of SEQ ID NO : 1-33 to which the fragment corresponds can be determined based on the intended purpose for the fragment using one or more analytical methods described herein or otherwise known in the art.

A"full length"polynucleotide 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, alternatively, 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 identical nucleotide 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 one or more computer algorithms or programs known in the art or described herein. For example, percent identity can 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.

Alternatively, a suite of commonly used and freely available sequence comparison algorithms which can be used 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 Penaltyfor 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 identical 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. The

phrases"percent similarity"and"% similarity, "as applied to polypeptide sequences, refer to the percentage of residue matches, including identical residue matches and conservative substitutions, between at least two polypeptide sequences aligned using a standardized algorithm. In contrast, conservative substitutions are not included in the calculation of percent identity between polypeptide sequences.

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.

Alternatively the NCBI BLAST software suite may be used. For example, for a pairwise comparison of two polypeptide sequences, one may use the"BLAST 2 Sequences"tool Version 2.0. 12 (April-21-2000) with blastp set at default parameters. Such default parameters may be, for example: Matrix : BLOSUM62 Open Gap : 11 and Extension Gap : 1 penalties Gap x drop-off. 50 Expect : 10 Word Size : 3 Filter : on Percent identity may be measured over the length of an entire defined polypeptide sequence, for example, as defined by a particular SEQ 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 , g/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 Tm and conditions for nucleic acid hybridization are well known and can be found in Sambrook, J. and D. W.

Russell (2001; Molecular Cloning : A Laboratory Manual, 3rd ed. , vol. 1-3, Cold Spring Harbor Press, Cold Spring Harbor NY, ch. 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 ug/rtlL 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 acids 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 present in solution and another nucleic acid 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 polynucleotide 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 CGDD 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 CGDD 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, antibodies, or other chemical compounds on a substrate.

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

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

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 CGDD 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 CGDD.

"Probe"refers to nucleic acids encoding CGDD, their complements, or fragments thereof, which are used to detect identical, allelic or related nucleic acids. 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, 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, for example, Sambrook, J. and D. W. Russell (2001; Molecular Cloning: A Laboratory Manual, 3rd ed. , vol. 1-3, Cold Spring Harbor Press, Cold Spring Harbor NY), Ausubel, F. M. et al. (1999; Short Protocols in Molecular Biology, 4i ed., John Wiley & Sons, New York NY), and 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 nucleic acid 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 and Russell (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 molecule, is composed of the same linear sequence of nucleotides as the reference DNA molecule 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 CGDD, nucleic acids encoding CGDD, 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 about 60% free, preferably at least about 75% free, and most preferably at least about 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 another embodiment, 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 in vitro fertilization, but rather is directed to the introduction of a recombinant DNA molecule. The transgenic organisms contemplated in accordance with the present invention include bacteria, cyanobacteria, fungi, plants and animals. The isolated DNA of the present invention can be introduced into the host by methods known in the art, for example infection, transfection, transformation or transconjugation. Techniques for transferring the DNA of the present invention into such organisms are widely known and provided in references such as Sambrook and Russell (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 during mRNA processing. The corresponding polypeptide may possess additional functional domains or lack domains that are present in the reference molecule.

Species variants are polynucleotides 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 or sequence similarity 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 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 or sequence similarity over a certain defined length of one of the polypeptides.

THE INVENTION Various embodiments of the invention include new human proteins associated with cell growth, differentiation, and death (CGDD), the polynucleotides encoding CGDD, and the use of these compositions for the diagnosis, treatment, or prevention of cell proliferative disorders including cancer, developmental disorders, neurological disorders, autoimmune/inflammatory disorders, reproductive disorders, and disorders of the placenta.

Table 1 summarizes the nomenclature for the full length polynucleotide and polypeptide embodiments 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 polypeptide embodiments 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 (Accelrys, Burlington MA).

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 proteins associated with cell growth, differentiation, and death. For example, SEQ ID NO : 2 is 100% identical, from residue Ml to residue K175, to H. sapiens cyclin H (GenBank ID gl3528978) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2. ) The BLAST probability score is 2.7e-93, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO : 2 also has homology to proteins that are localized to the nuclear region, and are components of heterotrimeric CAK (CDK-activating kinase), which is a component of the multimeric TFIIH, a complex involved in transcription and DNA repair, as determined by BLAST analysis using the PROTEOME database. SEQ ID NO : 2 also contains a domain present in cyclins, TFIIB, and retinoblastoma-associated proteins, as well as a cyclin ccll domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based TIGRFAM and SMART databases of conserved protein families/domains. (See Table 3. ) Data from BLIMPS analysis, as well as BLAST analyses against the PRODOM and DOMO databases, provide further corroborative evidence that SEQ ID NO : 2 is a cyclin H. In an alternative example, SEQ ID NO : 4 is 89% identical, from residue M1 to residue F420, to murine cell division cycle 10 (CDC10) (GenBank ID g2864606) as determined by the Basic Local Alignment Search Tool (BLAST). The BLAST probability score is 6.8e-200, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO : 4 also has homology to GTP-binding proteins that are localized to the cytoplasm and cytoskeleton, are members of the septin family of proteins that may play a role in cytokinesis, vesicle trafficking, and cytoskeletal organization, and are cell division cycle 10 related proteins, as determined by BLAST analysis using the PROTEOME database. SEQ ID NO : 4 also contains a cell division protein domain as determined by searching for statistically significant matches in the hidden Markov model (HMM) -based PFAM database of conserved protein families/domains. (See Table 3. ) Data from BLAST analyses against the PRODOM and DOMO databases, provide further corroborative evidence that SEQ ID NO : 4 is a septin. In an alternative example, SEQ ID NO: 17 is 100% identical, from residue MI to residue V263, to human type 1 septin 6 (GenBank ID gl5430743) as determined by the Basic Local Alignment Search Tool (BLAST).

(See Table 2. ) The BLAST probability score is 1.8e-137, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO: 17 also has homology to

proteins that have GTPase activity, and are GTP-binding cell division proteins resembling septins, as determined by BLAST analysis using the PROTEOME database. SEQ ID NO : 17 also contains a cell division protein domain as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM database of conserved protein families/domains. (See Table 3.) Data from MOTIFS and and BLAST analyses against the PRODOM and DOMO databases, provide further corroborative evidence that SEQ ID NO: 17 is a GTP-binding cell division protein. In an alternative example, SEQ ID NO : 25 is 92% identical, from residue L127 to residue P421, to human cell division cycle 25C (GenBank ID gl7512215) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2. ) The BLAST probability score is 1.6e-227, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO : 25 also has homology to proteins that are members of the CDC25 family of protein tyrosine phosphatases that trigger entry into mitosis, as determined by BLAST analysis using the PROTEOME database. SEQ ID NO : 25 also contains a rhodanese domain, characteristic of rhodanese/cdc25 phosphatase superfamily proteins, as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM and SMART databases of conserved protein families/domains. (See Table 3. ) Data from BLIMPS analysis, and BLAST analyses against the PRODOM and DOMO databases, provide further corroborative evidence that SEQ ID NO : 25 is a cdc25 phosphatase. In an alternative example, SEQ ID NO : 26 is 99% identical, from residue MI to residue L234, to human non-specific cross-reacting antigen (GenBank ID g 189085) as determined by the Basic Local Alignment Search Tool (BLAST). (See Table 2. ) The BLAST probability score is 1.4e-125, which indicates the probability of obtaining the observed polypeptide sequence alignment by chance. SEQ ID NO : 26 also has homology to proteins that are localized to the plasma membrane, are involved in cell adhesion and granulocyte activation, and are non-specific cross-reacting antigens of the carcinoembryonic family of antigens, as determined by BLAST analysis using the PROTEOME database. SEQ ID NO : 26 also contains immunoglobulin domains as determined by searching for statistically significant matches in the hidden Markov model (HMM)-based PFAM and SMART database of conserved protein families/domains. (See Table 3.) Data from BLIMPS and MOTIFS analyses, and BLAST analyses against the PRODOM and DOMO databases, provide further corroborative evidence that SEQ ID NO : 26 is a carcinoembryonic antigen. SEQ ID NO : 1, SEQ ID NO : 3, SEQ ID NO : 5-16, SEQ ID NO: 18-24, and SEQ ID N0 : 27-33 were analyzed and annotated in a similar manner. The algorithms and parameters for the analysis of SEQ ID NO : 1-33 are described in Table 7.

As shown in Table 4, the full length polynucleotide embodiments 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 embodiments, and of fragments of the polynucleotides which are useful, for example, in hybridization or amplification technologies that identify SEQ ID NO : 34-66 or that distinguish between SEQ ID NO : 34-66 and related polynucleotides.

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 polynucleotides. 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 FL XXXXXX N 1V2_YYYYY_N3 1V4 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, 21, 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 FLXXXXXX_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 (ie., 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 polynucleotides 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 polynucleotides. The tissues and vectors which were used to construct the cDNA libraries shown in Table 5 are described in Table 6.

Table 8 shows single nucleotide polymorphisms (SNPs) found in polynucleotide sequences of the invention, along with allele frequencies in different human populations. Columns 1 and 2 show the polynucleotide sequence identification number (SEQ ID NO:) and the corresponding Incyte project identification number (PID) for polynucleotides of the invention. Column 3 shows the Incyte identification number for the EST in which the SNP was detected (EST ID), and column 4 shows the identification number for the SNP (SNP ID). Column 5 shows the position within the EST sequence at which the SNP is located (EST SNP), and column 6 shows the position of the SNP within the full- length polynucleotide sequence (CB 1 SNP). Column 7 shows the allele found in the EST sequence.

Columns 8 and 9 show the two alleles found at the SNP site. Column 10 shows the amino acid encoded by the codon including the SNP site, based upon the allele found in the EST. Columns 11- 14 show the frequency of allele 1 in four different human populations. An entry of n/d (not detected) indicates that the frequency of allele 1 in the population was too low to be detected, while n/a (not available) indicates that the allele frequency was not determined for the population.

The invention also encompasses CGDD variants. Various embodiments of CGDD variants can have at least about 80%, at least about 90%, or at least about 95% amino acid sequence identity to the CGDD amino acid sequence, and can contain at least one functional or structural characteristic of

CGDD.

Various embodiments also encompass polynucleotides which encode CGDD. In a particular embodiment, the invention encompasses a polynucleotide sequence comprising a sequence selected from the group consisting of SEQ ID NO : 34-66, which encodes CGDD. The polynucleotide sequences of SEQ ID NO : 34-66, 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 variants of a polynucleotide encoding CGDD. In particular, such a variant polynucleotide will have at least about 70%, or alternatively at least about 85%, or even at least about 95% polynucleotide sequence identity to a polynucleotide encoding CGDD. A particular aspect of the invention encompasses a variant of a polynucleotide comprising a sequence selected from the group consisting of SEQ ID NO : 34-66 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 : 34-66. Any one of the polynucleotide variants described above can encode a polypeptide which contains at least one functional or structural characteristic of CGDD.

In addition, or in the alternative, a polynucleotide variant of the invention is a splice variant of a polynucleotide encoding CGDD. A splice variant may have portions which have significant sequence identity to a polynucleotide encoding CGDD, but will generally have a greater or lesser number of polynucleotides due to additions or deletions of blocks of sequence arising from alternate splicing 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 a polynucleotide encoding CGDD 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 encoding CGDD. For example, a polynucleotide comprising a sequence of SEQ ID NO : 38, a polynucleotide comprising a sequence of SEQ ID NO : 64, and a polynucleotide comprising a sequence of SEQ ID NO : 65 are splice variants of each other; a polynucleotide comprising a sequence of SEQ ID NO : 48 and a polynucleotide comprising a sequence of SEQ ID NO : 51 are splice variants of each other; a polynucleotide comprising a sequence of SEQ ID NO : 52 and a polynucleotide comprising a sequence of SEQ ID NO : 53 are splice variants of each other; a polynucleotide comprising a sequence of SEQ ID NO : 56, a polynucleotide comprising a sequence of SEQ ID NO : 57, and a polynucleotide comprising a sequence of SEQ ID NO : 58 are splice variants of each other; and a polynucleotide comprising a sequence of SEQ ID NO : 62 and a polynucleotide comprising a sequence of SEQ ID NO : 63 are splice variants of each other. Any one of the splice variants described above can encode a

polypeptide which contains at least one functional or structural characteristic of CGDD.

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 CGDD, 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 CGDD, and all such variations are to be considered as being specifically disclosed.

Although polynucleotides which encode CGDD and its variants are generally capable of hybridizing to polynucleotides encoding naturally occurring CGDD under appropriately selected conditions of stringency, it may be advantageous to produce polynucleotides encoding CGDD 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 CGDD 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 polynucleotides which encode CGDD and CGDD derivatives, or fragments thereof, entirely by synthetic chemistry. After production, the synthetic polynucleotide 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 polynucleotide encoding CGDD or any fragment thereof.

Embodiments of the invention can also include polynucleotides that are capable of hybridizing to the claimed polynucleotides, and, in particular, to those having the sequences shown in SEQ ID NO : 34-66 and fragments thereof, under various conditions of stringency (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 (Ausubel et al., supra, ch. 7; Meyers, R. A. (1995) Molecular Biology and Biotechnology, Wiley VCH, New York NY, pp. 856-853).

The nucleic acids encoding CGDD 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 (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 (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 (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 (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, polynucleotides or fragments thereof which encode CGDD may be cloned in recombinant DNA molecules that direct expression of CGDD, or fragments or functional equivalents thereof, in appropriate host cells. Due to the inherent degeneracy of the genetic code, other polynucleotides which encode substantially the same or a functionally equivalent polypeptides may be produced and used to express CGDD.

The polynucleotides of the invention can be engineered using methods generally known in the art in order to alter CGDD-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 CGDD, 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, polynucleotides encoding CGDD may be synthesized, in whole or in part, using one or more chemical methods well known in the art (Caruthers, M. H. et al. (1980) Nucleic Acids Symp. Ser. 7: 215-223; Horn, T. et al. (1980) Nucleic Acids Symp. Ser. 7: 225-232).

Alternatively, CGDD itself or a fragment thereof may be synthesized using chemical methods known in the art. For example, peptide synthesis can be performed using various solution-phase or solid-phase techniques (Creighton, T. (1984) Proteins, Structures and Molecular Properties, WH Freeman, New York NY, pp. 55-60; 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 CGDD, 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 (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 (Creighton, supra, pp. 28-53).

In order to express a biologically active CGDD, the polynucleotides encoding CGDD 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 polynucleotides encoding CGDD. Such elements may vary in their strength and specificity. Specific initiation signals may also be used to achieve more efficient translation of polynucleotides encoding CGDD. Such signals include the ATG initiation codon and adjacent sequences, e. g. the Kozak sequence. In cases where a polynucleotide sequence encoding CGDD 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 (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 polynucleotides encoding CGDD and appropriate transcriptional and translational control elements. These methods include it vitro recombinant DNA techniques, synthetic techniques, and iii vivo genetic recombination (Sambrook and Russell, supra, ch. 1-4, and 8; Ausubel et al.,

supra, ch. 1, 3, and 15).

A variety of expression vector/host systems may be utilized to contain and express polynucleotides encoding CGDD. 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 (Sambrook and Russell, supra ; Ausubel et al., 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; 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 polynucleotides to the targeted organ, tissue, or cell population (Di Nicola, M. et al. (1998) Cancer Gen. Ther. 5: 350-356; Yu, M. et al. (1993) Proc. Natl. Acad. Sci. USA 90: 6340-6344; Buller, R. M. et al. (1985) Nature 317: 813-815; McGregor, D. P. et al. (1994) Mol. Immunol. 31: 219-226; 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 polynucleotides encoding CGDD. For example, routine cloning, subcloning, and propagation of polynucleotides encoding CGDD can be achieved using a multifunctional E. coli vector such as PBLUESCRIPT (Stratagene, La Jolla CA) or PSPORT1 plasmid (Invitrogen). Ligation of polynucleotides encoding CGDD 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 (Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem.

264: 5503-5509). When large quantities of CGDD are needed, e. g. for the production of antibodies, vectors which direct high level expression of CGDD 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 CGDD. A number of vectors containing constitutive or inducible promoters, such as alpha factor, alcohol oxidase, and PGH promoters, may be used in the yeast Saccharomyces cerevisiae or Pichia pastors. In addition, such vectors direct either the secretion or intracellular retention of expressed proteins and enable

integration of foreign polynucleotide sequences into the host genome for stable propagation (Ausubel et al., supra ; Bitter, G. A. et al. (1987) Methods Enzymol. 153: 516-544; Scorer, C. A. et al. (1994) Bio/Technology 12: 181-184).

Plant systems may also be used for expression of CGDD. Transcription of polynucleotides encoding CGDD 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 (Coruzzi, G. et al. (1984) EMBO J. 3: 1671-1680; Broglie, R. et al. (1984) Science 224: 838-843; 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 (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, polynucleotides encoding CGDD 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 CGDD in host cells (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 (Harrington, J. J. et al. (1997) Nat. Genet. 15: 345-355).

For long term production of recombinant proteins in mammalian systems, stable expression of CGDD in cell lines is preferred. For example, polynucleotides encoding CGDD 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 (Wigler, M. et al. (1977) Cell 11: 223-232; Lowy, 1. 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; 71eo confers resistance to the aminoglycosides neomycin and G-418; and als and pat confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (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 (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), ß- glucuronidase and its substrate p-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 (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 CGDD is inserted within a marker gene sequence, transformed cells containing polynucleotides encoding CGDD can be identified by the absence of marker gene function.

Alternatively, a marker gene can be placed in tandem with a sequence encoding CGDD 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 polynucleotide encoding CGDD and that express CGDD 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 CGDD 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 CGDD is preferred, but a competitive binding assay may be employed. These and other assays are well known in the art (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; 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 CGDD include oligolabeling, nick translation, end-labeling, or PCR amplification using a labeled nucleotide.

Alternatively, polynucleotides encoding CGDD, 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 polynucleotides encoding CGDD 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 CGDD may be designed to contain signal sequences which direct secretion of CGDD 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 polynucleotides 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 polynucleotides encoding CGDD 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 CGDD protein containing a heterologous moiety that can be recognized by a commercially available antibody may facilitate the screening of peptide libraries for inhibitors of CGDD 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 CGDD encoding sequence and the heterologous protein sequence, so that CGDD may be cleaved away from the heterologous moiety following purification. Methods for fusion protein expression and purification are discussed in Ausubel et al. (supra, ch. 10 and 16).

A variety of commercially available kits may also be used to facilitate expression and purification of fusion proteins.

In another embodiment, synthesis of radiolabeled CGDD 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.

CGDD, fragments of CGDD, or variants of CGDD may be used to screen for compounds that specifically bind to CGDD. One or more test compounds may be screened for specific binding to CGDD. In various embodiments, 1,2, 3,4, 5,10, 20,50, 100, or 200 test compounds can be screened for specific binding to CGDD. Examples of test compounds can include antibodies, anticalins, oligonucleotides, proteins (e. g. , ligands or receptors), or small molecules.

In related embodiments, variants of CGDD can be used to screen for binding of test compounds, such as antibodies, to CGDD, a variant of CGDD, or a combination of CGDD and/or one or more variants CGDD. In an embodiment, a variant of CGDD can be used to screen for compounds that bind to a variant of CGDD, but not to CGDD having the exact sequence of a sequence of SEQ ID NO: 1-33. CGDD variants used to perform such screening can have a range of about 50% to about 99% sequence identity to CGDD, with various embodiments having 60%, 70%, 75%, 80%, 85%, 90%, and 95% sequence identity.

In an embodiment, a compound identified in a screen for specific binding to CGDD can be closely related to the natural ligand of CGDD, e. g. , a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner (Coligan, J. E. et al. (1991) Current Protocols in Immunology 1 (2): Chapter 5). In another embodiment, the compound thus identified can be a natural ligand of a receptor CGDD (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, a compound identified in a screen for specific binding to CGDD can be closely related to the natural receptor to which CGDD 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 CGDD which is capable of propagating a signal, or a decoy receptor for CGDD 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; Amgen Inc., Thousand Oaks CA), 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 IgG (Taylor, P. C. et al. (2001) Curr. Opin. Immunol. 13: 611-616).

In one embodiment, two or more antibodies having similar or, alternatively, different specificities can be screened for specific binding to CGDD, fragments of CGDD, or variants of CGDD. The binding specificity of the antibodies thus screened can thereby be selected to identify particular fragments or variants of CGDD. In one embodiment, an antibody can be selected such that its binding specificity allows for preferential identification of specific fragments or variants of CGDD. In another embodiment, an antibody can be selected such that its binding specificity allows for preferential diagnosis of a specific disease or condition having increased, decreased, or otherwise abnormal production of CGDD.

In an embodiment, anticalins can be screened for specific binding to CGDD, fragments of CGDD, or variants of CGDD. Anticalins are ligand-binding proteins that have been constructed based on a lipocalin scaffold (Weiss, G. A. and H. B. Lowman (2000) Chem. Biol. 7: R177-R184 ; Skerra, A. (2001) J. Biotechnol. 74: 257-275). The protein architecture oflipocalins can include a beta-barrel having eight antiparallel beta-strands, which supports four loops at its open end. These loops form the natural ligand-binding site of the lipocalins, a site which can be re-engineered in vitro by amino acid substitutions to impart novel binding specificities. The amino acid substitutions can be made using methods known in the art or described herein, and can include conservative substitutions (e. g. , substitutions that do not alter binding specificity) or substitutions that modestly, moderately, or significantly alter binding specificity.

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

Cells expressing CGDD or cell membrane fractions which contain CGDD are then contacted with a test compound and binding, stimulation, or inhibition of activity of either CGDD 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 CGDD, either in solution or affixed to a solid support, and detecting the binding of CGDD 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 (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 (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).

CGDD, fragments of CGDD, or variants of CGDD may be used to screen for compounds that modulate the activity of CGDD. Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for CGDD activity, wherein CGDD is combined with at least one test compound, and the activity of CGDD in the presence of a test compound is compared with the activity of CGDD in the absence of the test compound. A change in the activity of CGDD in the presence of the test compound is indicative of a compound that modulates the activity of CGDD. Alternatively, a test compound is combined with an in vitro or cell-free system comprising CGDD under conditions suitable for CGDD activity, and the assay is performed. In either of these assays, a test compound which modulates the activity of CGDD 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 CGDD 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 (iieo ; 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 CGDD 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 CGDD 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 CGDD 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 CGDD, e. g. , by secreting CGDD 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 CGDD and proteins associated with cell growth, differentiation, and death. In addition, examples of tissues expressing CGDD can be found in Table 6 and can also be found in Example XI. Therefore, CGDD appears to play a role in cell proliferative disorders including cancer, developmental disorders, neurological disorders, autoimmune/inflammatory disorders, reproductive disorders, and disorders of the placenta. In the treatment of disorders associated with increased CGDD expression or activity, it is desirable to decrease the expression or activity of CGDD. In the treatment of disorders associated with decreased CGDD expression or activity, it is desirable to increase the expression or activity of CGDD.

Therefore, in one embodiment, CGDD 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 CGDD. 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, colon, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms'tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann- Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; 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 reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, autoimmune disorders, ectopic pregnancy, teratogenesis; cancer of the breast, fibrocystic breast disease, galactorrhea; a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puberty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumors; and a disorder of the placenta such as preeclampsia, choriocarcinoma, abruptio placentae, placenta previa, placental or maternal floor infarction, placenta accreta, increate, and percreta, extrachorial placentas, chorangioma, chorangiosis, chronic villitis, placental villous endema, widespread fibrosis of the terminal villi, intervillous thrombi, hemorraghic endovasculitis, erythroblastosis fetalis, and nonimmune fetal hydrops.

In another embodiment, a vector capable of expressing CGDD 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 CGDD including, but not limited to, those described above.

In a further embodiment, a composition comprising a substantially purified CGDD 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 CGDD including, but not limited to, those provided above.

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

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

prevent a disorder associated with increased expression or activity of CGDD. Examples of such disorders include, but are not limited to, those cell proliferative disorders including cancer, developmental disorders, neurological disorders, autoimmune/inflammatory disorders, reproductive disorders, and disorders of the placenta described above. In one aspect, an antibody which specifically binds CGDD 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 CGDD.

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

In other embodiments, any protein, agonist, antagonist, antibody, complementary sequence, or vector embodiments 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 CGDD may be produced using methods which are generally known in the art. In particular, purified CGDD may be used to produce antibodies or to screen libraries of pharmaceutical agents to identify those which specifically bind CGDD. Antibodies to CGDD 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. In an embodiment, neutralizing antibodies (i. e., those which inhibit dimer formation) can be used therapeutically. Single chain antibodies (e. g. , from camels or llamas) may be potent enzyme inhibitors and may have application 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 CGDD 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 Corynebacteriumparvum are especially preferable.

It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to

CGDD 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 substantially identical to a portion of the amino acid sequence of the natural protein.

Short stretches of CGDD 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 CGDD 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 (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; 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 (Morrison, S. L. et al. (1984) Proc. Natl. Acad.

Sci. USA 81: 6851-6855; Neuberger, M. S. et al. (1984) Nature 312: 604-608; 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 CGDD-specific single chain antibodies. Antibodies with related specificity, but of distinct idiotypic composition, may be generated by chain shuffling from random combinatorial immunoglobulin libraries (Burton, D. R.

(1991) Proc. Natl. Acad. Sci. USA 88: 10134-10137).

Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature (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 CGDD 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 (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 CGDD and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies

reactive to two non-interfering CGDD 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 CGDD. Affinity is expressed as an association constant, Ka which is defined as the molar concentration of CGDD-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 CGDD epitopes, represents the average affinity, or avidity, of the antibodies for CGDD. The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular CGDD epitope, represents a true measure of affinity. High-affinity antibody preparations with Ka ranging from about 109 to 10"L/mole are preferred for use in immunoassays in which the CGDD-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 CGDD, 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 CGDD-antibody complexes. Procedures for evaluating antibody specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available (Catty, supra ; Coligan et al., supra).

In another embodiment of the invention, polynucleotides encoding CGDD, 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 CGDD. 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 CGDD (Agrawal, S. , ed. (1996) Antisense Therapeutics, Humana Press, 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 (Slater, J. E. et

al. (1998) J. Allergy Clin. Immunol. 102: 469-475; Scanlon, K. J. et al. (1995) FASEB J. 9: 1288- 1296). Antisense sequences can also be introduced intracellularly through the use of viral vectors, such as retrovirus and adeno-associated virus vectors (Miller, A. D. (1990) Blood 76: 271-278; Ausubel et al., supra ; Uckert, W. and W. Walther (1994) Pharmacol. Ther. 63: 323-347). Other gene delivery mechanisms include liposome-derived systems, artificial viral envelopes, and other systems known in the art (Rossi, J. J. (1995) Br. Med. Bull. 51: 217-225; Boado, R. J. et al. (1998) J. Pharm.

Sci. 87: 1308-1315; Morris, M. C. et al. (1997) Nucleic Acids Res. 25: 2730-2736).

In another embodiment of the invention, polynucleotides encoding CGDD 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)-X1 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 VDJ 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 Plasmodiumfalciparufrz and Trypanosoma cruzi). In the case where a genetic deficiency in CGDD expression or regulation causes disease, the expression of CGDD 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 CGDD are treated by constructing mammalian expression vectors encoding CGDD and introducing these vectors by mechanical means into CGDD-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. Récipon (1998) Curr.

Opin. Biotechnol. 9: 445-450).

Expression vectors that may be effective for the expression of CGDD 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). CGDD 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 P1ND ; 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 CGDD 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 CGDD expression are treated by constructing a retrovirus vector consisting of (i) the polynucleotide encoding CGDD 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, 1. 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 an embodiment, an adenovirus-based gene therapy delivery system is used to deliver polynucleotides encoding CGDD to cells which have one or more genetic abnormalities with respect to the expression of CGDD. 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).

In another embodiment, a herpes-based, gene therapy delivery system is used to deliver polynucleotides encoding CGDD to target cells which have one or more genetic abnormalities with respect to the expression of CGDD. The use of herpes simplex virus (HSV) -based vectors may be especially valuable for introducing CGDD 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). 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 embodiment, an alphavirus (positive, single-stranded RNA virus) vector is used to deliver polynucleotides encoding CGDD 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 CGDD into the alphavirus genome in place of the capsid-coding region results in the production of a large number of CGDD-coding RNAs and the synthesis of high levels of CGDD 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 CGDD 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 (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 RNA molecules encoding CGDD.

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 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 molecules encoding CGDD. 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.

In other embodiments of the invention, the expression of one or more selected polynucleotides of the present invention can be altered, inhibited, decreased, or silenced using RNA interference (RNAi) or post-transcriptional gene silencing (PTGS) methods known in the art. RNAi is a post-transcriptional mode of gene silencing in which double-stranded RNA (dsRNA) introduced into a targeted cell specifically suppresses the expression of the homologous gene (i. e. , the gene bearing the sequence complementary to the dsRNA). This effectively knocks out or substantially reduces the expression of the targeted gene. PTGS can also be accomplished by use of DNA or DNA fragments as well. RNAi methods are described by Fire, A. et al. (1998; Nature 391: 806-811) and Gura, T. (2000; Nature 404: 804-808). PTGS can also be initiated by introduction of a complementary segment of DNA into the selected tissue using gene delivery and/or viral vector delivery methods described herein or known in the art.

RNAi can be induced in mammalian cells by the use of small interfering RNA also known as siRNA. siRNA are shorter segments of dsRNA (typically about 21 to 23 nucleotides in length) that result in vivo from cleavage of introduced dsRNA by the action of an endogenous ribonuclease. siRNA appear to be the mediators of the RNAi effect in mammals. The most effective siRNAs appear to be 21 nucleotide dsRNAs with 2 nucleotide 3'overhangs. The use of siRNA for inducing RNAi in mammalian cells is described by Elbashir, S. M. et al. (2001; Nature 411: 494-498). siRNA can be generated indirectly by introduction of dsRNA into the targeted cell.

Alternatively, siRNA can be synthesized directly and introduced into a cell by transfection methods and agents described herein or known in the art (such as liposome-mediated transfection, viral vector methods, or other polynucleotide delivery/introductory methods). Suitable siRNAs can be selected by examining a transcript of the target polynucleotide (e. g., mRNA) for nucleotide sequences downstream from the AUG start codon and recording the occurrence of each nucleotide and the 3' adjacent 19 to 23 nucleotides as potential siRNA target sites, with sequences having a 21 nucleotide length being preferred. Regions to be avoided for target siRNA sites include the 5'and 3'untranslated regions (UTRs) and regions near the start codon (within 75 bases), as these may be richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the siRNP endonuclease complex. The selected target sites for siRNA can then be compared to the appropriate genome database (e. g. , human, etc. ) using BLAST or other sequence comparison algorithms known in the art. Target sequences with significant homology to other coding sequences can be eliminated from consideration. The selected siRNAs can be produced by chemical synthesis methods known in the art or by in vitro transcription using commercially available methods and kits such as the SILENCER siRNA construction kit (Ambion, Austin TX).

In alternative embodiments, long-tenn gene silencing and/or RNAi effects can be induced in selected tissue using expression vectors that continuously express siRNA. This can be accomplished using expression vectors that are engineered to express hairpin RNAs (shRNAs) using methods known in the art (see, e. g., Brummelkamp, T. R. et al. (2002) Science 296: 550-553; and Paddison, P. J. et al. (2002) Genes Dev. 16: 948-958). In these and related embodiments, shRNAs can be delivered to target cells using expression vectors known in the art. An example of a suitable expression vector for delivery of siRNA is the PSILENCER1.0-U6 (circular) plasmid (Ambion). Once delivered to the target tissue, shRNAs are processed in vivo into siRNA-like molecules capable of carrying out gene- specific silencing.

In various embodiments, the expression levels of genes targeted by RNAi or PTGS methods can be determined by assays for mRNA and/or protein analysis. Expression levels of the mRNA of a targeted gene can be determined, for example, by northern analysis methods using the NORTHERNMAX-GLY kit (Ambion); by microarray methods; by PCR methods; by real time PCR methods; and by other RNA/polynucleotide assays known in the art or described herein. Expression levels of the protein encoded by the targeted gene can be determined, for example, by microarray methods; by polyacrylamide gel electrophoresis; and by Western analysis using standard techniques known in the art.

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

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 CGDD expression or activity, a compound which specifically inhibits expression of the polynucleotide encoding CGDD may be therapeutically useful, and in the treatment of disorders associated with decreased CGDD expression or activity, a compound which specifically promotes expression of the polynucleotide encoding CGDD may be therapeutically useful.

In various embodiments, one or more 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 CGDD 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 CGDD 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 CGDD. 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 pointe 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 (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 CGDD, antibodies to CGDD, and mimetics, agonists, antagonists, or inhibitors of CGDD.- In various embodiments, the compositions described herein, such as pharmaceutical compositions, 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 allows 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 CGDD or fragments thereof. For example, liposome preparations containing a cell-impermeable macromolecule may promote cell fusion and intracellular delivery of the macromolecule. Alternatively, CGDD 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 CGDD or fragments thereof, antibodies of CGDD, and agonists, antagonists or inhibitors of CGDD, 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 LD^o (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 EDso 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 ßg to 100, 000/-zig, 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 CGDD may be used for the diagnosis of disorders characterized by expression of CGDD, or in assays to monitor patients being treated with CGDD or agonists, antagonists, or inhibitors of CGDD. Antibodies useful for diagnostic

purposes may be prepared in the same manner as described above for therapeutics. Diagnostic assays for CGDD include methods which utilize the antibody and a label to detect CGDD 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 CGDD, including ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of CGDD expression. Normal or standard values for CGDD expression are established by combining body fluids or cell extracts taken from normal mammalian subjects, for example, human subjects, with antibodies to CGDD under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods, such as photometric means. Quantities of CGDD 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, polynucleotides encoding CGDD may be used for diagnostic purposes. The polynucleotides which may be used include oligonucleotides, 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 CGDD may be correlated with disease. The diagnostic assay may be used to determine absence, presence, and excess expression of CGDD, and to monitor regulation of CGDD levels during therapeutic intervention.

In one aspect, hybridization with PCR probes which are capable of detecting polynucleotides, including genomic sequences, encoding CGDD or closely related molecules may be used to identify nucleic acid sequences which encode CGDD. 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 CGDD, 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 CGDD 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 : 34-66 or from genomic sequences including promoters, enhancers, and introns of the CGDD gene.

Means for producing specific hybridization probes for polynucleotides encoding CGDD include the cloning of polynucleotides encoding CGDD or CGDD derivatives into vectors for the production of mRNA probes. Such vectors are known in the art, are commercially available, and may be used to synthesize RNA probes in vitro by means of the addition of the appropriate RNA polymerases and the appropriate labeled nucleotides. Hybridization probes may be labeled by a variety of reporter groups, for example, by radionuclides such as 32p or 35S, or by enzymatic labels,

such as alkaline phosphatase coupled to the probe via avidintbiotin coupling systems, and the like.

Polynucleotides encoding CGDD may be used for the diagnosis of disorders associated with expression of CGDD. 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, colon, gall bladder, ganglia, gastrointestinal tract, heart, kidney, liver, lung, muscle, ovary, pancreas, parathyroid, penis, prostate, salivary glands, skin, spleen, testis, thymus, thyroid, and uterus; a developmental disorder such as renal tubular acidosis, anemia, Cushing's syndrome, achondroplastic dwarfism, Duchenne and Becker muscular dystrophy, epilepsy, gonadal dysgenesis, WAGR syndrome (Wilms'tumor, aniridia, genitourinary abnormalities, and mental retardation), Smith-Magenis syndrome, myelodysplastic syndrome, hereditary mucoepithelial dysplasia, hereditary keratodermas, hereditary neuropathies such as Charcot-Marie-Tooth disease and neurofibromatosis, hypothyroidism, hydrocephalus, seizure disorders such as Syndenham's chorea and cerebral palsy, spina bifida, anencephaly, craniorachischisis, congenital glaucoma, cataract, and sensorineural hearing loss; a neurological disorder such as epilepsy, ischemic cerebrovascular disease, stroke, cerebral neoplasms, Alzheimer's disease, Pick's disease, Huntington's disease, dementia, Parkinson's disease and other extrapyramidal disorders, amyotrophic lateral sclerosis and other motor neuron disorders, progressive neural muscular atrophy, retinitis pigmentosa, hereditary ataxias, multiple sclerosis and other demyelinating diseases, bacterial and viral meningitis, brain abscess, subdural empyema, epidural abscess, suppurative intracranial thrombophlebitis, myelitis and radiculitis, viral central nervous system disease, prion diseases including kuru, Creutzfeldt-Jakob disease, and Gerstmann- Straussler-Scheinker syndrome, fatal familial insomnia, nutritional and metabolic diseases of the nervous system, neurofibromatosis, tuberous sclerosis, cerebelloretinal hemangioblastomatosis, encephalotrigeminal syndrome, mental retardation and other developmental disorders of the central nervous system including Down syndrome, cerebral palsy, neuroskeletal disorders, autonomic nervous system disorders, cranial nerve disorders, spinal cord diseases, muscular dystrophy and other neuromuscular disorders, peripheral nervous system disorders, dermatomyositis and polymyositis, inherited, metabolic, endocrine, and toxic myopathies, myasthenia gravis, periodic paralysis, mental disorders including mood, anxiety, and schizophrenic disorders, seasonal affective disorder (SAD), akathesia, amnesia, catatonia, diabetic neuropathy, tardive dyskinesia, dystonias, paranoid psychoses, postherpetic neuralgia, Tourette's disorder, progressive supranuclear palsy, corticobasal degeneration, and familial frontotemporal dementia; 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 reproductive disorder such as a disorder of prolactin production, infertility, including tubal disease, ovulatory defects, endometriosis, a disruption of the estrous cycle, a disruption of the menstrual cycle, polycystic ovary syndrome, ovarian hyperstimulation syndrome, an endometrial or ovarian tumor, a uterine fibroid, autoimmune disorders, ectopic pregnancy, teratogenesis; cancer of the breast, fibrocystic breast disease, galactorrhea; a disruption of spermatogenesis, abnormal sperm physiology, cancer of the testis, cancer of the prostate, benign prostatic hyperplasia, prostatitis, Peyronie's disease, impotence, carcinoma of the male breast, gynecomastia, hypergonadotropic and hypogonadotropic hypogonadism, pseudohermaphroditism, azoospermia, premature ovarian failure, acrosin deficiency, delayed puberty, retrograde ejaculation and anejaculation, haemangioblastomas, cystsphaeochromocytomas, paraganglioma, cystadenomas of the epididymis, and endolymphatic sac tumors; and a disorder of the placenta such as preeclampsia, choriocarcinoma, abruptio placentae, placenta previa, placental or maternal floor infarction, placenta accreta, increate, and percreta, extrachorial placentas, chorangioma, chorangiosis, chronic villitis, placental villous endema, widespread fibrosis of the terminal villi, intervillous thrombi, hemorraghic endovasculitis, erythroblastosis fetalis, and nonimmune fetal hydrops. Polynucleotides encoding CGDD 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 CGDD expression. Such qualitative or quantitative methods are well known in the art.

In a particular embodiment, polynucleotides encoding CGDD may be used in assays that detect the presence of associated disorders, particularly those mentioned above. Polynucleotides complementary to sequences encoding CGDD 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 polynucleotides encoding CGDD 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 CGDD, 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 CGDD, 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 CGDD 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 CGDD, or a fragment of a polynucleotide complementary to the polynucleotide encoding CGDD, 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 polynucleotides encoding CGDD 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 polynucleotides encoding CGDD 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 CGDD include radiolabeling or biotinylating nucleotides, coamplification of a control nucleic acid, and interpolating results from standard curves (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 polynucleotides 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, CGDD, fragments of CGDD, or antibodies specific for CGDD 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 (Seilhamer et al. ,"Comparative Gene Transcript Analysis, "U. S. Patent No. 5,840, 484; hereby 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). 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 an embodiment, the toxicity of a test compound can be 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 embodiment relates to the use of the polypeptides disclosed herein 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 interest. In some cases, further sequence data may be obtained for definitive protein identification.

A proteomic profile may also be generated using antibodies specific for CGDD to quantify the levels of CGDD 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 (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 W095/25116 ; Shalon, D. et al. (1995) PCT application W095/35505 ; Heller, R. A. et al. (1997) Proc. Natl. Acad. Sci. USA 94: 2150-2155; Heller, M. J. et al. (1997) U. S. Patent No. 5,605, 662). Various types of microarrays are well known and thoroughly described in Schena, M. , ed. (1999; DNA Microarrays: A Practical Approach, Oxford University Press, London).

In another embodiment of the invention, nucleic acid sequences encoding CGDD 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 (Harrington, J. J. et al.

(1997) Nat. Genet. 15: 345-355; Price, C. M. (1993) Blood Rev. 7: 127-134; Trask, B. J. (1991) Trends Genet. 7: 149-154). Once mapped, the nucleic acid sequences 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) (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 (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 CGDD 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 (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, CGDD, 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 CGDD 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 (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 CGDD, or fragments thereof, and washed.

Bound CGDD is then detected by methods well known in the art. Purified CGDD 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 CGDD specifically compete with a test compound for binding CGDD.

In this manner, antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with CGDD.

In additional embodiments, the nucleotide sequences which encode CGDD 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. 60/379,849, U. S. Ser. No. 60/384,661, U. S. Ser. No. 60/397,112, U. S. Ser.

No. 60/405, 420, and U. S. Ser. No. 60/412,516, are hereby expressly incorporated by reference.

EXAMPLES

I. Construction of cDNA Libraries Incyte cDNAs are derived from cDNA libraries described in the LIFESEQ database (Incyte, Palo Alto CA). Some tissues are homogenized and lysed in guanidinium isothiocyanate, while others are 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 are centrifuged over CsCI cushions or extracted with chloroform. RNA is precipitated from the lysates with either isopropanol or sodium acetate and ethanol, or by other routine methods.

Phenol extraction and precipitation of RNA are repeated as necessary to increase RNA purity.

In some cases, RNA is treated with DNase. For most libraries, poly (A) + RNA is isolated using oligo d (T) -coupled paramagnetic particles (Promega), OLIGOTEX latex particles (QIAGEN, Chatsworth CA), or an OLIGOTEX mRNA purification kit (QIAGEN). Alternatively, RNA is 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 is provided with RNA and constructs the corresponding cDNA libraries. Otherwise, cDNA is synthesized and cDNA libraries are constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen), using the recommended procedures or similar methods known in the art (Ausubel et al., supra, ch. 5). Reverse transcription is initiated using oligo d (T) or random primers. Synthetic oligonucleotide adapters are ligated to double stranded cDNA, and the cDNA is digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA is size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (Amersham Biosciences) or preparative agarose gel electrophoresis. cDNAs are ligated into compatible restriction enzyme sites of the polylinker of a suitable plasmid, e. g., PBLUESCRIPT plasmid (Stratagene), PSPORT1 plasmid (Invitrogen, Carlsbad CA), PCDNA2.1 plasmid (Invitrogen), PBK-CMV plasmid (Stratagene), PCR2- TOPOTA plasmid (Invitrogen), PCMV-ICIS plasmid (Stratagene), pIGEN (Incyte, Palo Alto CA), pRARE (Incyte), or pINCY (Incyte), or derivatives thereof. Recombinant plasmids are transformed into competent E. coli cells including XLl-Blue, XLl-BlueMRF, or SOLR from Stratagene or DHSa, DH10B, or ElectroMAX DH1OB from Invitrogen.

II. Isolation of cDNA Clones Plasmids obtained as described in Example I are recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids are 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 are resuspended in 0.1 ml

of distilled water and stored, with or without lyophilization, at 4°C.

Alternatively, plasmid DNA is 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 are carried out in a single reaction mixture. Samples are processed and stored in 384- well plates, and the concentration of amplified plasmid DNA is quantified fluorometrically using PICOGREEN dye (Molecular Probes, Eugene OR) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).

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

Sequencing reactions are 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 are 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 are 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 are identified using standard methods (Ausubel et al., supra, ch. 7). Some of the cDNA sequences are selected for extension using the techniques disclosed in Example VIM.

Polynucleotide sequences derived from Incyte cDNAs are 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 are 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 Horno sapiens, Rattus <BR> <BR> <BR> <BR> norvegicus, Mus musculus, Caerzorlzabditis elegans, Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Candida albicans (Incyte, 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, J. 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 are performed using programs based on BLAST, FASTA, BLIMPS, and HMMER. The Incyte cDNA sequences are

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) are used to extend Incyte cDNA assemblages to full length. Assembly is performed using programs based on Phred, Phrap, and Consed, and cDNA assemblages are screened for open reading frames using programs based on GeneMark, BLAST, and FASTA. The full length polynucleotide sequences are translated to derive the corresponding full length polypeptide sequences. Alternatively, a polypeptide may begin at any of the methionine residues of the full length translated polypeptide. Full length polypeptide sequences are 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 (MiraiBio, Alameda 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 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 are also used to identify polynucleotide sequence fragments from SEQ ID NO : 34-66. 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 proteins associated with cell growth, differentiation, and death are 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 (Burge, C. and S. Karlin (1997) J. Mol. Biol. 268: 78-94; 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 is set to 30 kb. To determine which of these Genscan predicted cDNA sequences encode proteins associated with cell growth, differentiation, and death, the encoded polypeptides are analyzed by querying against PFAM models for proteins associated with cell growth, differentiation, and death. Potential proteins associated with cell growth, differentiation, and death are also identified by homology to Incyte cDNA sequences that have been annotated as proteins associated with cell growth, differentiation, and death. These selected Genscan-predicted sequences are then compared by BLAST analysis to the genpept and gbpri public databases. Where necessary, the Genscan-predicted sequences are 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 is 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 is available, this information is used to correct or confirm the Genscan predicted sequence. Full length polynucleotide sequences are 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 are 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 are extended with exons predicted by the Genscan gene identification program described in Example IV. Partial cDNAs assembled as described in Example III are mapped to genomic DNA and parsed into clusters containing related cDNAs and Genscan exon predictions from one or more genomic sequences. Each cluster is analyzed using an algorithm based on graph theory and dynamic programming to integrate cDNA and genomic information, generating possible splice variants that are subsequently confirmed, edited, or extended to create a full length sequence. Sequence intervals in which the entire length of the interval is present on more than one sequence in the cluster are identified, and intervals thus identified are considered to be equivalent by transitivity. For example, if an interval is present on a cDNA and two genomic sequences, then all three intervals are considered to be equivalent. This process allows unrelated but consecutive genomic sequences to be brought together, bridged by cDNA sequence. Intervals thus identified are 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) are given preference over linkages which change parent type (cDNA to genomic sequence). The resultant stitched sequences are translated and compared by BLAST analysis to the

genpept and gbpri public databases. Incorrect exons predicted by Genscan are corrected by comparison to the top BLAST hit from genpept. Sequences are further extended with additional cDNA sequences, or by inspection of genomic DNA, when necessary.

"Stretched"Sequences.

Partial DNA sequences are extended to full length with an algorithm based on BLAST analysis. First, partial cDNAs assembled as described in Example m are queried against public databases such as the GenBank primate, rodent, mammalian, vertebrate, and eukaryote databases using the BLAST program. The nearest GenBank protein homolog is then compared by BLAST analysis to either Incyte cDNA sequences or GenScan exon predicted sequences described in Example IV. A chimeric protein is 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 are used as probes to search for homologous genomic sequences from the public human genome databases. Partial DNA sequences are therefore "stretched"or extended by the addition of homologous genomic sequences. The resultant stretched sequences are examined to determine whether they contain a complete gene.

VI. Chromosomal Mapping of CGDD Encoding Polynucleotides The sequences used to assemble SEQ ID NO : 34-66 are 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 : 34-66 are 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 are used to determine if any of the clustered sequences have been previously mapped.

Inclusion of a mapped sequence in a cluster results 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 (Sambrook and Russell, supra, ch. 7; Ausubel et al. , supra, ch. 4).

Analogous computer techniques applying BLAST are used to search for identical or related molecules in databases such as GenBank or L1FESEQ (Incyte). 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, polynucleotides encoding CGDD are analyzed with respect to the tissue sources from which they are derived. For example, some full length sequences are assembled, at least in part, with overlapping Incyte cDNA sequences (see Example III). 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 CGDD. cDNA sequences and cDNA library/tissue information are found in the LIFESEQ database (Incyte, Palo Alto CA).

VIII. Extension of CGDD Encoding Polynucleotides Full length polynucleotides are produced by extension of an appropriate fragment of the full length molecule using oligonucleotide primers designed from this fragment. One primer is synthesized to initiate 5'extension of the known fragment, and the other primer is synthesized to initiate 3'extension of the known fragment. The initial primers are 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 is avoided.

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

High fidelity amplification is obtained by PCR using methods well known in the art. PCR is performed in 96-well plates using the PTC-200 thermal cycler (MJ Research, Inc. ). The reaction mix contains 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+ are 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 is determined by dispensing 100 ttl PICOGREEN quantitation reagent (0.25% (v/v) PICOGREEN ; Molecular Probes, Eugene OR) dissolved in 1X TE and 0. 5, ul 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 is scanned in a Fluoroskan II (Labsystems Oy, Helsinki, Finland) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5, ul to 10, ul aliquot of the reaction mixture is analyzed by electrophoresis on a 1 % agarose gel to determine which reactions are successful in extending the sequence.

The extended nucleotides are 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 are separated on low concentration (0.6 to 0.8%) agarose gels, fragments are 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 are selected on antibiotic-containing media, and individual colonies are picked and cultured overnight at 37°C in 384-well plates in LB/2x carb liquid media.

The cells are lysed, and DNA is 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 is quantified by PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries are reamplified using the same conditions as described above. Samples are 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 polynucleotides 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 CGDD Encoding Polynucleotides Common DNA sequence variants known as single nucleotide polymorphisms (SNPs) are identified in SEQ ID NO : 34-66 using the LIFESEQ database (Incyte). Sequences from the same gene are 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 is used to distinguish SNPs from other sequence variants. Preliminary filters remove the majority of basecall errors by requiring a minimum Phred quality score of 15, and remove sequence alignment errors and errors resulting from improper trimming of vector sequences, chimeras, and splice variants. An automated procedure of advanced chromosome analysis is applied to the original chromatogram files in the vicinity of the putative SNP. Clone error filters use statistically generated algorithms to identify errors introduced during laboratory processing, such as those caused by reverse transcriptase, polymerase, or somatic mutation. Clustering error filters use 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 removes duplicates and SNPs found in immunoglobulins or T-cell receptors.

Certain SNPs are 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 comprises 92 individuals (46 male, 46 female), including 83 from Utah, four French, three Venezualan, and two Amish individuals. The African population comprises 194 individuals (97 male, 97 female), all African Americans. The Hispanic population comprises 324 individuals (162 male, 162 female), all Mexican Hispanic. The Asian population comprises 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 are first analyzed in the Caucasian population; in some cases those SNPs which show no allelic variance in this population are not further tested in the other three populations.

X. Labeling and Use of Individual Hybridization Probes Hybridization probes derived from SEQ ID NO : 34-66 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 moi of <BR> <BR> <BR> [Y-32P] adenosine triphosphate (Amersham Biosciences), and T4 polynucleotide kinase (DuPont NEN,<BR> Boston MA). The labeled oligonucleotides are substantially purified using a SEPHADEX G-25 superfine size exclusion dextran bead column (Amersham Biosciences). An aliquot containing 10' 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 II, Eco RI, Pst I, Xba I, or Pvu II (DuPont NEN).

The DNA from each digest is fractionated on a 0.7% agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham NH). Hybridization is carried out for 16 hours at 40°C. To remove nonspecific signals, blots are sequentially washed at room temperature under conditions of up to, for example, 0.1 x saline sodium citrate and 0. 5% sodium 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 et al., 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, M. , ed.

(1999) DNA Microarrays : A Practical Approach, Oxford University Press, London). 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 (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/jul oligo- (dT) primer (21mer), 1X first strand buffer, 0.03 units/yl RNase inhibitor, 500 uM dATP, 500, uM dGTP, 500 yM dTTP, 40 jtM 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 85° C to the stop the reaction and degrade the RNA. Samples are purified using two successive CHROMA SPIN 30 gel filtration spin columns (Clontech, 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 ; 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 , ag. 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-Aldrich, St. Louis MO) 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 nul of the array element DNA, at an average concentration of 100 ng/jul, is loaded into the open capillary printing element by a high-speed robotic apparatus. The apparatus then deposits about 5 nl of array element sample per slide.

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

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

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

Hybridization Hybridization reactions contain 9 al of sample mixture consisting of 0. 2 ug 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 al 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).

Array elements that exhibit at least about a two-fold change in expression, a signal-to-background ratio of at least about 2.5, and an element spot size of at least about 40%, are considered to be differentially expressed.

Expression For example, SEQ ID NO : 40 showed differential expression associated with osteosarcoma as

determined by microarray analysis. Messenger RNA from normal human osteoblasts (primary culture, NHOst 5488) was compared with mRNA from biopsy specimens and osteosarcoma tissues.

The expression of SEQ ID NO : 40 was decreased by at least two-fold in femur tumor tissue from patients with osteosarcoma compared to normal osteoblasts. Therefore, SEQ ID NO : 40 may be used in the development of therapeutics and in monitoring treatment of and diagnostic assays for osteosarcoma.

In an alternative example, SEQ ID NO : 42 and SEQ ID NO : 44-47 showed differential expression associated with breast cancer as determined by microarray analysis. The gene expression profile of a nonmalignant mammary epithelial cell line was compared to the gene expression profiles of breast carcinoma lines at different stages of tumor progression. Cell lines compared included: a) BT-20, a breast carcinoma cell line derived irez vitro from the cells emigrating out of thin slices of tumor mass isolated from a 74-year-old female, b) BT-474, a breast ductal carcinoma cell line that was isolated from a solid, invasive ductal carcinoma of the breast obtained from a 60-year-old woman, c) BT-483, a breast ductal carcinoma cell line that was isolated from a papillary invasive ductal tumor obtained from a 23-year-old normal, menstruating, parous female with a family history of breast cancer, d) Hs 578T, a breast ductal carcinoma cell line isolated from a 74-year-old female with breast carcinoma, e) MCF7, a nonmalignant breast adenocarcinoma cell line isolated from the pleural effusion of a 69-year-old female, f) MCF-10A, a breast mammary gland (luminal ductal characteristics) cell line isolated from a 36-year-old woman with fibrocystic breast disease, g) MDA- MB-468, a breast adenocarcinoma cell line isolated from the pleural effusion of a 51-year-old female with metastatic adenocarcinoma of the breast, h) MDA-MB-435S, a spindle shaped strain that evolved from the parent line (435) as isolated from the pleural effusion of a 31-year old female with metastatic, ductal adenocarcinoma of the breast, i) MDA-MB-231, a breast adenocarcinoma cell line isolated from the pleural effusion of a 51-year old female, j) T-47D, a breast carcinoma cell line isolated from a pleural effusion obtained from a 54-year old female with an infiltrating ductal carcinoma of the breast, k) Sk-BR-3, a breast adenocarcinoma cell line isolated from a malignant pleural effusion of a 43-year old female, 1) HMEC, a primary breast epithelial cell line isolated from a normal donor. SEQ ID NO : 42 showed at least a two-fold increase in expression in Hs 578T breast carcinoma cells compared to HMEC breast epithelial cells. SEQ ID NO : 44 showed at least a two- fold decrease in expression in BT-20, MDA-MB-468, BT-483, MCF7, BT-474, and Hs578T breast carcinoma cells compared to HMEC breast epithelial cells. SEQ ID NO : 45 showed at least a two- fold decrease in expression in MDA-MB-435S, MCF-10A, MDA-MB-231, BT-20, T-47D, and Sk- BR-3 cells compared to HMEC breast epithelial cells. In addition, SEQ ID NO : 45 showed at least a two-fold decrease in expression in T-47D, BT-20, MDA-MB-231, and Sk-BR-3 breast carcinoma cells compared to nonmalignant MCF-10A cells. SEQ ID NO : 45 showed at least a two-fold decrease

in expression in MCF-10A cells treated with UV (254 nm) radiation. Mammalian cells require high- fidelity DNA repair to reduce the occurrence of genetic mutations caused by DNA-damaging agents such as UV radiation. In the absence of efficient DNA repair, accumulation of DNA lesions frequently leads to genomic instability and cancer development. Deficient DNA repair capacity appears to be a predisposing factor for breast cancer. SEQ ID NO : 46 showed at least a two-fold increase in expression in Hs 578T breast carcinoma cells compared to HMEC breast epithelial cells.

In addition, SEQ ID NO : 46 showed at least a two-fold decrease in expression in MCF7 cells treated with epidermal growth factor compared to untreated cells. SEQ ID NO : 47 showed at least a two-fold increase in expression in BT-20 breast carcinoma cells compared to nonmalignant MCF-lOA cells.

Therefore, SEQ ID NO : 42 and SEQ ID NO : 44-47 may be used in the development of therapeutics and in monitoring treatment of and diagnostic assays for breast cancer.

In an alternative example, SEQ ID NO : 42 and SEQ ID NO : 46 showed differential expression associated with prostate cancer as determined by microarray analysis. The expression of SEQ ID NO : 42 showed at least a two-fold decrease in LNCaP, PC-3, and DU 145 prostate carcinoma cells compared to PrEC prostate epithelial cells. The expression of SEQ ID NO : 46 showed at least a two- fold decrease in PC-3 prostate carcinoma cells compared to PrEC prostate epithelial cells. The PC-3 cell line was isolated from a 62-year old male with grade IV prostate adenocarcinoma. The LNCaP cell line was isolated from a lymph node biopsy of a 50-year old male with metastatic prostate carcinoma. The DU 145 cell line was isolated from a metastatic site in the brain of 69-year old male with widespread metastatic prostate carcinoma. The PrEC cell line is a prostate epithelial cell line isolated from a normal donor. Therefore, SEQ ID NO : 42 and SEQ ID NO : 46 may be used in the development of therapeutics and in monitoring treatment of and diagnostic assays for prostate cancer.

In an alternative example, SEQ ID NO : 44 showed differential expression associated with Tangier disease. Normal and Tangier disease derived fibroblasts were compared. Human fibroblasts were obtained from skin explants from both normal subjects and two patients with homozygous Tangier disease. Cell lines were immortalized by transfection with human papillomavirus 16 genes E6 and E7 and a neomycin resistance selectable marker. In addition, both types of cells were cultured in the presence of cholesterol and compared with the same cell type cultured in the absence of cholesterol. TD derived cells are shown to be deficient in an assay of apoA-I mediated tritiated cholesterol efflux. Therefore, SEQ ID NO : 44 may be used in the development of therapeutics and in monitoring treatment of and diagnostic assays for Tangier disease.

In an alternative example, SEQ ID NO : 47 showed differential expression associated with ovarian cancer. The expression of SEQ ID NO : 47 showed at least a two-fold increase in tumorous ovarian tissue from a patient with adenocarcinoma compared to non-tumorous matched tissue from the same donor as determined by microarray analysis. Therefore, SEQ ID NO : 47 may be used in the

development of therapeutics and in monitoring treatment of and diagnostic assays for ovarian cancer.

In an alternative example, SEQ ID NO : 47 showed tissue-specific expression. RNA samples isolated from a variety of normal human tissues were compared to a common reference sample.

Tissues contributing to the reference sample were selected for their ability to provide a complete distribution of RNA in the human body and include brain (4%), heart (7%), kidney (3%), lung (8%), placenta (46%), small intestine (9%), spleen (3%), stomach (6%), testis (9%), and uterus (5%). The normal tissues assayed were obtained from at least three different donors. RNA from each donor was separately isolated and individually hybridized to the microarray. Since these hybridization experiments were conducted using a common reference sample, differential expression values are directly comparable from one tissue to another. The expression of SEQ ID NO : 47 was increased by at least two-fold in heart (atrium and ventricle), kidney, and fallopian tube tissues as compared to the reference sample. Therefore, SEQ ID NO : 47 can be used as a tissue marker for heart (atrium and ventricle), kidney, and fallopian tube tissues.

In an alternative example, SEQ ID NO : 56-58 showed tissue-specific expression as determined by microarray analysis. RNA samples isolated from a variety of normal human tissues were compared to a common reference sample. Tissues contributing to the reference sample were selected for their ability to provide a complete distribution of RNA in the human body and include brain (4%), heart (7%), kidney (3%), lung (8%), placenta (46%), small intestine (9%), spleen (3%), stomach (6%), testis (9%), and uterus (5%). The normal tissues assayed were obtained from at least three different donors. RNA from each donor was separately isolated and individually hybridized to the microarray. Since these hybridization experiments were conducted using a common reference sample, differential expression values are directly comparable from one tissue to another. The expression of SEQ ID NO : 56-58 was increased by at least two-fold in spinal cord and liver as compared to the reference sample. Therefore, SEQ ID NO : 56-58 can each be used as a tissue marker for spinal cord and liver.

In an alternative example, SEQ ID NO : 60 showed differential expression associated with colon cancer as determined by microarray analysis. Matched normal and tumor tissue samples from a 64-year-old female diagnosed with moderately differentiated colon adenocarcinoma (Huntsman Cancer Institute, Salt Lake City, UT) were compared by competitive hybridization. The expression of SEQ ID NO : 60 showed at least a two-fold increase in expression in colon tumor tissue compared to microscopically normal tissue from the same donor. Therefore, in various embodiments, SEQ ID NO : 60 can be used for one or more of the following: i) monitoring treatment of colon cancer, ii) diagnostic assays for colon cancer, and iii) developing therapeutics and/or other treatments for colon cancer.

In an alternative example, the C3A cell line was used to investigate transcript profiling in

response to gemfibrozil dissolved in carboxymethyl cellulose. SEQ ID NO : 61 showed at least a two- fold decrease in expression in C3A cells treated with gemfibrozil compared to untreated cells as determined by microarray analysis. C3A cells were treated with 120,600, 800 or 1200 RM gemfibrozil for 1,3 or 6 hours. Therefore, in various embodiments, SEQ ID NO : 61 can be used for one or more of the following: i) monitoring treatment of coronary heart disease, hyperlipoproteinemia, obesity, gall bladder disease, stroke, and hyperlipidemia, ii) diagnostic assays for coronary heart disease, hyperlipoproteinemia, obesity, gall bladder disease, stroke, and hyperlipidemia, and iii) developing therapeutics and/or other treatments for coronary heart disease, hyperlipoproteinemia, obesity, gall bladder disease, stroke, and hyperlipidemia.

XII. Complementary Polynucleotides Sequences complementary to the CGDD-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring CGDD. 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 CGDD. 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 CGDD-encoding transcript.

XIII. Expression of CGDD Expression and purification of CGDD is achieved using bacterial or virus-based expression systems. For expression of CGDD 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 CGDD upon induction with isopropyl beta-D- thiogalactopyranoside (IPTG). Expression of CGDD 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 CGDD 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 Spodopterafrugiperda (Sf9) insect cells in most cases, or human hepatocytes, in some cases.

Infection of the latter requires additional genetic modifications to baculovirus (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, CGDD 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 Schistosorna japoni. cu » a, 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 CGDD 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 et al. (supra, ch. 10 and 16). Purified CGDD obtained by these methods can be used directly in the assays shown in Examples XVII and XVIII, where applicable.

XIV. Functional Assays CGDD function is assessed by expressing the sequences encoding CGDD 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, ug 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. i-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 Cytometry, Oxford, New York NY).

The influence of CGDD on gene expression can be assessed using highly purified populations of cells transfected with sequences encoding CGDD 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 CGDD and other genes of interest can be analyzed by northern analysis or microarray techniques.

XV. Production of CGDD Specific Antibodies CGDD 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 CGDD 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 (Ausubel et al., 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 (Ausubel et al., supra). Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. Resulting antisera are tested for antipeptide and anti-CGDD activity by, for example, binding the peptide or CGDD 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 CGDD Using Specific Antibodies Naturally occurring or recombinant CGDD is substantially purified by immunoaffinity chromatography using antibodies specific for CGDD. An immunoaffinity column is constructed by covalently coupling anti-CGDD 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 CGDD are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of CGDD (e. g. , high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/CGDD 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 CGDD is collected.

XVII. Identification of Molecules Which Interact with CGDD CGDD, or biologically active fragments thereof, are labeled with"'I Bolton-Hunter reagent (Bolton, A. E. and W. M. Hunter (1973) Biochem. J. 133: 529-539). Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled CGDD, washed, and any wells with labeled CGDD complex are assayed. Data obtained using different concentrations of CGDD are used to calculate values for the number, affinity, and association of CGDD with the candidate molecules.

Alternatively, molecules interacting with CGDD 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).

CGDD 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).

XVIII. Demonstration of CGDD Activity CGDD activity is demonstrated by measuring the induction of terminal differentiation or cell cycle progression when CGDD is expressed at physiologically elevated levels in mammalian cell culture systems. cDNA is subcloned into a mammalian expression vector containing a strong promoter that drives high levels of cDNA expression. Vectors of choice include PCMV SPORT (Life Technologies, Gaithersburg, MD) and PCR 3.1 (Invitrogen, Carlsbad, CA), both of which contain the cytomegalovirus promoter. 5-10 ofrecombinant vector are transiently transfected into a human cell line, preferably of endothelial or hematopoietic origin, 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, Palo Alto, CA), CD64, or a CD64-GFP fusion protein. Flow cytometry detects and quantifies the uptake of fluorescent molecules that diagnose events preceding or coincident with cell cycle progression or terminal differentiation. 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; up or down-regulation of DNA synthesis as measured by decrease in bromodeoxyuridine uptake; alterations in expression of cell surface and intracellular proteins as measured by reactivity with specific antibodies; and alterations in plasma membrane composition as measured by the binding of fluorescein-conjugated Annexin V protein to the cell

surface. Methods in flow cytometry are discussed in Ormerod, M. G. (1994) Flow Cytometry, Oxford, New York, NY.

Alternatively, an in vitro assay for CGDD activity measures the transformation of normal human fibroblast cells overexpressing antisense CGDD RNA (Garkavtsev, I. and K. Riabowol (1997) Mol. Cell Biol. 17: 2014-2019). cDNA encoding CGDD is subcloned into the pLNCX retroviral vector to enable expression of antisense CGDD RNA. The resulting construct is transfected into the ecotropic BOSC23 virus-packaging cell line. Virus contained in the BOSC23 culture supernatant is used to infect the amphotropic CAK8 virus-packaging cell line. Virus contained in the CAK8 culture supernatant is used to infect normal human fibroblast (Hs68) cells. Infected cells are assessed for the following quantifiable properties characteristic of transformed cells: growth in culture to high density associated with loss of contact inhibition, growth in suspension or in soft agar, formation of colonies or foci, lowered serum requirements, and ability to induce tumors when injected into immunodeficient mice. The activity of CGDD is proportional to the extent of transformation of Hs68 cells.

Alternatively, CGDD can be expressed in a mammalian cell line by transforming the cells with a eukaryotic expression vector encoding CGDD. Eukaryotic expression vectors are commercially available, and the techniques to introduce them into cells are well known to those skilled in the art. To assay the cellular localization of CGDD, cells are fractionated as described by Jiang, H. P. et al. (1992; Proc. Natl. Acad. Sci. 89: 7856-7860). Briefly, cells pelleted by low-speed centrifugation are resuspended in buffer (10 mM TRIS-HC1, pH 7. 4/ 10 mM NaCl/3 mM MgC/5 mM EDTA with 10 ug/ml aprotinin, 10 ug/ml leupeptin, 10 ug/ml pepstatin A, 0.2 mM phenylmethylsulfonyl fluoride) and homogenized. The homogenate is centrifuged at 600 x g for 5 minutes. The particulate and cytosol fractions are separated by ultracentrifugation of the supernatant at 100,000 x g for 60 minutes. The nuclear fraction is obtained by resuspending the 600 x g pellet in sucrose solution (0.25 M sucrose/10 mM TRIS-HC1, pH 7. 4/ 2 mM MgCl2) and recentrifuged at 600 x g. Equal amounts of protein from each fraction are applied to an SDS/10% polyacrylamide gel and blotted onto membranes. Western blot analysis is performed using CGDD anti-serum. The localization of CGDD is assessed by the intensity of the corresponding band in the nuclear fraction relative to the intensity in the other fractions. Alternatively, the presence of CGDD in cellular fractions is examined by fluorescence microscopy using a fluorescent antibody specific for CGDD.

Alternatively, CGDD activity may be demonstrated as the ability to interact with its associated Ras superfamily protein, in an in vitro binding assay. The candidate Ras superfamily proteins are expressed as fusion proteins with glutathione S-transferase (GST), and purified by affinity chromatography on glutathione-Sepharose. The Ras superfamily proteins are loaded with GDP by incubating 20 mM Tris buffer, pH 8.0, containing 100 mM NaCl, 2 mM EDTA, 5 mM

MgC12, 0.2 mM DTT, 100, uM AMP-PNP and 10 M GDP at 30°C for 20 minutes. CGDD is expressed as a FLAG fusion protein in a baculovirus system. Extracts of these baculovirus cells containing CGDD-FLAG fusion proteins are precleared with GST beads, then incubated with GST- Ras superfamily fusion proteins. The complexes formed are precipitated by glutathione-Sepharose and separated by SDS-polyacrylamide gel electrophoresis. The separated proteins are blotted onto nitrocellulose membranes and probed with commercially available anti-FLAG antibodies. CGDD activity is proportional to the amount of CGDD-FLAG fusion protein detected in the complex.

Alternatively, as demonstrated by Li and Cohen (Li, L. and S. N. Cohen (1995) Cell 85: 319- 329), the ability of CGDD to suppress tumorigenesis can be measured by designing an antisense sequence to the 5'end of the gene and transfecting NIH 3T3 cells with a vector transcribing this sequence. The suppression of the endogenous gene will allow transformed fibroblasts to produce clumps of cells capable of forming metastatic tumors when introduced into nude mice.

Alternatively, an assay for CGDD activity measures the effect of injected CGDD on the degradation of maternal transcripts. Procedures for oocyte collection from Swiss albino mice, injection, and culture are as described in Stutz et al., (supra). A decrease in the degradation of maternal RNAs as compared to control oocytes is indicative of CGDD activity. In the alternative, CGDD activity is measured as the ability of purified CGDD to bind to RNAse as measured by the assays described in Example XVII.

Alternatively, an assay for CGDD activity measures syncytium formation in COS cells transfected with an CGDD expression plasmid, using the two-component fusion assay described in Mi (supra). This assay takes advantage of the fact that human interleukin 12 (IL-12) is a heterodimer comprising subunits with molecular weights of 35 kD (p35) and 40 kD (p40). COS cells transfected with expression plasmids carrying the gene for p35 are mixed with COS cells cotransfected with expression plasmids carrying the genes for p40 and CGDD. The level of IL-12 activity in the resulting conditioned medium corresponds to the activity of CGDD in this assay. Syncytium formation may also be measured by light microscopy (Mi et al., supra).

An alternative assay for CGDD activity measures cell proliferation as the amount of newly initiated DNA synthesis in Swiss mouse 3T3 cells. A plasmid containing polynucleotides encoding CGDD is transfected into quiescent 3T3 cultured cells using methods well known in the art. The transiently transfected cells are then incubated in the presence of [Hjthymidine or a radioactive DNA precursor such as [a3zP] ATP. Where applicable, varying amounts of CGDD ligand are added to the transfected cells. Incorporation of [3Wthymidine into acid-precipitable DNA is measured over an appropriate time interval, and the amount incorporated is directly proportional to the amount of newly synthesized DNA and CGDD activity.

Alternatively, CGDD activity is measured by the cyclin-ubiquitin ligation assay (Townsley,

F. M. et al. (1997) Proc. Natl. Acad. Sci. USA 94: 2362-2367). The reaction contains in a volume of 10 ltl, 40 mM Tris. HCl (pH 7. 6), 5 mM Mg Cl, 0. 5 mM ATP, 10 mM phosphocreatine, 50, Ag of creatine phosphokinase/ml, 1 mg reduced carboxymethylated bovine serum albumin/ml, 50, uM ubiquitin, 1, uM ubiquitin aldehyde, 1-2 pmol 125I-labeled cyclin B, 1 pmol E1, l, uM okadaic acid, 10 , ug of protein of M-phase fraction 1A (containing active E3-C and essentially free of E2-C), and varying amounts of CGDD. The reaction is incubated at 18 °C for 60 minutes. Samples are then separated by electrophoresis on an SDS polyacrylamide gel. The amount of zsI-cyclin-ubiquitin formed is quantified by PHOSPHORIMAGER analysis. The amount of cyclin-ubiquitin formation is proportional to the activity of CGDD in the reaction.

Alternatively, an assay for CGDD activity uses radiolabeled nucleotides, such as [a32P] ATP, to measure either the incorporation of radiolabel into DNA during DNA synthesis, or fragmentation of DNA that accompanies apoptosis. Mammalian cells are transfected with plasmid containing cDNA encoding CGDD by methods well known in the art. Cells are then incubated with radiolabeled nucleotide for various lengths of time. Chromosomal DNA is collected, and radioactivity is detected using a scintillation counter. Incorporation of radiolabel into chromosomal DNA is proportional to the degree of stimulation of the cell cycle. To determine if CGDD promotes apoptosis, chromosomal DNA is collected as above, and analyzed using polyacrylamide gel electrophoresis, by methods well known in the art. Fragmentation of DNA is quantified by comparison to untransfected control cells, and is proportional to the apoptotic activity of CGDD.

Alternatively, cyclophilin activity of CGDD is measured using a chymotrypsin-coupled assay to measure the rate of cis to trans interconversion (Fischer, G. et al. (1984) Biomed. Biochim. Acta 43: 1101-1111). The chymotrypsin is used to estimate the trans-substrate cleavage activity at Xaa-Pro peptide bonds, wherein the rate constant for the cis to trans isomerization can be obtained by measuring the rate constant of the substrate hydrolysis at the slow phase. Samples are incubated in the presence or absence of the immunosuppressant drugs CsA or FK506, reactions initiated by addition of chymotrypsin, and the fluorescent reaction measured. The enzymatic rate constant is calculated from the equation pp = kH20 + kenz, wherein first order kinetics are displayed, and where one unit of PPIase activity is defined as kenz (s-l).

Alternatively, cyclophilin activity of CGDD is monitored by a quantitative immunoassay that measures its affinity for stereospecific binding to the immunosuppressant drug cyclosporin (Quesniaux, V. F. et al. (1987) Eur. J. Immunol. 17: 1359-1365). In this assay, the cyclophilin- cyclosporin complex is coated on a solid phase, with binding detected using anti-cyclophilin rabbit antiserum enhanced by an antiglobulin-enzyme conjugate.

Alternatively, activity of CGDD is monitored by a binding assay developed to measure the non-covalent binding between FKBPs and immunosuppressant drugs in the gas phase using

electrospray ionization mass spectrometry (Trepanier, D. J. et al. (1999) Ther. Drug Monit. 21: 274- 280). In electrospray ionization, ions are generated by creating a fine spray of highly charged droplets in the presence of a strong electric field; as the droplet decreases in size, the charge density on the surface increases. Ions are electrostatically directed into a mass analyzer, where ions of opposite charge are generated in spatially separate sources and then swept into capillary inlets where the flows are merged and where reactions occur. By comparing the charge states of bound versus unbound CGDD/immunosuppressive drug complexes, relative binding affinities can be established and correlated with in vitro binding and immunosuppressive activity.

Various modifications and variations of the described compositions, 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. It will be appreciated that the invention provides novel and useful proteins, and their encoding polynucleotides, which can be used in the drug discovery process, as well as methods for using these compositions for the detection, diagnosis, and treatment of diseases and conditions.

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.

Nor should the description of such embodiments be considered exhaustive or limit the invention to the precise forms disclosed. Furthermore, elements from one embodiment can be readily recombined with elements from one or more other embodiments. Such combinations can form a number of embodiments within the scope of the invention. It is intended that the scope of the invention be defined by the following claims and their equivalents.

Table 1 Incyte Project ID Polypeptide Incyte Polynucleotide Incyte SEQ ID NO: Polypeptide ID SEQ ID NO: Polynucleotide ID Incyte Full Length Clones 7511094 1 7511094CD1 34 7511094CB1 2220889CA2 7511619 2 7511619CD1 35 7511619CB1 7511720 3 7511720CD1 36 7511720CB1 7511661 4 7511661CD1 37 7511661CB1 7512204 5 7512204CD1 38 7512204CB1 90019612CA2 7512197 6 7512197CD1 39 7512197CB1 7511656 7 7511656CD1 40 7511656CB1 7512297 8 7512297CD1 41 7512297CB1 7512392 9 7512392CD1 42 7512392CB1 7511589 10 7511589CD1 43 7511589CB1 8131015CA2 7511699 11 7511699CD1 44 7511699CB1 7512595 12 7512595CD1 45 7512595CB1 2803911 13 2803911CD1 46 2803911CB1 7511806 14 7511806CD1 47 7511806CB1 7511853 15 7511853CD1 48 7511853CB1 95120972CA2, 95120988CA2, 95121024CA2, 95121032CA2, 95121048CA2, 95121064CA2, 95121080CA2, 95121108CA2, 7512306 16 7512306CD1 49 7512306CB1 95121172CA2 7512607 17 7512607CD1 50 7512607CB1 95170124CA2, 95188291CA2 7511851 18 7511851CD1 51 7511851CB1 7512569 19 7512569CD1 52 7512569CB1 7512570 20 7512570CD1 53 7512570CB1 7512846 21 7512846CD1 54 7512846CB1 7517814 22 7517814CD1 55 7517814CB1 90187858CA2 7519828 23 7519828CD1 56 7519828CB1 95029192CA2 7519833 24 7519833CD1 57 7519833CB1 95029168CA2 7519834 25 7519834CD1 58 7519834CB1 95029276CA2 7519939 26 7519939CD1 59 7519939CB1 95103184CA2 7520133 27 7520133CD1 60 7520133CB1 95103753CA2 Table 1 Incyte Project ID Polypeptide Incyte Polynucleotide Incyte SEQ ID NO: Polypeptide ID SEQ ID NO: Polynucleotide ID Incyte Full Length Clones 7520171 28 7520171CD1 61 7520171CB1 95103419CA2 7522453 29 7522453CD1 62 7522453CB1 7522454 30 7522454CD1 63 7522454CB1 7517956 31 7517956CD1 64 7517956CB1 90071047CA2 7517957 32 7517957CD1 65 7517957CB1 90019644CA2, 95122447CA2 7519293 33 7519293CD1 66 7519293CB1 95096945CA2 Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 1 7511094CD1 g6090856 3.1E-68 [Homo sapiens] FLASH Kimura, T. et al. Reply: Searching for FLASH domains. Nature 401, 662-663 (1999). 432684#CASP8A 2.5E-69 [Homo sapiens][Activator; REceptor (signalling)] Caspase 8 associated protein 2 (FLASH), P2 an apoptosis activator that interacts with and activates caspase-8 (CASP8) in Fas (TNFRSF6) -mediated apoptosis, coordinates NF-KB (NFKB1) activation in TNF signaling. Choi, Y. H. et al. FLASH coordinates NF-kappa B activity via TRAF2. J Biol Chem 276, 25073-7. (2001). 746279#Casp8ap2 2.5E-60 [Mus musculus] Caspase 8 associated protein 2 (FLASH), an apoptosis activator that interacts with and activates caspase-8 (Casp8) in Fas (Tnfrsf6)-mediated apoptosis. Imai, Y. et al. The CED-4-homologous protein FLASH is involved in FAs-mediated activation of caspase-8 during apoptosis. Nature 398, 777-85 (1999). 2 7511619CD1 g18496883 2.7E-93 [Homo sapiens] cyclin H 334500#CCNH 2.2E-94 [Homo sapiens][Regulatory subunit; Cyclin][Nuclear] Cyclin H, component of heterotrimeric CAK (CDK-activating kinase), which is a component of the multimeric TFIIH, a complex involved in transcriptiion and DNA repair. Makela, T. P. et al. A cyclin associated with the CDK-activating kinase MO15. Nature 371, 254-7 (1994). 3 7511720CD1 g4063709 1.2E-44 [Homo sapiens] cornichon 342412#CNIL 9.6E-46 [Homo sapiens] Cornichon-like, protein with similarity to Droscophila cornichon protein, may play a signaling role during T-cell activation. Utku, N. et al. The human homolog of Drosophila cornichon protein is differentially expressed in alloactivated T-cells. Biochim Biophys Acta 1449, 203-10 (1999). 584537#Cnil 9.3E-32 [Mus musculus] Cornichon-like, protein with similarity to Drosophila cornichon protein which plays a role in embryonic development via the epidermal growth factor receptor (Egfr) signaling pathway.

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: Hwang, S. Y. et al. The mouse cornichon gene family. Dev Genes Evol 209, 120-5 (1999). 4 7511661CD1 g2864606 6.8E-200 [Mus musculus] CDC10 Soulier, S. et al. Biochim. Biophys. Acta 1442, 339-346 (1998) 619384#CDC10 2.3E-214 [Homo sapiens][Structural protein; Hydrolase; GTP-binding protein/GTPase][Cytoplasmic; Cytoskeletal] Cell division cycle 10 (S. cerevisiae homolog), a member of the septin family of proteins that contains a GTP-binding motif, localized in the cell cortex and cleavage furrow and associated with actin, may play a role in cytokinesis and vesicle trafficking Huang, H. et al. Cancer Res 60, 6868-74 (2000). Nakatsuru, S. et al. Biochem Biophys Res Commun 202, 82-7 (1994). 320582#Sept7 5.5E-201 [Mus musculus] Septin 7 (cell divisiion cycle 10 S. cerevisiae homolog), a putative GTP- binding protein, binds to Borg 3 which is an effector in the Cdc42 signaling pathway, may be involved in cytoskeletal organization Soulier, S. et al. Biochim Biophys Acta 1442, 339-46 (1998). Joberty, G. et al. Nat Cell Biol 3, 861-6 (2001). 657537#Cdc10 1.1E-200 [Rattus norvegicus] Cell division cycle 10 (S. cerevisiae homolog), a member of the septin family of proteins, may play a role in cytokinesis and vesicle trafficking and the formation and maintenance of neuronal spines and assembly of proteins at the postsynaptic site Hsu, S. C. et al. Neuron 20, 1111-22 (1998). 5 7512204CD1 g219539 2.9E-76 [Homo sapiens] CGM7 Kuroki, M. et al. J. Biol. Chem. 266, 11810-11817 (1991) 334640#CEACA 2.4E-77 [Homo sapiens][Plasma membrane; Unspecified membrane] Member of the CEA subgroup M4 of the immunoglobulin superfamily Muenzner, P. et al. J Biol Chem 276, 24331-40 (2001). Thompson, J. et al. Int J Cancer 55, 311-9 (1993).

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 6 7512197CD1 g595924 2.0E-33 [Homo sapiens] Bak protein Kiefer, M. C. et al. Bak, Nature 374, 736-739 (1995) 334316#BAK1 1.6E-34 [Homo sapiens][Cytoplasmic; Mitochondrial] BCL2 antagonist - killer, induces apoptosis, induces cytochrome a release from mitohondria and heterodimerizes with BCLx (BCL2L1) and BCL2, interacts with human papilloma virus protein E7 and shows increased expression in Alzheimer's disease 757362#Bak1 3.6E-23 [Rattus norvegicus] Protein with strong similarity to Bcl2 antagonist - killer (mouse BAk1), which induces apoptosis and mitochondrial cytochrome c release and binds mouse Bcl21 and mouse Bcl2, contains two Bcl-2 family domains, which are found in apoptosis regulators 580933#Bak1 1.8E-21 [Mus musculus] Bcl2 antagonist - killer, induces apoptosis, induces cytochrome c release from mitochondria and heterodimerizes with Bcl-x (Bcl21) and Bcl2 Lindsten, t. et al. Mol Cell 6, 1389-1399 (2000). Korsmeyer, S. J. et al. Cell DEath Differ 7, 1166-73 (2000). 7 7511656CD1 g2895085 1.9E-53 [Homo sapiens] hD54+ins2 isoform Byrne, J. A. et al. Oncogene 16, 873-881 (1998) Nourse, C. R. et al. Biochim. Biophys. Acta 1443,1 55-168 (1998) Byrne, J. A. et al. Genomics 35, 523-532 (1996) 338646#TPD52L 1.5E-54 [Homo sapiens] Tumor protein D52-like 2, a member of the D52-like family of proteins 2 with coiled-coil domains, participates in homo- and heterologous interactiions that may be important in vesicle transport and control of cell proliferation Byrne, J. A. et al. Oncogene 16, 873-81 (1998). 581715#Tpd52 4.4E-41 [Mus musculus] Protein with a coiled-coil domain and PEST domains, has strong similarity to human TPD52, which is expressed in cancer cells Byrne, J. A. et al (supra) 8 7512297CD1 g19070547 0.0 [Homo sapiens] (AF349467) amplified in breast cancer 2 Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 9 7512392CD1 g15987491 2.8E-81 [Homo sapiens] tumor endothelial marker 5 precursor Carson-Walte,r E. B. et al. Cancer REs. 61, 6649-6655 (2001) 762413#TEM5 2.3E-82 [Homo sapiens] Member of the secretin family of G protein-coupled receptors (GPCR), contains eight leucine rich repeats and a leucine rich repeat C-terminal cysteine rich domain, has a region of low similarity to a regiion of slit homolog 1 (mouse Slit1) 10 7511589CD1 g4008085 8.6E-16 [Homo sapiens] cyclin E2 Gudas, J. M. et al. Mol. Cell. Biol. 19, 612-622 (1999) 760108#CCNE2 6.9E-17 [Homo sapiens][Regulatory subunit; Cyclin][Nuclear] Cyclin E2, a G1-specific CDK kinase regulatory subunit that interacts with CDK2 and CDK3 and is overexpressed in transformed cells Gudas, J. M. et al. Mol Cell Biiol 19, 612-22 (1999). Muller-Tidow, C. et al. Cancer Res 61, 647-53 (2001). 584431#Cene2 6.5E-14 [Mus musculus][Regulatory subunit; Cyclin] Cyclin E2, a G1-specific CDK kinase regulatory subunit that interacts with CDK2; human CCNE2 is overexpressed in transformed cells Lauper, N. et al. Oncogene 17, 2637-43 (1998). 11 7511699CD1 g5305409 2.6E-27 [Homo sapiens] frizzled 6 335406#FZD6 2.1E-28 [Homo sapiens][Receptor (signalling)][Plasma membrane] Frizzled 6, a protein with seven transmembrane domains that has similarity to the frizzled family of receptors, which bind Wnt proteins and are implicated in tissue polarity, development, and carcinogenesis Kirikoshi, H. et al. Biochem Biophys Res Commun 271, 8-14 (2000). Wang, Y. et al. J Biol Chem 271, 4468-76 (1996).

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 582945#Fzd6 2.5E-23 [Mus musculus][Receptor (signalling)][Plasma membrane] Frizzled 6, a protein with seven transmembrane domains that has similarity to the frizzled family of receptors, which bind Wnt proteins and are implicated in tissue polarity, development, and carcinogenesis He, X. et al. Science 275, 1652-4 (1997). Sheldahl, L. C. et al. Curr Biol 9, 695-8 (1999). 12 7512595CD1 g5640021 6.8E-81 [Rattus norvegicus] schlafen-4 598352#FLJ1026 9E-121 [Homo sapiens] Protein with moderate similarity to members of the Schlafen family of 0 growth regulatory proteins, which are expressed in lymphoid tissues, and may have a regulatory role in thymocyte development 757212#Slfn4 5.5E-82 [Rattus norvegicus] Protein with high similarity to schlafen 3 (mouse Slfn3), which is involved in negative control of cell proliferation and possibly regulates thymocyte development 585871#Slfn3 3.9E-78 [Mus musculus] Schlafen 3, member of the Schlafen family of growth regulatory proteins, expressed in lymphoid tissues, may have a regulatory role in thymocyte development Schwarz, D. A. et al. Immunity 9, 657-68 (1998). 13 2803911CD1 g21979456 0.0 raptor [Homo sapiens] 14 7511806CD1 g4323587 3.3E-13 [Homo sapiens] apoptosis-inducing factor AIF Susin, S. A. et al. Nature 397, 441-446 (1999) 339966#PDCD8 2.7E-14 [Homo sapiens][Oxidoreductase; Small molecule-binding protein][Nuclear; Cytoplasmic; Mitochondrial] Programmed cell death 8 (apoptosis-inducing factor), a caspase-independent apoptotic protease activator and flavoprotein, translocates from the mitochondria to the nucleus to play a role in chromatin condensatiion and DNA fragmentatiion Joza, N. et al. Nature 410, 549-54 (2001). Pardo, J. et al. J Immunol 167, 1222-9 (2001).

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 704471#Pdcd8 9.4E-11 [Rattus norvegicus] Programmed cell death 8 (apoptosis-inducing factor), an apoptosis activator that translocates from the mitochondria to the nucleus to play a role inDNA fragmentation during induced photoreceptor apoptosis Hisatomi, T. et al. Am J Pathol 158, 1271-8 (2001). 15 7511853CD1 g17064172 0.0 [Homo sapiens] NALP4 709951#Rnh2 9.5E-149 [Mus musculus] Ribonuclease-angiogenin inhibitor 2, a putative spermatogonia-specific ribonuclease inhibitor, contains six leucine-rich repeats Wang, P. J. et al., An abundance of X-linked enes expressed in spermatogonia, Nat Genet 27, 422-6. (2001). 59454#FLJ2051 7E-114 [Homo sapiens] protein containing a pyrin domain, an NAHT domain, and leucine rich 0 repeats, may play a role in apoptotic or inflammatory signaling pathways 16 7512306CD1 g683460 2.0E-17 [Homo sapiens] melanoma growth regulatory protein Bosserhoff, A. K. et al., Structure and promoter analysis of the gene encoding the human melanoma-inhibiting protein MIA, J. Biol. Chem. 271, 490-495 (1996) 567906#MIA 1.6E-18 [Homo saiens][Extracellular (excluding cell wall)] Melanoma inhibitory activity (cartilage derived retinoic acid sensitive protein) a marker for malignant melanoma, inhibits the growth of melanoma cells, may also be involved in cartilage development Bosserhoff, A. K. et al., Mouse CD-RAP/MIA gene: structure, chromosomal localization, and expression in cartilage and chondrosarcoma, Dev Dyn 208, 516-25 (1997). 17 7512607CD1 g15430743 1.8E-137 [Homo sapiens] SEPTIN6 type I Ono, R. et al., Cancer Res. 62:333-337 (2002) 743346#SEP2 1.4E-138 [Homo sapiens][Hydrolase; GTP-binding protein/GTPase] Protein with high similarity to murine Sep6, a member of the GTP-binding cell division family that resemble septins, member of the cell division protein FtsA family, which colcatlize to the septal ring with FtsZ and act in bacterial cell division Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 609262#Sept6 8.3E-134 [Mus musculus][Hydrolase; GTP-binding protein/GTPase] Member of the GTP-binding cell division family, has high similarity to septins, which are filamentous proteins with GTP- binding domains and that may interact during cytokinesis 18 7511851CD1 g17064172 0.0 [Homo sapiens] NALP4 709951#Rnh2 3.9E-175 [Mus musculus] RIbonuclease-angiogenin inhibior 2, a putative spermatogonia-specific ribonucleas inhibitor, contains six leucine-rich repeats Wang, P. J. et al., (supra) 599454#FLJ2051 .6E-117 [Homo sapiens] Protein containing a pyrin domain, an NACHT domain, and leucine rich 0 repeats, may play a role i apoptotic or inflammatory signaling pathways 19 7512569CD1 g3511153 3.1E-62 [Hmo sapiens] cell division protein 712625#FTSJ1 2.4E-63 [Homo sapiens] Member of the FtsJ cell division family 647518#orf6.8880 2.9E-37 [Candida albicans] Member of the FtsJ-like methyltransferase family, which may interact with RNA, has moeerate similarity to a region of S. cerevisiae Spblp, which is a putative S- adenosyl-methionine-dependent methyltransferase Huang, S. et al., Specificity of cotranslational amino-terminal processing of proteins in yeast, Biochemistry 26, 8242-6 (1987). 20 7512570CD1 g3511153 3.1E-30 [Homo sapiens] cell division protein 712625#FTSJ1 2.5E-31 [Homo sapiens] Member of the FtsJ cell division family 647518#orf6.8880 7.2E-25 [Candida albicans] Member of the FtsJ-like methyltransferase family, which may interact with RNA, has moderate similarity to a region of S. cerevisiae Spb1p, which is a putative S- adenosyl-methionine-dependent methyltransferase Huang, S. et al., (supra) 21 7512846CD1 g5533375 0.0 [Homo sapiens] cell division cntrol protein 16 743488#CDC16 0.0 [Homo sapiens][Cytoplasic; Centrosome/spindle pole body] Cell division cycle 16, putative component of the anaphase promoting complex that localizes to the centrosome and the mitotic spindle, plays an essential role in the metaphase to anaphase transition and is downregulated in response to ionizing radiation Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: Tugendreich, S. et a., CDC27Hs colocalizes with CDC16Hs to the centrosome and mitotic spindle and is essential for the metaphase to anaphase transition, Cell 81, 261-8 (1995). 376401#cut9 2.0E-59 [Schizosaccharomyces pombe] Component of the APC, possible target of the spindle checkpoint Funabiki, H. et al., Cut2 proteolysis required for sister-chromatid seperation in fission yeast, Nature 381, 438-41 (1996). 22 7517814CD1 g3283051 3.3E-110 [Homo sapiens] cell division cycle protein 23 Zhao, N. et al., Human CDC23: cDNA cloning, mapping to 5q31, genomic structure, and evaluation as a candidate tumor suppressor gene in myeloid leukemias, Genomics 53, 184- 190 (1998) 2.5E-111 [Homo sapiens][Nuclear] Cell division cycle 23, putative component of the anaphase promoting complex (APC), which promotes the metaphase to anaphase transition via APC- catalyzed formation of cyclin B-ubiquitin conjugates, contains nine tetratrico peptide repeats Yu, H. et al., Identification of cullin homology region in a subunit of the anaphase promoting complex., Science 279, 1219 (1998). 6.9E-24 [Schizosaccharomyces pombe][Unknown] SUbunit of the APC/cyclosome complex with similarity to S. cerevisiae Cdc23p Yamashita, Y. M. et al., Fission yeast APC/cyclosome subunits, Cut20/Apc4 and Cut23/Apc8, in regulating metaphse-anaphase progression and cellular stress responses., enes Cells 4, 445-63 (1999). May, K. M. et al., Polo boxes and Cut23 (Apc8) mediate an interaction between polo kinase and the anaphase promoting complex for fission yeast mitosis., J Cell Biol 156, 23-8. (2002). 23 7519828CD1 g17512215 9.9E-134 [Homo sapiens] Cell division cycle 25C Bordo, D. and Bork, P. The rhodanese/Cdc25 phosphatase superfamily: Sequence-structure- function relations. EMBO Rep. 3, 741-746. (2002) Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: Ito, Y. et al. Expression of cdc25A and cdc25B proteins in thyroid neoplasms. Br J Cancer. 86, 1909-13. (2002) 661160#CDC25C 7.1E-137 [Homo sapiens][Protein phosphatase; Hydrolase] [Nuclear; Cytoplasmic] Cell division cycle 25C, a member of the Cdc25 family of protein tyrosine phosphatases that positively regulate the cell division cyce and dephosphorylates the cyclin B-dependent protein kinase CDC2 and thereby trigers entry into mitosis Strausfeld, U. et al., Dephosphorylation and activation of a p34cdc2/cyclin B complex in vitro by human CDC25 protein., Nature 351, 242-5 (1991). Korner, K. et al., In vivo structuere of the cell cycle-regulated human cdc25C promoter, J Biol Chem 275, 18676-81 (2000). Morris, M. C. et al., An essential phosphorylation-site domain of human cdc25C interacts with both 14-3-3 and cyclins., J Biol Chem 275, 28849-57 (2000). 584467#Cdc25c 1.4E-131 [Mus musculus][Protein phosphatase; Hydrolase] Cel division cycle 25 homolog C (S. cerevisiae), dual-specificity phosphatase, dephosphorylates the cyclin-dependent kinase Cdc2a and may thereby trigger entry of cells into mitosis, may play a role in gametogenesis Wu, S. et al., The distinct and developmentally regulated patterns of expression of members of the mouse Cdc25 gene family suggest differential functions during gametogenesis., Dev Biol 170, 195-206 (1995). Chen, M. S. et al., absence of apparent phenotype in mice lacking cdc25c protein phosphatase., Mol Cell Biol 21, 3853-61. (2001). Kanatsu-Shinohara, M. et al., Acquisition of meiotic competence in mouse oocytes: absolute amounts of p34(cdc2), cyclin B1, cdc25C, and wee1 in meiotically incompetent and competent oocytes., Biol Reprod 63, 1610-6. (2000). 24 7519833CD1 g17512215 1.6E-53 [Homo sapiens] Cell division cycle 25C Bordo and Bork, (supra) Ito, Y. et al., (supra) Table Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 661160#CDC25C 6.7E-31 [Homo sapeins][Protein phosphatase; Hydrolase] [Nuclear; Cytoplasmic] Cell division cycle 25C, a member of the Cdc25 family of protein tyrosine phosphatases that positively regulate the cell division cycle and dephosphorylates the cyclin B-dependent protein kinase CDC2 and thereby triggers entry into mitosis Strausfeld, U. et al., (supra) Korner, K. et al., (supra) Morris, M. C. et al., (supra) 584467A#Cdc25c 4.7E-30 [Mus musculus][Protein phosphatase; Hydrolase] Cell division cycle 25 homolog C (S. cerevisiae), dual-specificity phosphatase, deposphorylates the cyclin-dependent kinase Cdc2a and may thereby trigger entry of cells into mitosis, may play a role in gametogenesis Wu, S. et al., (supra) Chen, M. S. et al., (supra) Kanatsu-Shinohara, M. et al., (supra) 25 7519834CD1 g17512215 1.6E-227 [Homo sapiens] Cell division cycle 25C Bordo and Bork, (supra) Ito, Y. et al., (supra) 661160#CDC25C 6.9E-148 [Homo sapeins][Protein phosphatase; Hydrolase] [Nuclear; Cytoplasmic] Cell division cycle 25C, a member of the Cdc25 family of protein tyrosine phosphatases that positively regualte the cell division cycle and dephosphorylates the cyclin B-dependent protein kinase CDC2 and thereby triggers entry into mitosis Strausfeld, U. et al., (supra) Korner, K. et al., (supra) Morris, M. C. et al., (supra) Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 584467#Cdc25c 1.1E-138 [Mus musculus][Protein phosphatase; Hydrolase] Cell divison cycle 25 homolog C (S. cerevisiae), dual-specificity phosphatase, dephosphorylates the cyclin-dependent kinase Cdc2a and may thereby trigger entry fo cells into mitosis, may play a role in gametogenesis Wu, S. et al., (supra) Chen, M. S. et al., (supra) Kanatsu-Shinohara, M. et al., (supra) 26 7519939CD1 g189085 1.4E-125 [Homo sapiens] non-specific cross reacting antigen Tawaragi, Y. et al., Primary structure of nonspecific crossreacting antigen (NCA), a member of carcinomebryonic antigen (CEA) gene family, deduced from cDNA sequence, Biochem. biophys. Res. Commun. 150, 89-96 (1988) 789611#CEACA 1.1E-126 [Homo sapiens][Receptor (signaling][Plasma membrane] Carcinoembryonic antigen- M6 related cell adhesion molecule 6, a GPI-anchored member of the carcinoembryonic antigen family, involved in cell adhesion and granulocyte activation, mediates internalization of N. gonorrhoeae, upregulated in pancreatic carcinoma Screaton, R. A. et al., The specificity for the differentiation blocking activity of carcinoembryonic antigen resides in its glycophosphatidyl-inositol anchor, J Cell Biol 150, 613-26 (2000). Kinugasa, T. et al., Expression of four CEA family antigens (CEA, NCA, BGP and CGM2) in normal and cancerous gastric epithelial cells: up-regulation of BGP and CGM2 in carcinomas, Int J Cancer 76, 148-53 (1998). Ross, D. D. et al., The 95-kilodalton membrane glycoprotein overexpressed in novel multidrug-resistant breast cancer cells is NCA, the nonspecific cross-reacting antigen of carcinoembryonic antigen, Cancer Res 57, 5460-4 (1997). Robbins, P. F. et al., Definition of the expression of the human carcinoembryonic antigen and non-specific cross-reacting antigen in human breast and lung carcinomas, Int J Cancer 53, 892-7 (1993).

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 27 7520133CD1 g1853971 8.3E-178 [Homo sapiens] Mr 110,000 antigen Shimada, S. et al., Molecular cloning and characterization of the complementary DNA of an M(r) 110,000 antigen expressed by human gastric carcinoma cells and upregulated by gamma-interferon, Cancer Res. 54, 3831-3836 (1994) 428282#ADRM1 6.2E-179 [Homo sapiens][Plasma membrane] Cell membrane glycoprotein 110 kDa, putative integral plasma membrane glycoprotein, putative tumor antigen and is expressed on human gastric carcinoma cells; upregulated in response to IFNgamma (IFNG) Shimada, S. et al. (supra) 711654#Gp110 6.8E-173 [Rattus norvegicus] Integral plasma membrane glycoprotein, type I transmembrane glycoprotein that mediates Ca2+ -independent cell-cell adhesion, functions as a carrier in the transport of bile salts from hepatocytes into bile Lucka, L. et al., A short isoform of carcinoembryonic-antigen-related rat liver cell-cell adhesion molecule (C-CAM/gp110) mediates intercellular adhesion. Sequencing and recombinant functional analysis, Eur J Biochem 234, 527-35 (1995). Becker, A. et al., Characterisation of the ATP-dependent taurocholate-carrier protein (gp110) of the hepatocyte canalicular membrane, Eur J Biochem 214, 539-48 (1993). Muller, M. et al., ATP-dependent transport of taurocholate across the hepatocyte canalicular membrane mediated by a 110-kDa glycoprotein binding ATP and bile salt, J Biol Chem 266, 18920-6 (1991). 28 7520171CD1 g3298345 6.4E-84 [Homo sapiens] uroplakin 3 Yuasa, T. et al., Expression of uroplakin Ib and uroplakin III genes in tissues and peripheral blood of patients with transitional cell carcinoma, Jpn. J Cancer Res. 89, 879-82 (1998) Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 568106#UPK3 3.4E-84 [Homo sapiens][Unspecified membrane] Uroplakin 3, a urothelium-specific transmembrane protein, presence in peripheral blood may be a diagnostic marker for metastatic transitional cell carcinoma, may play a role in primary vesicoureteral reflux Yuasa, T. et al. (supra). Lobban, E. D. et al., uroplakin gene expression by normal and neoplastic human urothelium, Am J Pathol 153, 1957-67 (1998). Hu, P. et al., Ablation of uroplakin III gene results in small urothelial plaques, urothelial leakage, and vesicoureteral reflux, J Cell Biol 151, 961-72 (2000). 746537#Upk3 3.6E-71 [Mus musculus][Plasma membrane] Uroplakin 3, an integral membrane protein required for the formation of normal urothelial plaques in the urothelium, may contribute to the permeability barrier of the urinary tract, plays a role in primary vesicoureteral reflux Hu, P. et al. (supra) 29 7522453CD1 g7671653 6.9E-43 [Homo sapiens] dJ298J18.3 (novel protein similar to putative HIV-1 induced protein HIN-1 and Drosophila ovarian tumor locus protein OTU) 586233#Fmn 1.0E-15 [Mus musculus][Nuclear; Cytoplasmic] Formin (limb deformity), a protein that plays a role in embryonic pattern formation in limb and kidney, various forms are produced by alternative splicing Chen, D. C. et al., Formin isoforms are differentially expressed in the mouse embryo and are required for normal expression of fgf-4 and shh in the limb bud, Dev Suppl 121, 3151-62 (1995). Wynshaw-Boris, A. et al., The role of a single formin isoform in the limb and renal phenotypes of limb deformity, Mol Med 3, 372-84 (1997). 750720#FHOD2 5.1E-15 [Homo sapiens] Protein with high similarity to mouse Fmnl, which binds profilin and is involved in cell adhesion, contains a formin homology 2 domain, which is found in proteins that control rearrangements of the actin cytoskeleton Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: 30 7522454CD1 g7671653 2.5E-43 [Homo sapiens] dJ298J18.3 (novel protein similar to putative HIV-1 induced protein HIN-1 and Drosophila ovarian tumor protein OTU) 31 7517956CD1 g219539 3.4E-115 [Homo sapiens] CGM7 Kuroki, M. et al., Molecular cloning of nonspecific cross-reacting antigens in human granulocytes, J. Biol. Chem, 266, 11810-11817 (1991) 334640#CEACA 2.6E-116 [Homo sapiens][Plasma membrane; Unspecified membrane] Member of the CEA subgroup M4 of the immunoglobulin superfamily Muenzner, P. et al., Pathogenic Neisseria trigger expression of their carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1; previously CD66a) receptor on primary endothelial cells by activating the immediate early response transcription factor, nuclear factor-kappaB, J Biol Chem 276, 24331-40 (2001). Thompson, J. et al., A polymerase-chain-reaction assay for the specific identification of transcripts encoded by individual carcinoembryonic antigen (CEA)-gene-family members, Int J cancer 55, 311-9 (1993). 32 7517957CD1 g219539 3.7E-49 [Homo sapiens] CGM7 Kuroki, M. et al. (supra) 334640#CEACA 2.8E-50 [Homo sapiens][Plasma membrane; Unspecified membrane] Member of the CEA subgroup M4 of the immunoglobulin sperfamily Muenzner, P. et al. (supra) 710029#Ceacam1 6.0E-1 [Mus musculus][Adhesin/agglutinin][Plasma membrane] CEA-related cell adhesion molecule 1, a cell adhesion and signal transduction molecule, serves as a hepatitis virus receptor, negatively regulates tumor cell growth, may play roles in epithelial cell renewal and differentiation and in the immune response Dveksler, G. S. et al., Several members of the mouse carcinoembryonic antigen-related glycoprotein family are functional receptors for the coronavirus mouse hepatitis virus-A59, J Virol 67, 1-8 (1993).

Table 2 Polypeptide SEQ Incyte GenBank ID NO: Probability Annotation ID NO: Polypeptide ID or PROTEOME Score ID NO: Daniels, E. et al., biliary glycoprotein 1 expression during embryogenesis: correlation with jevents of epithelial differentiation, mesenchymal-epithelial interactions, absorption, and myogenesis, Dev Dyn 206, 272-90 (1996). Kunath, T. et al., Inhibition of colonic tumor cell growth by biliary glycoprotein, Oncogene 11, 2375-82 (1995). 33 7519293CD1 g807819 2.6E-146 [Homo sapiens] PDGF receptor beta-like tumor suppressor Fujiwara, Y. et al., Isolation of a candidate tumor suppressor gene on chromosome 8p21.3- p22 that is homologous to an extracellular domain of the PDGF receptor beta gene, Oncogene 10, 891-895 (1995) 343630#PDGFRL 1.9E-147 [Homo sapiens][Protein kinase; Transferase; Receptor (signaling)] Platelet-derived growth factor receptor-like, protein with similarity to the extracellular ligand-binding domain of PDGFRB; mutations in the gene are associated with hepatocellular carcinomas; colorectal cancers, and non-small cell lung cancers Fujiwara, Y. et al. (supra) Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites 1 7511094Cd1 190 S12 S110 S155 N101 N108 N114 S174 T67 T72 T121 2 7511619CD1 207 S6 S14 S95 S181 N4 N93 N119 domain present in chylins, TFIIB and Retinoblastoma- HMMER_SMART associated protein domain: K62-Q152 cell : cylin cell: Y2-K205 HMMER_TIGRFAM CYCLIN CELL CYCLE DIVISION PROTEIN BLIMPS_PRODOM PD02331: A74-V120, E141-D173 CYCLIN H MO15-ASSOCIATED PROTEIN CELL BLAST_PRODOM CYCLE DIVISION NUCLEAR P37 P34 PD030211: M1- M75 CYCLIN CELL CYCLE DIVISION PROTEIN C G1/S BLAST_PRODOM SPECIFIC NUCLEAR H HOMOLOG PD002975: P45- P123 CYCLIN; DM05249 BLAST_DOMO #P51946#10-277: H10-K175 #P51947#47-277: L48-K175 #P36613#24-286: A36-R185 3 7511720CD1 96 signal_cleavage: M1-A30 HMMER Signal Peptide: M1-A30, M11-A30 HMMER Cornichon protein: A6-S96 HMMER_PFAM Cytosolic domains: M1-A6, P80-S96 TMHMMER Transmembrane domains: A7-I29, L57-M79 Non-cytosolic domain: A30-Y56 PROTEIN TRANSMEMBRANE CORNCHON; BLAST_PRODOM DEVELOPMENTAL CORNICHON-LIKE ER-DERIVED VESICLES ERV14 ENDOPLASMIC PD008226: M1-W87 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites CORNICHON; DM04292 BLAST_DOMO #P53173#1-137: A7-Y91 #P49858#1-143: M1-W87 4 7511661CD1 420 S4 S44 S67 S119 N16 N405 Cell division protein: R47-D306 HMMER-PFAM SD294 S358 S406 T93 T183 T211 T236 GTP BINDING PROTEIN CELL DIVISION SEPTIN BLAST_PRODOM HOMOLOG CONTROL CYCLOE BRAIN H5 PD002565: R47-V56, T52-N308 CELL DIVISION CONTROL PROTEIN CYCLE GTP BLAST_PRODOM BINDING SEPTIN B PD131891: P318-E388 SEPTIN 2 CELL DIVISION GTP BINDING PD131893: BLAST_PRODOM E323-K416 do HCDC10; PEANUT; DM00875#JC2352#12-207: V31- BLAST_DOMO V56, T52-E210 do HCDC10; PEAUT; DM00875#P40797#123-320; V31- BLAST_DOMO V56, T52-T211 do SPAC8A4.07; HCDC10; PEANUT; SPR28; BLAST_DOMO DM00647#JC2352#209-324: T211-R327 do HCDC10; PEANUT; DM00875#P42208#18-215: V31- BLAST_DOMO V56, T52-E210 5 7512204CD1 239 S44 S96 S151 S1282 N57 N104 N111 signal_cleavage: M1-A64 SPSCAN S204 S219 T26 N126 T113 T121 T194 T198 T227 Signal Pptide: M1-V34 HMMER Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequencves, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites RECURSOR GLYCOPROTEIN SIGNAL PREGNANCY BLAST_PRODOM SPECIFIC CARCINOEMBRYONIC ANTIGEN IMMUNOGLOBULIN FOLD PREGNANCY MULTIGENE PD000677: D29-T113 CARINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#A40428#38-128: I38-Y129 CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#P40199#38-146: I38-H141 CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#A26902#38-146: I38-H141 CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#C40428#38-128: I38-A127 6 7512197CD1 77 S21 T70 BCL2 HOMOLOGOUS ANTAGONIST/KILIER BLAST_PRODOM APOPTOSIS REGULATOR TRANSMEMBRANE BAK BAK2 3D STRUCTURE PD0021493: M1-L71 7 7511656CD1 186 S21 S96 S132 S146 N92 N164 TUMOR COILED D52-LIKE D53 N8 CSPP28 MD52 R10 BLAST_PRODOM T42 T54 T62 T85 HD53 HD54+INS2 T143 T153 PD008422: K125-A142 PROTEIN D52 TUMOR D53 N8 CSPP28 MD52 R10 BLST_PRODOM HD53 HD54+INS2 PD008422: K15-A142 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequencesd, Domains and Motifs Analytical Methods ID Polypetide residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites 8 7512297CD1 753 S6 S26 S62 S115 N126 N320 N427 Signal Peptide: M1-S22 HMMER S155 S295 S342 S418 S436 S504 S624 S740 T70 T448 T492 T543 T566 T724 9 7512392CD1 204 S33 S36 S62 S101 N77 N94 signal_cleavage: M1-G26 SPSCAN Signal Peptide: M1-G26 HMMER Signal Peptide: A6-G26 HMMER Signal Peptide: P5-G26 HMMER Signal Peptide: M1-G29 HMMER Signal Peptide: L9-G26 HMMER Leucine-rich repeats, typical (most populated) subfamily: HMMER_SMART L124-G147, L100-G123, L75-G99 Leucine Rich Repeat: HMMER_PFAM E126-P1249, L102-G125, G78-S101 Leucine-rich repeat signature BLIMPS-PRINTS PR00019: L127-L140 10 7511589CD1 47 S2 S16 S20 T34 T35 11 7511699CD1 63 N38 signal_cleavage: M1-G18 SPSCAN Signal Peptide: M1-G18 HMMER Signal Peptide: M3-G18 HMMER Signal Peptide: M3-S20 HMMER Signal Peptide: M1-S20 HMMER Signal Peptide: M1-C24 HMMER Signal Peptide: M1-T23 HMMER FRIZZLED 6 PD060353: M1-T23 BLAST _PRODOM (P=4.7e-08) Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites 12 7512595CD1 891 S2 S84 S250 S382 N148 N169 ATP/GTP-binding site motif A (P-loop): G578-T585 MOTIFS S457 S473 S499 S582 S622 S786 S798 S817 S849 T123 T193 T269 T355 T485 T557 T631 T671 Y67 Y662 Y803 13 2803911CD1 1335 S48 S136 S178 N170 N736 N741 WD40 repeasts: R1154-D1194, S1200-D1240, S1283-S1329, HMMER_SMART S205 S236 S367 N758 N1281 E1052-K1097, S1246-N1281, R1012-D1050, N1105-D1151 S483 S590 S599 S743 S791 S903 S963 S11140 S1176 S1189 S1282 T126 T395 T548 T638 T714 T731 T865 T926 T937 T1011 T1071 T1086 T1121 T1152 T1163 T1206 T1257 WD domain, G-beta repeat: V1158-D1194, G1302-S1329, HMMER_PFAM R1203-D1240, D1080-K1097, V1029-D1050, S1246- N1281, L1115-D1151 G-protein beta WD-40 repeats IPB001680: T1086-K1097 BLIMPS_BLOCKS Beta G-protein (transducin) signature PR00319: 11227- BLIMPS_PRINTS P1241, P1263-Y1280 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sitesd and Databases NO: ID Sites PROTEIN TRANSMEMBRANE GNDIKII BLAST_PRODOM INTERGENIC REGION PUTATIVE GUANINE NUCLEOTIDE BINDING C57A7.11 TRANSPORT- PD043909: W49-E214, P232-Q281, I298-I397, R404-D512, Q215-C240 PROTEIN TRNSMEMBRANE PUTATIVE GUANNE BLAST_PRODOM NUCLEOTIDE BINDING C57A7.11 TRANSPORT GND1IKI1 INTERGENIC REGION PD043325: V652- E689, V802-I837, H947-K1221 PROTEIN TRANSMEMBRANE PUTATIVE GUANINE BLAST_PRODOM NUCLEOTIDE BINDING C57A7.11 TRANSPORT GND11K11 INTERGENIC REGION PD043911: E530- G628 14 7511806CD1 43 S27 15 7511853CD1 806 S37 S40 S125 S146 N228 N601 N675 signal_cleavage: M1-T16 SPSCAN S157 S230 S275 N739 N791 S393 S462 S523 S543 S591 S632 S638 S677 T69 T217 T574 T695 T772 T783 T793 T802 Y47 Y109 Y645 Leucine rich repeat, ribonuclease inhibitory type: N675-H702, HMMER_SMART S732-H759, S562-H589, D704-C731, A647-R674 SPLICE VARIANT RECEPTOR VASOPROESSIN RNI- BLAST_PRODOM LIKE SITE AII/AVP RECRUTMENT KIAA0926 CASPASE PD156095: M280-Q454 Table 3 SEQ Incyte Amino Acid Potential Potential Signasture Sequences, Domains and Motifs Analytical Methods ID Poypeptide Residues Phospyorylation Glycosylation Sites and Databases NO: ID Sites RBONUCLASE INHIBITORREPEAT LEUCINE BLAST_PRODOM REPEAT 3D-STRUCTURE PLACENTAL RIBONUCLEASE/ANGIOGENIN RAI RI RECEPTOR PD017636: L681-E776, L624-L729 MHC CLASS II TRANSACTIVATOR CIITA BLAST_PRODOM TRANSCRIPTION REGULATION ACTIVATOR ATP- BINDING DISEASE PD025523: Q72-L498 ATP/GTP-binding site motif A (P-loo): G80-T87 MOTIFS C-type lectin domain signature: C706-C731 MOTIFS 16 7512306CD1 60 S44 signal_cleavage: M1-G24 SPSCAN Signal Peptide: M1-S18,D M1-G21, M1-G24 HMMER Cytosolic domain: M1-S4 TMHMMER Transmembrane domain: L5-M27 Non-cytosolic domain: P28-Q60 PROTEIN INHIBITORY MELANOMA ACTIVITY BLAST_PRODOM DERIVED GROWTH REGULATORY PRECURSOR RETINOIC FACTOR PD012895: M1-S42 117 7512607CD1 275 S27 S114 S161 N34-N226 signal_cleavage: M1-G54 SPSCAN S190 S221 S240 T57 T65 T142 T169 T217 Cell division protein: Q39-S275 HMMER_PFAM GPT-BINDING PROTEIN CELL DIVISION SEPTIN BLASTpPRODOM HOMOLOG CONTROL CYCLOE BRAIN H5 PD002565: Q39-V263 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites HCDC10; PEANUT; DM00875# BLAST_DOMO P42207#16-212: V23-P216 P28661#125-322: V23-P216 JC2352#12-207: V23-P216 P42208#18-215: V23-P216 ATP/GTP-binding site motif A (P-loop): G49-S56 MOTIFS 18 7511851CD1 862 S37 S40 S125 S146 N228 N601 N682 signal_cleavage: M1-T16 SPSCAN S157 S230 S275 N731 N847 S393 S462 S523 S543 S591 S632 S638 S661 S733 T69 T217 T574 T679 T751 T814 T828 T839 T849 T858 Y47 Y109 Y645 Leucine rich repeat, ribonuclease inhibitor type: N731-H758, HMMER_SMART N788-H815, D760-S787, N675-S701, S562-H589, A647- N674 SPLICE VARIANT RECEPTOR VASOPRESSIN RNI- BLAST_PRODOM LIKE SITE AII/AVP RECRUITMENT KIAA0926 CASPASE PD156095: M280-Q454 RIBONUCLEASE INHIBITOR REPEAT LEUCINE BLAST_PRODOM REPEAT 3D-STRUCTURE PLACENTAL RIBONUCLEASE/ANGIOGENIN RAI RI RECEPTOR PD017636: L681-S787, L737-E832 MHC CLASS II TRANSACTIVATOR CIITA BLAST_PRODOM TRANSCRIPTION REGULATION ACTIVATOR ATP- BINDING DISEASE PD025523: Q72-L498 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites ATP/GTP-binding site motif A (P-loop): G80-T87 MOTIFS 19 7512569CD1 170 S60 T4 T97 T127 signal_cleavage: M1-S60 SPSCAN Y13 FtsJ-like methyltransferase: W21-Q167 HMMER_PFAM rrmJ: ribosomal RNA large subunit methyltransferase: D10- HMMER_TIGRFAM L155 PROTEIN INTERGENIC REGION C4F10.03C BLAST_PRODOM CHROMOSOME I JM23 R74.7 ORC2TIP1 KAR4PBN1 PD150824: M1-V42 PROTEIN CELL DIVISION FTSJ INTERGENIC REGION BLAST_PRODOM F45G2.9 J C4F10.03C CHROMOSOME PD149883: R44- A118 DIVISION; FTSJ; DM02985# BLAST_DOMO P38238#1-208: M1-D120 P25582#5-203: R3-D120 20 7512570CD1 109 S60 T4 Y13 N103 signal_cleavage: M1-S60 SPSCAN PROTEIN INTERGENIC REGION C4F10.03C BLAST_PRODOM CHROMOSOME I JM23 R74.7 ORC2TIP1 KAR4PBN1 PD150824: M1-V42 DIVISION; FTSJ; DM02985# BLAST_DOMO P38238#1-208: M1-I63 P25582#5-203: R3-Q61 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites 21 7512846CD1 569 S33 S62 S119 S127 N145 N427 N554 Tetratricopeptide repeats: E394-N427, A428-D461, S130- HMMER_SMART S152 S236 S288 C163 S348 S535 S544 S563 T177 T274 T327 T500 T503 T523 T530 T534 Y19 Y69 Y74 Y286 Y416 TPR Domain: E394-N427, A428-D461, S130-C163 HMMER_PFAM BTB (also known as BR-C/Ttk) domain proteins. PF00651: BLIMPS_PFAM Y53-L65 PROTEIN REPEAT DOMAIN TPR NUCLEAR. PD00126: BLIMPS_PRODOM G435-L455, I139-D144 CELL DIVISION PROTEIN CYCLE MITOSIS REPEAT BLAST_PRODOM TPR DOMAIN NUCLEAR CONTROL PD024394: L3- C250 CDC16HS PD071385: L455-T569 BLAST_PRODOM TPR REPEAT DM03987 BLAST_DOMO #A56519#1-559: M1-G366, W367-P510 #P41889#82-636: L14-S429 #P09798#228-796: L3-L455 Leucine zipper pattern: L400-L421 MOTIFS 22 7517814CD1 229 S51 S111 S159 T77 Signal Peptide: M1-S21, M1-L29, M7-S21 HMMER T214 Y99 CELL DIVISION CYCLE PROTEIN 23 CELL DIVISION BLAST_PRODOM PD179957: L20-L227 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites 23 7519828CD1 439 S8 S20 S78 S198 N59 N76 Rhodanese Homology Domain: A277-E391 HMMER_SMART S216 S262 S281 S315 S347 T9 T67 T89 T236 T333 Y270 Rhodanese-like domain: L280-E388 HMMER_PFAM M-phase inducer phosphatase signature PR00716: R253- BLIMPS_PRINTS Y270, V271-F291, V293-L313, K335-R355, N365-F386, C393-L410, L410-E428 PHOSPHATASE CELL DIVISION HYDROLASE BLAST_PRODOM MPHASE INDUCER MITOSIS MULTIGENE FAMILY PROTEIN PD008585: M1-N155, S148-R223 PROTEIN TRANSFERASE SULFURTRANSFERASE BLAST_PRODOM THIOSULFATE PHOSPHATASE HYDROLASE PUTATIVE CELL DIVISION MITOSIS PD002675: E289- F386 do PHOSPHATASE; INDUCER; M-PHASE; CDC25; BLAST_DOMO DM01001#P30307#199-469: L158-D436 DM01001#I49126#185-442: E184-K435 DM01001#P48967#184-442: E184-K435 24 7519833CD1 141 S8 S20 S78 T9 T67 N59 N76 PHOSPHATASE CELL DIVISION HYDROLASE BLAST_PRODOM T89 MPHASE INDUCER MITOSIS MULTIGENE FAMILY PROTEIN PD008585: M1-H119 25 7519834CD1 421 S8 S20 S78 S164 N59 N76 N153 Rhodanese Homology Domain: A259-E373 HMMER_SMART S211 S244 S263 S297 S329 T9 T67 T89 T184 T315 Y252 Rhodanese-like domain: L262-E370 HMMER_PFAM Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites M-phae inducer phosphatase signature PR00716: S235- BLIMPS_PRINTS Y252, V253-F273, V275-L295, K317-R337, N347-F368, C375-L392, L392-E410 PHOSPHATASE CELL DIVISION HYDROLASE BLAST_PRODOM MPHASE INDUCER MITOSIS MULTIGENE FAMILY PROTEIN PD008585: M1-L174, L206-E271 PROTEIN TRANSFERASE SULFURTRANSFERASE BLAST_PRODOM THIOSULFATE PHOSPHATASE HYDROLASE PUTATIVE CELL DIVISION MITOSIS PD002675: E271- F368 do PHOSPHATASE; INDUCER; M-PHASE; CDC25; BLAST_DOMO DM01001#P30307#199-469: V154-D418 DM01001#P49126#185-442: P172-K417 DM01001#P48967#184-442: P172-K417 DM01001#P30308#273-549: N150-K417 26 7519939CD1 256 S96 S188 S204 N104 N111 N115 Signal Peptide: M1-A34 HMMER S247 T33 T113 N152 N173 N197 Y213 N224 Immunoglobulin: N152-L234, S40-Y141 HMMER_SMART Immunoglobulin domain: K160-I217, G48-V124 HMMER_PFAM Vascular cell adhesion molecule-1 (VCAM-1) signature BLIMPS_PRINTS PR01474: D159-V171, N195-V205, K206-N219 PRECURSOR GLYCOPROTEIN SIGNAL PREGNANCY- BLAST_PRODOM SPECIFIC CARCINOEMBRYONIC ANTIGEN IMMUNOGLOBULIN FOLD PREGNANCY MULTIGENE PD000677: N29-T113 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation Glycosylation Sites and Databases NO: ID Sites PRECURSOR GLYCOPROTEIN SIGNAL PREGNANCY- BLAST_PRODOM SPECIFIC IMMUNOGLOBULIN FOLD MULTIGENE FAMILY PREGNANCY BETA1 PD002253: P145-E216 ANTIGEN PRECURSOR GLYCOPROTEIN SIGNAL BLAST_PRODOM IMMUNOGLOBULIN FOLD CARCINOEMBRYONIC GPI ANCHOR TRANSMEMBRANE BILIARY PD149875: Q114-E143 CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#A26902#38-146: I38-P147 DM00372#P40199#38-146: I38-P147 IMMUNOGLOBULIN BLAST_DOMO DM00001#A26902#148-232: S148-V233 DM00001#P40199#148-232: S148-V233 Actinin-type actin-binding domain signature 1: Q172-N181 MOTIFS 27 7520133CD1 369 S19 S32 S90 S102 N162 signal_cleavage: M1-G17 SPSCAN S143 S208 S224 S354 T114 T323 T359 T362 T365 T369 Dopamine D4 receptor signature PR00569: G179-G196, BLIMPS_PRINTS T221-A235, L343-R364 PROTEIN CELL MEMBRANE GLYCOPROTEIN BLAST_PRODOM C56G2.7 CHROMOSOME III T9J22.26 PD022277: S15-I172 PD021964: S253-Q338 28 7520171CD1 212 S76 N74 N139 signal_cleavage: M1-A18 SPSCAN Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation GLycosylation Sites and Databases NO: ID Sites Signal Peptide: M1-G16 HMMER Signal Peptide: M1-A18 HMMER Signal Peptide: M1-N20 HMMER Signal Peptide: M1-P23 HMMER Signal Peptide: M1-L21 HMMER UROPLAKIN III UROPLKIN PRECURSOR UPIII BLAST_PRODOM GLYCOPROTEIN TRANSMEMBRANE SIGNAL PD127194: M1-Y162 29 7522453CD1 720 S59 S142 S162 N104 N243 N274 Lysosome-associated membrane glycoprotein (Lamp) BLIMPS_BLOCKS S231 S244 S276 N401 N632 IPB002000: A499-P512 S281 S285 S356 S403 S413 S554 S630 T316 Y48 Y381 Wilm's tumour protein signature PR00049: G498-P512 BLIMPS_PRINTS Proline rich extensin signature PR01217: P502-P523 BLIMPS_PRINTS OVARIAN TUMOR LOCUS PROTEIN PD146246: P34- BLAST_PRODOM E62 A453-S583 ACROSIN BLAST_DOMO DM03631#P12978#10-163: L501-Y567 DM03631#P48038#288-430: S474-A529 do FORMIN; BLAST_DOMO DM04565#Q05859#5-1205: K408-P553, A451-Q560 DM04565#Q05860#176-1467: K408-P553, A451-Q560 30 7522454CD1 641 S59 S142 S162 N104 N243 N274 S231 S244 S276 N401 N553 S281 S285 S356 S403 S413 S551 T316 Y48 Y381 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation GLycosylation Sites and Databases NO: ID Sites 31 7517956CD1 256 S44 S96 S203 S214 N57 N104 N111 signal_cleavage: M1-A64 SPSCAN T113 T121 T179 N126 Signal Peptide: M1-V34 HMMER Cytosolic domain: S177-Q256 TMHMMER Transmembrane domain: A154-L176 Non-cytosolic domain: M1-V153 PRECURSOR GLYCOPROTEIN SIGNAL BLAST_PRODOM PREGNANCYSPECIFIC CARCINOEMBRYONIC ANTIGEN IMMUNOGLOBULIN FOLD PREGNANCY MULTIGENE PD000677: H29-T113 ANTIGEN CARCINOEMBRYONIC CGM1 PRECURSOR BLAST_PRODOM CD66D IMMUNOGLOBULIN FOLD SIGNAL GLYCOPROTEIN TRANSMEMBRANE PD030936: R181- H228 CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM00372#A40428#38-128: I38-Y129 DM00372#P40199#38-146: I38-P149 DM02898#A40428#I49-227: P149-Y222 DM02898#P40198#I49-235: P149-H228 32 7517957CD1 136 S83 S94 T59 signal_cleavage: M1-A62 SPSCAN Cytosolic domain: S57-Q136 TMHMMER Transmembrane domain: A34-L56 Non-cytosolic domain: M1-V33 ANTIGEN CARCINOEMBRYONIC CGM1 PRECURSOR BLAST_PRODOM CD66D IMMUNOGLOBULIN FOLD SIGNAL GLYCOPROTEIN TRANSMEMBRANE PD030936: R61- H108 Table 3 SEQ Incyte Amino Acid Potential Potential Signature Sequences, Domains and Motifs Analytical Methods ID Polypeptide Residues Phosphorylation GLycosylation Sites and Databases NO: ID Sites CARCINOEMBRYONIC ANTIGEN PRECURSOR BLAST_DOMO AMINO-TERMINAL DOMAIN DM02898#A40428#149-227: P29-Y102 DM02898#P40198#149-235: P29-H108 33 7519293CD1 277 S118 S182 S187 N132 signal_cleavage: M1-E17 SPSCAN T41 T111 T134 T221 T225 T236 T245 T272 Signal Peptide: M1-A15 HMMER Signal Peptide: M1-D18 HMMER Signal Peptide: M1-G21 HMMER Signal Peptide: M1-E17 HMMER Immunoglobulin: A180-S277, A81-P170 HMMER_SMART Immunoglobulin domain: G188-A261, G89-V145 HMMER_PFAM PDGF RECEPTOR BETA-LIKE TUMOR SUPPRESSOR BLAST_PRODOM PD104162: M1-T168 PD179025: V220-S277 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 34/7511094CB1 1-234, 1-409, 1-446, 1-450, 1-466, 1-516, 1-525, 1-551, 1-567, 1-575, 2-576, 383-1027, 470-1000, 481-1004, 507-1168, 532-934, 533-1010, 1422 550-1000, 709-1303, 776-1402, 815-1308, 820-1371, 902-1422, 905-1421, 941-1364, 950-1395, 1086-1422 35/7511619CB1 1-267, 1-455, 1-466, 1-511, 1-515, 1-528, 1-555, 1-597, 1-613, 1-622, 1-630, 1-653, 1-660, 1-1476, 8-244, 8-462, 8-475, 8-506, 9-320, 11- 1476 292, 13-300, 19-300, 22-302, 27-296, 32-295, 32-298, 33-291, 37-233, 39-297, 42-284, 43-287, 46-305, 46-307, 51-316, 51-366, 52-229, 54- 626, 55-132, 55-660, 56-517, 56-533, 57-380, 61-334, 61-428, 61-635, 61-658, 63-336, 65-263, 65-284, 65-337, 71-321, 71-626, 73-294, 73- 347, 73-355, 79-338, 82-380, 86-303, 88-305, 91-632, 97-329, 97-400, 104-660, 111-529, 112-658, 119-660, 122-660, 126-315, 128-364, 133-406, 133-415, 144-360, 144-613, 159-384, 165-458, 167-419, 210-466, 212-452, 235-473, 235-480, 256-509, 288-557, 316-768, 316- 834, 359-616, 409-644, 427-660, 657-1065, 657-1080, 657-1114, 657-1115, 657-1119, 657-1128, 669-1019, 671-1079, 671-1115, 672-909, 672-1115, 673-1127, 685-928, 688-980, 701-1115, 701-1124, 702-1116, 721-1120, 762-1114, 771-1054, 771-1117, 773-1109, 775-1084, 802-1115, 809-1102, 813-906, 836-1114, 867-1124, 868-1115, 880-1115, 940-1116, 986-1432, 992-1237, 997-1242, 998-1355, 1002-1114, 1010-1124, 1037-1355, 1070-1344, 1087-1344, 1119-1355, 1179-1476, 1221-1473, 1290-1355 36/7511720CB1/ 1-251, 19-249, 19-1264, 31-296, 36-333, 38-307, 40-349, 51-299, 54-299, 55-346, 56-276, 57-249, 58-286, 58-351, 60-353, 60-354, 61-182, 1280 61-303, 61-309, 61-336, 61-354, 62-268, 62-329, 62-333, 62-345, 62-346, 62-347, 62-354, 63-182, 63-312, 64-307, 64-347, 66-302, 69-312, 71-242, 73-349, 74-332, 74-342, 74-354, 81-287, 81-324, 81-329, 82-327, 85-349, 86-354, 87-306, 87-339, 87-350, 87-354, 89-329, 89-336, 91-352, 92-331, 92-340, 92-343, 94-309, 94-319, 94-330, 94-332, 94-338, 94-344, 94-350, 94-351, 94-354, 94-373, 95-242, 95-312, 95-344, 95-354, 96-354, 97-354, 98-352, 100-315, 101-308, 101-325, 102-312, 102-349, 102-354, 103-234, 103-347, 106-354, 107-236, 107-296, 111-354, 120-293, 160-692, 194-354, 354-601, 354-735, 355-620, 355-643, 355-651, 355-652, 363-606, 365-611, 365-839, 366-632, 366- 879, 372-692, 376-1262, 377-633, 378-1035, 380-819, 386-634, 405-593, 419-640, 420-686, 422-905, 426-689, 428-659, 428-661, 432-684, 435-709, 437-685, 442-713, 442-881, 448-694, 452-617, 452-664, 455-677, 457-759, 464-761, 481-927, 490-764, 491-959, 514-805, 520-794, 533-770, 543-1190, 544-933, 545-836, 552-785, 556-1262, 564-833, 575-1002, 578-832, 578-835, 580-788, 582-841, 586-735, 586-1036, 586-1260, 590-1275, 603-786, 603-900, 603-1059, 616-844, 619-1181, 622-1262, 625-854, 629-1198, 645-737, 645-842, 645-921, 645-1249, 660-905, 602-1262, 666-861, 667-1260, 671-914, 673-1135, 673-1279, 674-871, 674-1029, 677-934, 678-925, 681-927, 697-951, 697-957, 704-997, 717-986, 721-1261, 727-1262, 731-988, 735-1253, 744-1215, 746-1230, 749-996, 748-1033, 761-1066, 763-1280, 766-1023, 769-1011, 769-1024, 776-1280, 778-1280, 779-1280, 786-1216, 786-1280, 788-1097, 793-1260, 794-1023, 794-1051, 794-1070, 796-1074, 796-1110, 796-1255, 796-1261, 798-1280, 801-1120, 802-1260, 802-1280, 803-1223, 804-1259, 805-1269, 806-1280, 807-1036, 807-1068, 807-1260, 807-1261, 807-1270, 808-1267, 809-1255, 810-1278, 811-1280, 812-1268, 815-1267, 816-1267, 817-1260, 818-1262, 820-1100, 822-1262, 823-1275, 827-1077, 827-1097, 828-1081, 829-1266.

Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 832-1263, 834-1280, 835-1260, 836-1086, 837-1267, 839-1260, 839-1267, 839-1280, 841-1263, 842-1261, 843-1260, 847-1280, 848-1268, 849-1267, 852-1266, 854-1174, 857-1089, 857-1279, 859-1260, 864-1279, 867-1141, 874-1256, 883-1081, 887-1261, 888-1135, 888-1146, 888-1230, 888-1270, 888-1272, 889-1260, 894-1011, 894-1260, 895-1267, 899-1260, 899-1263, 903-1262, 904-1190, 906-1280, 907-1168, 909-1267, 911-1202, 912-1116, 914-1260, 914-1263, 914-1267, 917-1121, 917-1198, 919-1208, 938-1260, 947-1212, 950-1200, 959-1267, 960-1116, 967-1260, 968-1260, 971-1053, 971-1232, 973-1267, 983-1225, 992-1223, 992-1252, 992-1262, 998-1252, 1017-1260, 1029- 1280, 1030-1262, 1038-1262, 1043-1273, 1051-1262, 1052-1263, 1061-1267, 1067-1260, 1070-1224, 1073-1260, 1078-1267, 1095-1244, 1101-1215, 1107-1278, 1112-1280, 1143-1261, 1182-1280 37/7511661CB1/ 1-249, 38-1574, 79-249, 108-224, 122-199, 122-220, 122-249, 123-667, 123-719, 123-792, 123-903, 124-452, 144-249, 202-1023, 249-416, 1589 249-436, 249-441, 249-492, 249-499, 249-501, 249-503, 249-504, 249-510, 249-528, 249-535, 249-540, 249-641, 249-645, 249-655, 240- 663, 249-730, 249-768, 249-782, 249-783, 249-792, 249-798, 249-799, 249-815, 249-816, 249-825, 249-829, 249-832, 249-838, 249-884, 249-894, 249-900, 250-888, 253-413, 254-695, 256-853, 262-53,1 271-879, 285-590, 290-951, 293-1032, 302-1032, 304-846, 306-568, 308- 543, 308-550, 310-623, 320-651, 331-519, 337-609, 342-588, 343-1064, 352-926, 356-1177, 359-552, 362-613, 380-1016, 392-956, 397- 1016, 400-642, 410-677, 415-688, 420-620, 420-660, 420-904, 421-726, 422-944, 425-657, 425-676, 428-992, 429-669, 429-679, 431-1059, 434-971, 446-755, 460-840, 464-750, 466-742, 469-657, 474-741, 479-1175, 483-710, 488-728, 524-1240, 525-783, 528-1173, 529-809, 529-1065, 533-803, 534-683, 541-643, 541-772, 542-814, 546-795, 554-853, 561-841, 562-813, 565-1114. 569-854, 571-850, 574-784, 575-1173, 582-851, 583-870, 588-830, 588-1157, 605-855, 609-854, 609-873, 614-860, 616-874, 616-1039, 617-1401, 620-852, 620-909, 622-890, 628-828, 628-1115, 629-862, 629-878, 629-883, 635-855, 635-1416, 636-838, 643-803, 646-1159, 649-907, 651-858, 660-1474, 665-916, 667-903, 667-905, 667-912, 667-937, 667-1141, 675-965, 677-899, 682-1314, 683-922, 683-951, 683-1237, 684-914, 700-947, 708-1016, 712-972, 717-959, 717-1316, 717-1317, 719-993, 722-1047, 724-1142, 726-1324, 728-1004, 734- 967, 735-1027, 735-1053, 738-1348, 742-1073, 749-1115, 750-1022, 758-1052, 764-1339, 765-1013, 766-1020, 774-907, 774-1013, 777- 1049, 791-1354, 792-1150, 798-1386, 804-1104, 804-1475, 805-1096, 808-1091, 809-1115, 811-1446, 813-1065, 813-1070, 813-1078, 819- 1091, 822-1391, 829-1073, 844-1056, 844-1175, 851-1378, 861-1077, 861-1310, 863-1456, 868-1101, 872-1120, 877-1228, 880-1476, 883- 1374, 885-1307, 886-1163, 886-1476, 893-1153, 898-1093, 900-1472, 909-1455, 917-1195, 919-1192, 920-1196, 921-1185, Table 4 Polynculeotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 936-1477, 943-1478, 944-1191, 946-1187, 950-1160, 950-1185, 950-1186, 950-1187, 950-1211, 950-1236, 952-1204, 956-1077, 956-1185, 956-1238, 956-1244, 960-1219, 962-1231, 963-1197, 966-1258, 969-1228, 972-1573, 978-1246, 978-1497, 979-1243, 982-1263, 982-1572, 984-1284, 990-1265, 993-1120, 993-1190, 993-1292, 1000-1524, 1004-1267, 1004-1565, 1005-1312, 1014-1248, 1014-1295, 1014-1300, 10151312, 1017-1037, 1023-1317, 1032-1247, 1035-1576, 1035-1589, 1039-1277, 1039-1469, 1046-1325, 1057-1361, 1064-1269, 1081- 1276, 1086-1331, 1087-1370, 1087-1392, 1088-1332, 1088-1378, 1094-1319, 1094-1400, 1099-1325, 1102-1245, 1105-1360, 1105-1411, 1111-1354, 1124-1379, 1128-1373, 1131-1371, 1137-1420, 1137-1429, 1151-1397, 1153-1367, 1153-1420, 1153-1428, 1155-1466, 1156- 1349, 1156-1420, 1162-1351, 1162-1396, 1162-1400, 1162-1404, 1170-1475, 1171-1470, 1172-1407, 1172-1479, 1174-1456, 1174-1482, 1174-1528, 1179-1426, 1181-1442, 1190-1470, 1191-1481, 1192-1469, 1192-1589, 1193-1468, 1195-1589, 1212-1432, 1213-1440. 1214-1476, 1217-1401, 1220-1482, 1224-1532, 1225-1475, 1226-1504, 1238-1466, 1238-1475, 1240-1431, 1240-1486, 1248-1486, 1248- 1513, 1251-1449, 1252-1486, 1271-1528, 1280-1362, 1288-1485, 1292-1405, 1292-1443, 1295-1471, 1310-1559, 1324-1501, 1333-1589, 1496-1589 38/7512204CB1/ 1-244, 1-1038, 3-269, 3-327, 3-374, 3-466, 4-262, 4-264, 4-438, 14-310, 78-908, 79-297, 159-1014, 584-1029, 620-1038, 636-966, 653- 1038 1004, 886-1029 39/7512197CB1/ 1-239, 1-278, 3-255, 5-300, 5-1257, 17-416, 18-220, 18-255, 18-263, 18-399, 28-291, 30-311, 39-293, 42-335, 46-150, 46-170, 46-268, 58- 1257 283, 64-246, 225-347, 252-455, 404-873, 460-550, 460-695, 460-704, 460-772, 460-898, 460-1109, 467-729, 471-983, 485-1139, 488-637, 492-1133, 514-879, 525-960, 527-941, 528-767, 548-1170, 549-1217, 550-828, 551-827, 551-832, 551-837, 551-857, 551-861, 553-841, 564-779, 569-1038, 581-848, 581-851, 587-879, 601-1138, 601-1173, 601-1189, 605-1161, 626-825, 632-1257, 658-1130, 671-1184, 676- 1177, 683-974, 683-1205, 690-957, 694-928, 718-960, 724-1137, 724-1242, 725-1230, 726-1243, 772-989, 792-1074, 792-1243, 799-1054, 799-1243, 805-1242, 815-1257, 822-1100, 825-1242, 846-1192, 864-1241, 864-1243, 870-1242, 871-1242, 872-1242, 881-1129, 881-1243, 891-1123, 891-1125, 891-1233, 897-1171, 939-1097, 962-1242, 965-1239, 983-1238, 989-1243, 989-1257, 1000-1242, 1008-1242. 1011- 1243, 1024-1160, 1047-1242, 1072-1243, 1078-1215, 1094-1255, 1094-1257, 1095-1257, 1130-1234, 1131-1243, 1132-1242, 1165-1242, 1172-1257 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 40/7511656CB1/ 1-235, 1-267, 4-313, 5-208, 5-263, 9-235, 9-241, 9-2274, 13-120, 13-128, 17-321, 20-274, 25-296, 30-297, 30-337, 30-338, 34-298, 34-347, 2275 35-303, 38-241, 38-348, 39-239, 39-274, 39-298, 39-320, 40-277, 42-298, 43-294, 46-302, 52-361, 60-213, 65-340, 68-316, 73-363, 78-230, 78-492, 82-314, 84-381, 84-409, 86-373, 88-327, 88-412, 89-375, 90-396, 91-277, 92-401, 96-334, 97-359, 100-323, 133-693, 147-374, 157- 429, 196-349, 196-452, 196-823, 233-416, 234-372, 234-418, 238-406, 247-1071, 335-600, 347-983, 426-702, 426-709, 426-936, 426-978, 442-710, 444-1144, 464-759, 465-1004, 466-1047, 478-623, 485-718, 517-817, 520-767, 522-1166, 525-875, 525-1111, 526-1153, 533-781, 548-837, 575-748, 575-1046, 575-1091, 575-1093, 576-783, 576-826, 579-1124, 579-1185, 600-1229, 609-833, 609-859, 617-883, 621- 1215, 626-1367, 630-884, 683-1184, 640-827, 641-1217, 649-938, 657-901, 661-949, 665-1392, 666-1345, 667-950, 667-1169, 676-1256, 680-1000, 683-966, 685-1350, 689-964, 705-1366, 707-1112, 715-944, 715-1398, 720-986. 722-1019, 727-967, 737-1409, 741-1312, 744-1004, 752-920, 753-1345, 760-1333, 761-1030, 762-1268, 766-1317, 770-1039, 780-927, 780- 1257, 786-1042, 786-1384, 786-1391, 787-1309, 790-1349, 792-1235, 803-1362, 805-1047, 807-1065, 807-1647, 810-1445, 814-1268, 824- 1012, 825-1207, 831-1101, 833-1260, 836-1341, 845-1093, 849-1390, 857-1162, 857-1167, 859-1058, 859-1127, 859-1153, 859-1496, 876- 1178, 886-1442, 888-1369, 897-1358, 898-1160, 904-1262, 904-1506, 908-1179, 909-1191, 912-1035, 913-1201, 915-1163, 928-1140, 930- 1199, 950-1132, 952-1638, 962-1198, 962-1594, 980-1103, 981-1297, 984-1450, 991-1381, 991-1390, 997-1514, 1000-1308, 1000-1649, 1002-1159, 1005-1203, 1005-1356, 1005-1461, 1006-1280, 1009-1217, 1009-1663, 1012-1364, 1016-1291, 1016-1720, 1017-1465, 1018- 1434, 1019-1306, 1019-1393, 1022-1292, 1023-1599, 1024-1707, 1031-1287, 1031-1303, 1033-1358, 1034-1839, 1035-1260, 1040-1330, 1042-1318, 1042-1348, 1047-1758, 1061-1963, 1070-1777, 1074-1367, 1075-1630, 1083-1623, 1084-1691, 1087-1704, 1091-1674. 1093-1330, 1110-1338, 1116-1392, 1126-1659, 1127-1782, 1128-1814, 1135-1590, 1148-1345, 1148-1361, 1148-1414, 1149-1389, 1149- 1640, 1151-1281, 1152-1779, 1159-1377, 1162-1563, 1169-1735, 1177-1657, 1187-1761, 1189-1447, 1194-1804, 1196-1793, 1200-1736, 1202-1479, 1205-1446, 1207-2101, 1208-1490, 1208-1753, 1216-1444, 1216-1477, 1216-1479, 1216-1663, 1217-1488, 1218-1823, 1228- 1372, 1230-1540, 1237-1475, 1237-1482, 1238-1471, 1239-1771, 1240-1726, 1251-1482, 1252-1764, 1257-1670, 1262-1705, 1265-1509, 1265-1536, 1265-1650, 1266-1409, 1266-1471, 1267-1498, 1274-1518, 1275-1662, 1286-1724, 1287-1436, 1287-1579, 1287-1663, 1287- 1865, 1289-1516, 1289-1539, 1290-1735, 1294-1572, 1295-1513, 1295-1635, 1297-1594, 1299-1733, 1301-2103, 1305-1916, 1312-1595, 1316-1585, 1328-1598, 1330-1599, 1331-1587, 1332-1567, 1335-1547, 1335-1596, 1335-1616, 1336-1450, 1336-1489, 1336-1563, 1336- 1805, 1336-1993, 1337-1546, 1337-1596, 1337-1627, 1338-1576, 1338-1581, 1338-1599, 1338-1632, 1338-1636, 1338-1863, 1338-1959, 1342-1590.

Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 1345-1636, 1349-1648, 1354-1617, 1355-1599, 1356-1777, 1357-1623, 1357-1630, 1357-1654, 1357-1777, 1364-1579, 1374-1890, 1377- 1821, 1379-1596, 1379-1624, 1379-1662, 1380-1680, 1380-1681, 1380-1686, 1390-1924, 1390-2131, 1390-2262, 1397-1679, 1397-1987, 1398-1669, 1398-1960, 1402-1567, 1402-1661, 1402-1687, 1403-1637, 1403-1657, 1404-1632, 1404-1665, 1404-1693, 1404-2054, 1408- 1661, 1418-1649, 1421-1768, 1421-2026, 1422-1644, 1422-1966, 1423-1688, 1424-1695, 1428-1618, 1429-2261, 1430-1692, 1431-1552, 1431-2017, 1433-1874, 1434-1808, 1437-1895, 1450-1702, 1451-1771, 1454-1723, 1463-1708, 1465-1917, 1467-2161, 1469-1772, 1470- 1795, 1471-1897, 1476-1759, 1476-1843, 1476-2032, 1478-1748, 1478-1777, 1478-2163, 1481-1778, 1483-1734, 1483-1770, 1483-1774, 1485-1713, 1485-1745, 1496-2262, 1501-1697, 1502-1769, 1502-1782, 1510-1761, 1510-2262, 1512-1688, 1525-1785, 1527-2189, 1530- 1852, 1530-1882, 1533-2018, 1534-1821, 1534-2259, 1536-1795, 1539-1735, 1540-2225, 1542-2207, 1548-2262, 1558-2210, 1569-2083, 1584-2241, 1590-1862, 1591-1826, 1591-1835, 1591-1874, 1592-1835, 1592-1841, 1593-1822, 1593-2150, 1595-1883, 1597-2267, 1601-1839, 1602- 1853, 1602-1857, 1603-1866, 1605-1889, 1608-2007, 1615-1864, 1615-1922, 1618-1867, 1625-1903, 1627-1921, 1628-1830, 1633-1816, 1639-1896, 1647-2262, 1654-1864, 1654-1879, 1658-1905, 1659-2043, 1660-1930, 1662-1914, 1663-2262, 1664-1885, 1671-1887, 1675- 2153, 1679-1923, 1691-2216, 1693-2216, 1697-1831, 1697-1972, 1700-2166, 1704-1972, 1706-1955, 1706-1985, 1716-1991, 1716-2022, 1719-2005, 1720-2270, 1734-1998, 1735-1964, 1736-1981, 1436-2020, 1739-2223, 1743-2236, 1744-2111, 1744-2262, 1749-2014, 1749- 2039, 1753-2025, 1757-2247, 1760-2024, 1762-2275, 1764-2275, 1766-1880, 1768-1970, 1771-2262, 1772-2275, 1773-2225, 1780-2275, 1786-2262, 1796-2098, 1798-2269, 1799-2223, 1801-2257, 1804-1999, 1804-2000, 1804-2032, 1085-2259, 1808-2062, 1809-2275, 1812- 2222, 1812-2275, 1815-2053, 1816-2264, 1817-2082, 1820-2063, 1820-2194, 1822-2275, 1824-2257, 1824-2262, 1828-2260, 1832-2260, 1833-2262, 1833-2268, 1834-2144, 1835-2266, 1837-2227, 1839-2262, 1841-2154, 1843-2120, 1843-2144, 1843-2265, 1845-1989, 1845-2128, 1846- 2262, 1846-2269, 1846-2273, 1848-2266, 1849-2275, 1850-2267, 1854-2265, 1855-2106, 1855-2107, 1862-2247, 1863-2097, 1864-2262, 1868-2262, 1869-1968, 1869-2274, 1870-2270, 1871-2269, 1875-2262, 1876-2135, 1877-2223, 1883-2124, 1886-2275, 1891-2275, 1893- 2116, 1893-2275, 1894-2260, 1900-2264, 1902-2264, 1903-2262, 1905-2262, 1910-2275, 1931-2275, 1935-2267, 1939-2193, 1940-2246, 1941-2261, 1943-2265, 1946-2262, 1947-2226, 1948-2054, 1950-2208, 1951-2267, 1971-2269, 1974-2268, 1975-2228, 1982-2227, 1982 2262, 1982-2265, 1983-2190, 1983-2262, 1984-2268, 1985-2262, 1986-2274, 1986-2275, 1990-2112, 1990-2265, 1996-2265, 2004-2262, 2021-2272, 2028-2275, 2039-2262, 2041-2275, 2056-2262, 2076-2264, 2081-2267, 2112-2273, 2127-2264, 2136-2262, 2138-2271, 2140- 2262, 214-2265, 2145-2265, 2148-2262, 2152-2262, 2166-2275, 2167-2248 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 41/7512297CB1/ 1-232, 1-486, 1-543, 1-594, 1-2521, 11-542, 13-213, 13-725, 20-701, 25-224, 30-682, 30-849, 31-337, 31-416, 31-499, 31-506, 31-531, 31- 2521 609, 31-688, 31-721, 32-316, 47-652, 48-555, 69-592, 70-616, 103-803, 157-400, 184-760, 201-645, 206-696, 207-453, 240-791, 245-872, 255-931, 263-970, 323-896, 429-1010, 433-606, 445-613, 463-1006, 510-1072, 511-880, 554-1047, 597-1050, 676-991, 680-993, 718-997, 775-1049, 881-1725, 1070-1689, 1076-1748, 1111-1391, 1150-1803, 1165-1642, 1181-1425, 1181-1429, 1181-1576, 1186-1760, 1236- 1883, 1239-1515, 1253-1518, 1270-1833, 1293-1930, 1313-1552, 1330-1971, 1336-1926, 1347-1933, 1358-1933, 1363-1614, 1371-1678, 1384-1678, 1403-1724, 1410-2118, 1446-1774, 1461-2124, 1468-2084, 1516-1789, 1523-2189, 1537-2100, 1551-2227, 1555-1653, 1569- 1782, 1578-2011, 1582-1994, 1606-1849, 1629-2227, 1688-2003, 1689-2046, 1690-1938, 1700-2169, 1703-2265, 1708-2208, 1716-2172, 1719-2171, 1739-2328, 1740-2075, 1770-2411, 1811-2274, 1830-2063, 1832-2439, 1873-2374, 1874-2469, 1943-2516, 1953-2521, 1996-2211, 1996-2242, 1996-2464, 2023-2518, 2025-2393, 2040-2469, 2042-2459, 2043-2514, 2054-2521, 2056-2496, 2057-2514, 2062- 2521, 2067-2512, 2084-2518, 2085-2521, 1086-2343, 2090-2351, 2090-2509, 2090-2521, 2095-2458, 2098-2521, 2100-2515, 2105-2481, 2109-2521, 2112-2514, 2115-2386, 2117-2519, 2120-2521, 2123-2446, 2123-2521, 2138-2509, 2138-2513, 2144-2509, 2160-2516, 2165- 2514, 2166-2509, 2183-2518, 2184-2514, 2193-2426, 2197-2488, 2209-2326, 2209-2424, 2209-2474, 2209-2512, 2212-2521, 2216-2521, 2222-2521, 2253-2460, 2263-2519, 2271-2509, 2361-2508, 2361-2519, 2444-2515 42/7512392CB1/ 1-5261, 418-964, 496-731, 823-1178, 904-1484, 904-1486, 908-1486, 909-1486, 953-1544, 1037-1634, 1076-1658, 1102-1419, 1166-1526, 5281 1185-1830, 1191-1452, 1223-1343, 1464-2215, 1826-2288, 1855-2246, 1859-2082, 1942-2468, 2032-2291, 2035-2182, 2035-2468, 2035- 2475, 2035-2505, 2035-2535, 2035-2550, 2035-2557, 2035-2583, 2035-2608, 2035-2665, 2035-2666, 2035-2673, 2115-2780, 2139-2670, 2169-2794, 2169-2814, 2289-2916, 2309-2673, 2311-2988, 2329-2881, 2332-2976, 2369-2988, 2382-2734, 2394-2998, 2419-3050, 2441- 2990, 2523-3345, 2526-3066, 2534-2974, 2572-3014, 2594-2794, 2594-2807, 2616-2995, 2629-3016, 2652-3014, 2661-3015, 2681-3011, 2710-3024, 2733-2999, 2741-3018, 2747-3635, 2750-3022, 2758-3002, 2815-2998, 2819-3011, 2858-3001, 2879-3430, 2897-3532, 2994- 3764, 2999-3761, 3079-3621, 3083-3856, 3142-3700, 3224-3843, 3236-3781, 3256-3850, 3286-3934, 3292-3941, 3293-4047, 3295-3921, 3367-4036, 33898-3950, 3425-3708, 3444-3932, 3444-3950, 3457-3827, 3457-3847, 3457-4053, 3501-4020, 3533-4170, 3538-3933, 3556- 4094, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 3569-4382, 3596-3745, 3603-4120, 3614-4348, 3640-4079, 3662-4401, 3689-3947, 3696-4111, 3730-3833, 3730-4042, 3730-4149, 3730- 4182, 3730-4189, 3730-4269, 3731-4421, 3752-4322, 3771-4059, 3776-4002, 3776-4083, 3776-4368, 3787-4113, 3816-4009, 3828-4463, 3856-4168, 3857-4045, 3862-4366, 3872-4075, 3874-4330, 3875-4425, 3875-4438, 3878-4130, 3908-4080, 3920-4343, 3920-4625, 3932- 4162, 3945-4528, 3972-4606, 3979-4285, 3980-4540, 3986-4308, 3998-4549, 4009-4278, 4021-4581, 4027-4153, 4032-4576, 4033-4629, 4038-4296, 4038-4441, 4045-4258, 4049-4573, 4051-4320, 4055-4298, 4098-4361, 4119-4397, 4144-4728, 4145-4349, 4170-4302, 4172- 4867, 4174-4526, 4179-4679, 4181-4574, 4183-4329, 4208-4757, 4237-4791, 4238-4855, 4243-4427, 4245-4522, 4246-4877, 4259-4542, 4261-4788, 4285-4833, 4288-4528, 4304-4521, 4304-4680, 4310-4410, 4310-4693, 4310-4721, 4326-4848, 4326-4876, 4327-4880, 4332- 4630, 4348-4907, 4365-4909, 4376-4780, 4377-4628, 4378-4608, 4378-4648, 4378-4662, 4384-4767, 4392-4696, 4399-4502, 4404-4645, 4406-4714, 4407-4876, 4411-4692, 4416-4654, 4422-4690, 4434-4727, 4436-4724, 4453-4662, 4462-4717, 4468-4658, 4484-5085, 4487-4848, 4487- 5044, 4509-5014, 4512-4763, 4519-4774, 4519-4798, 4520-4621, 4520-4923, 4523-4711, 4525-4930, 4541-4785, 4543-4775, 4546-4667, 4547-4800, 4548-4808, 4548-4809, 4549-4840, 4558-4819, 4569-4678, 4578-4863, 4580-4829, 4580-4842, 4583-4897, 4585-4856, 4592- 4833, 4594-4847, 4596-5077, 4608-4858, 4608-4875, 4608-4887, 4615-4923, 4624-4864, 4631-4881, 4640-5175, 4651-4856, 4651-4880, 4655-4934, 4656-4879, 4663-4696, 4664-5196, 4668-4841, 4671-4787, 4671-4873, 4690-4939, 4704-4922, 4704-4939, 4735-4939, 4746- 5070, 4803-4921, 4851-5271, 4904-4975, 4937-5038, 4937-5172, 4937-5230, 4937-5231, 4937-5246, 4940-5257, 4955-5196, 4955-5252, 4959-5174, 4959-5220, 4959-5222, 4972-5210, 4973-5206, 4986-5252, 4986-5253, 4988-5252, 5000-5257, 5027-5237, 5028-5252, 5035- 5246, 5044-5252, 5048-5235, 5052-5281, 5074-5182, 5110-5251 43/7511589cb1/ 1-639, 15-1452, 547-1378, 561-1378, 566-1378, 612-1378, 613-1378, 615-1378, 629-1378, 630-1378, 734-1378, 765-1378, 778-1378, 814- 1452 1378, 838-1378, 851-1193, 851-1208, 891-1121, 909-1452, 1059-1418 44/7511699CB1/ 1-141, 1-2640, 50-141, 53-141, 107-732, 142-277, 347-638, 347-679, 347-732, 347-744, 347-749, 347-802, 347-804, 347-816, 347-851, 2640 347-855, 350-749, 350-854, 449-749, 462-839, 464-769, 538-1121, 557-749, 561-816, 881-1069, 914-1375, 1423-2320, 1473-2013, 1514- 2362, 1514-2466, 1514-2477, 1519-2320, 1559-2194, 1604-2163, 1604-2273, 1604-2283, 1604-2286, 1604-2403, 1604-2481, 1604-2519, 1604-2640, 1608-2325, 1652-1926, 1679-2321, 1696-1966, 1718-2349, 1738-1925, 1740-2045, 1758-2038, 1776-2033, 1792-2205, 1856- 2385, 1888-2314, 1910-2200, 1930-2423, 1942-2020, 1964-2481, 2002-2267, 2002-2387, 2015-2483, 2029-2317, 2029-2449 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 45/7512595CB1/ 1-267, 1-289, 1-539, 1-678, 14-516, 14-517, 30-500, 37-540, 180-215, 286-522, 300-488, 531-837, 652-916, 734-1038, 734-1139, 1081- 2920 1360, 1081-1418, 1095-1287, 1302-1771, 1583-1847, 1650-1990, 1650-2145, 1771-2121, 1833-2451, 1892-2605, 1893-2532, 1956-2229, 1960-2181, 1960-2201, 1960-2337, 1960-2477, 1972-2671, 1978-2535, 1990-2624, 2003-2445, 2006-2445, 2026-2738, 2030-2595, 2041- 2660, 2049-2286, 2060-2737, 2073-2674, 2080-2589, 2118-2737, 2141-2527, 2169-2701, 2177-2790, 2187-2606, 2240-2920, 2252-2605, 2258-2867, 2308-2434, 2324-2920, 2343-2575, 2358-2847, 2395-2741, 2399-2822 46/2803911CB1/ 1-791, 319-972, 338-981, 545-1131, 590-1188, 653-1352, 680-1090, 693-1235, 693-1282, 706-850, 712-1359, 740-1544, 741-1544, 742- 6736 1544, 743-1544, 777-1241, 823-1544, 842-1508, 1043-1475, 1045-1476, 1108-1455, 1161-1437, 1173-1765, 1309-1815, 1331-4766, 1569- 6724, 1942-2581, 2131-2692, 2145-2807, 2154-2824, 2173-2873, 2462-3080, 2528-3064, 2946-3448, 3260-3885, 3566-4048, 3711-4312, 374-4346, 3757-4305, 3837-4551, 3918-4538, 3933-4442, 3951-4489, 3964-4484, 4026-4538, 4070-4538, 4108-4710, 4133-4731, 4133- 4788, 4301-4902, 4301-4934, 4301-5020, 4329-4989, 4339-4868, 4342-4765, 4368-5012, 4446-5240, 4495-5046, 4510-5244, 4510-5246, 4518-5214, 4529-5237, 4555-5052, 4574-5199, 4578-5263, 4593-4841, 4616-5302, 4629-5162, 4629-5418, 4643-5200, 4649-5302, 4676- 4901, 4691-5392, 4699-5319, 4723-5346, 4761-5346, 4782-5422, 4800-5410, 4805-5526, 4830-5459, 4844-5536, 4847-5519, 4857-5484, 4860-5479, 4860-5536, 4862-5328, 4863-5521, 4870-5570, 4871-5464, 4876-5503, 4880-5411, 4880-5550, 4897-5469, 4907-5499, 4907- 5597, 4936-5603, 4942-5794, 4946-5588, 4963-5488, 4968-5537, 4972-5471, 4992-5686, 4998-5745, 5005-5629, 5006-5802, 5013-5736, 5019- 5595, 5019-5672, 5023-5755, 5026-5617, 5030-5635, 5040-5654, 5042-5533, 5042-5559, 5042-5585, 5042-5608, 5042-5614, 5044-5588, 5045-5608, 5048-5536, 5056-5745, 5063-5616, 5078-5581, 5078-5658, 5101-5719, 5104-5648, 5107-5745, 5146-5851, 5171-5672, 5175- 5735, 5184-5831, 5199-5874, 5204-5700, 5220-5868, 5240-5929, 5287-5828, 5287-5829, 5344-5851, 5406-5677, 5406-5982, 5471-6255, 5525-6139, 5529-6150, 5572-6244, 5577-6456, 5586-6146, 5596-6323, 5598-6197, 5602-6399, 5606-6130, 5615-6291, 5635-6317, 5645- 6135, 5656-6148, 5675-6279, 5683-6225, 5684-5989, 5687-6345, 5711-6535, 5716-6455, 5718-6285, 5719-6210, 5742-6284, 5745-6291, 5754-6225, 5755-6337, 5756-6364, 5758-6387, 5772-6268, 5798-6466, 5800-6473, 5810-6466, 5815-6605, 5828-6466, 5831-6412, 5838- 6375, 5841-6455, 5844-6072, 5844-6139, 5846-6336, 5857-6594, 5861-6117, 5864-6602, 5867-5980, 5869-6647, 5875-6140, 5876-6431, 5883-6215, 5883-6290, 5883-6298, 5890-6466, 5894-6480, 5894-6599, 5895-6465, 5897-6386, 5904-6170, 5904-6187, 5906-6465, 5908-6422, 5908- 6599, 5908-6728, 5912-6483, 5914-6346, 5914-6460, 5915-6196, 5919-6465, 5930-6456, 5945-6466, 5958-6526, 5966-6434, 5980-6479, 5992-6469, 5993-6472, 6037-6717, 6043-6472, 6053-6582, 6067-6471, 6088-6476, 6174-6454, 6183-6714, 6217-6489, 6220-6476, 6221- 6529, 6496-6724, 6575-6736 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 47/7511806CB1/ 1-207, 14-209, 14-300, 14-2092, 21-222, 25-300, 27-300, 154-394, 298-556, 299-518, 299-554, 324-635, 324-790, 345-907, 352-491, 354- 2113 7114, 393-977, 394-706, 394-714, 395-902, 438-1042, 456-1114, 459-1095, 512-770, 512-1115, 519-1163, 532-1094, 538-637, 551-1136, 552-1135, 572-804, 572-1035, 572-1193, 588-887, 600-1099, 602-1009, 620-899, 623-885, 649-949, 671-912, 673-766, 673-767, 687-1231, 698-960, 702-1260, 709-942, 710-1247, 715-1116, 715-1247, 718-1371, 721-975, 726-1216, 726-1217, 738-931, 760-1321, 765-1045, 772- 1361, 782-935, 783-1048, 783-1067, 788-1074, 788-1209, 791-1299, 798-1084, 806-1060, 812-1112, 815-1367, 817-1077, 817-1281, 823- 1059, 824-1098, 825-1028, 837-1360, 849-1518, 867-1130, 869-1135, 872-1134, 872-1136, 873-1479, 894-1197, 901-1448, 906-1156, 906- 1166, 916-1231, 931-1128, 931-1157, 931-1170, 931-1173, 931-1174, 931-1183, 931-1206, 932-1184, 935-1631, 936-1361, 937-1134, 941- 1578, 9843-1496, 949-1547, 956-1478, 960-1194, 960-1339, 960-1417, 960-1506, 960-1512, 961-1507, 967-1247, 968-1253, 970-1507, 970-1726, 977-1519, 999-1272, 1002-1272, 1013-1553, 1019-1290, 1035-1614, 1036-1338, 1040-1642, 1045-1731, 1052-1610, 1057-1436, 1061-1741, 1066-1623, 1067-1265, 1068-1365, 1068-1396, 1070-1323, 1071-1315, 1082-1706, 1096-1417, 1096- 1543, 1097-1507, 1100-1603, 1100-1650, 1101-1353, 1104-1709, 1109-1366, 1126-1237, 1142-1754, 1144-1813, 1154-1728, 1158-1734, 1160-1778, 1162-1781, 1167-1687, 1173-1596, 1173-1658, 1174-1752, 1180-1476, 11898-1841, 1204-1464, 1204-1467, 1204-1767, 1209- 1819, 1216-1807, 1227-1905, 1230-1599, 1235-1431, 1237-1841, 1241-1474, 1242-1810, 1246-1427, 1255-1511, 1266-1855, 1266-1856, 1268-1532, 1268-1576, 1282-1554, 1283-1690, 1288-1791, 1292-1807, 1294-1789, 1301-1904, 1306-1610, 1306-1617, 1306-1917, 1306- 1961, 1312-1894, 1331-1791, 1348-1846, 1366-1880, 1367-1921, 1368-1639, 1368-1641, 1373-2078, 1380-1930, 1382-1682, 1395-1586, 1395-1636, 1395-1644, 1398-1607, 1398-1679, 1398-1696, 1411-1940, 1412-1682, 1412-1996, 1421-1721, 1429-1720, 1430-1941, 1433- 1797, 1444-1777, 1448-2032, 1449-2028, 1452-1782, 1458-2089, 1467-1874, 1469-1762, 1484-1803, 1487-1970, 1490-1711, 1494-2081, 1504- 2083, 1510-2025, 1514-2021, 1522-1842, 1525-2091, 1525-2092, 1527-2079, 1532-1794, 1537-2018, 1544-2010, 1546-2083, 1547-1811, 1547-2039, 1547-2083, 1553-2092, 1563-2071, 1563-2082, 1565-2087, 1567-1854, 1572-2093, 1577-2089, 1578-2083, 1587-1759, 1587- 2100, 1592-2090, 1594-2090, 1597-1941, 1598-1873, 1608-2083, 1619-2023, 1620-1860, 1620-2042, 1625-2078, 1625-2081, 1626-1848, 1626-1881, 1626-2055, 1629-2082, 1636-2011, 1636-2076, 1637-1912, 1637-2100, 1638-2083, 1642-2071, 1644-2087, 1645-1787, 1649- 2112, 1650-2015, 1651-2079, 1652-2081, 1653-2097, 1654-2081, 1660-2081, 1661-2078, 1661-2081, 1662-2081, 1663-1975, 1663-2076, 1663-2080, 1663-2082, 1665-2078, 1666-2078, 1666-2091, 1668-2083, 1670-2078, 1671-2080, 1673-2081, 1675-2078, 1677-2078, 1678- 2079, 1681-2079, 1682-2078, 1682-2081, 1685-2073, 1686-2078, 1687-2078, 1689-2013, 1689-2081, 1690-2034, 1690-2045, 1690-2078, 1694-2073, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 1698-2078, 1700-2010, 1700-2113, 1708-2078, 1711-2078, 1713-2043, 1715-2078, 1715-2086, 1718-2043, 1718-2078, 1725-2087, 1733- 2113, 1736-2080, 1748-2078, 1759-2081, 1764-2078, 1764-2082, 1765-2079, 1771-2078, 1774-2081, 1778-2102, 1781-2094, 1787-2095, 1802-2082, 1811-2000, 1811-2024, 1811-2025, 1811-2086, 1814-2028, 1831-2086, 1844-2081, 1845-2074, 1845-2081, 1851-2078, 1879- 2084, 1887-2033, 1903-2066, 1908-2069, 1910-2081, 1913-2024, 1936-2080, 1937-2080, 1942-2078, 1945-2081, 1952-2078, 1979-2095 48/7511853CB1 1-338, 1-345, 1-348, 1-351, 1-352, 1-353, 1-2799, 16-651, 16-702, 16-707, 16-809, 16-823, 16-877, 16-898, 16-940, 16-946, 19-566, 21- 2799 844, 99-353, 240-352, 625-1404, 711-1022, 995-1908, 1422-1615, 1499-2074, 1622-2357, 1635-2616, 1708-2616, 1916-2597, 1916-2695, 2020-2695, 2089-2686, 2244-2778, 2342-2790, 2416-2796, 2528-2791, 2538-2799 49/7512306CB1/399 1-175, 4-388, 4-399, 44-154, 58-285, 283-399, 285-383 50/7512607CB1/ 1-163, 1-205, 1-211, 1-228, 1-230, 1-233, 1-253, 1-279, 1-322, 1-368, 1-379, 1-424, 1-485, 1-507, 1-528, 1-553, 1-623, 1-629, 1-632, 1-751, 1316 1-1298, 4-276, 4-516, 5-270, 5-281, 10-475, 10-639, 18-263, 26-280, 27-453, 27-859, 28-256, 28-467, 31-303, 32-282, 32-285, 32-298, 32- 713, 35-298, 36-338, 37-663, 37-773, 38-603, 41-303, 44-141, 44-288, 45-300, 45-733, 46-581, 47-309, 47-320, 50-174, 58-589, 77-577, 89- 347, 90-312, 90-394, 90-649, 97-777, 117-410, 131-403, 132-411, 137-726, 156-711, 164-445, 178-444, 220-522, 225-858, 231-414, 231- 471, 233-863, 238-847, 264-517, 281-557, 283-561, 311-598, 332-620, 346-883, 347-851, 347-853, 372-610, 394-661, 420-859, 434-610, 449-733, 463-616, 481-748, 487-744, 493-738, 507-790, 533-810, 587-757, 598-859, 601-800, 601-847, 627-859, 632-827, 857-1137, 857- 1151, 864-1141, 869-1145, 887-1133, 890-1146, 892-1159, 894-1181, 931-1181, 941-1157, 988-1316, 101-1182, 1036-1282, 1056-1298 51/7511851CB1/ 1-338, 1-345, 1-348, 1-351, 1-352, 1-353, 1-2967, 16-651, 16-702, 16-707, 16-809, 16-823, 16-877, 16-898, 16-940, 16-946, 19-566, 21- 2967 844, 99-353, 240-352, 625-1404, 711-1022, 995-1908, 1422-1615, 1499-2067, 1513-2161, 1514-2273, 1917-2861, 2584-2964, 2696-2959, 2706-2967 52/7512569CB1/ 1-305, 1-389, 25-206, 25-299, 25-334, 25-522, 25-615, 26-612, 26-676, 28-279, 29-213, 38-323, 39-281, 39-293, 39-316, 41-259, 44-230, 1400 44-343, 46-426, 48-604, 49-323, 49-330, 49-428, 49-672, 52-512, 53-374, 54-339, 55-627, 56-347, 56-674, 56-677, 57-577, 57-580, 64-297, 64-591, 74-338, 74-394, 80-328, 80-642, 83-274, 89-624, 99-340, 137-367, 138-401, 139-413, 183-414, 183-505, 184-440, 203-441, 203- 612, 203-668, 250-510, 251-399, 262-678, 285-526, 306-427, 323-634, 325-584, 341-562, 369-523, 374-636, 374-637, 387-678, 505-678, 508-744, 679-824, 679-893, 679-900, 679-1113, 696-1291, 720-913, 723-1206, 728-1093, 731-1019, 739-1212, 746-1221, 749-1029, 753- 1010, 757-1118, 758-1156, 758-1157, 761-1013, 785-893, 808-1051, 815-1185, 833-1108, 835-1107, 854-1121, 854-1126, 891-1393, 895- 1400, 899-1383, 912-1172, 919-1171, 928-1400, 932-1179, 937-1164, 937-1186, 938-1221, 940-1202, 963-1198, 964-1077, 965-1289, 966- 1393, 977-1180, 982-1400, 983-1378, 991-1378, 996-1221, 1040-1400, 1063-1330, 1087-1379, 1101-1386, Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 1107-1378, 1109-1389, 1109-1392, 1113-1144, 1147-1385, 1246-1378, 1252-1378, 1299-1378 53/7512570CB1/831 1-305, 1-784, 25-206, 25-299, 25-334, 25-489, 25-513, 28-279, 29-213, 38-323, 39-281, 39-293, 39-316, 41-259, 44-230, 44-343, 46-426, 49-323, 49-330, 49-428, 52-616, 53-374, 54-339, 56-347, 64-297, 74-338, 74-394, 80-328, 80-504, 83-274, 99-340, 137-367, 138-401, 139- 413, 183-414, 183-505, 184-440, 203-441, 250-514, 251-399, 285-512, 306-427, 369-512, 508-773, 508-774, 508-781, 508-784, 508-787, 508-824, 508-831, 542-780, 641-773, 647-773, 694-773 54/7512846CB1/ 1-238, 1-248, 4-518, 7-2053, 13-491, 14-521, 21-255, 33-255, 37-727, 47-387, 48-302, 48-488, 48-492, 48-497, 48-503, 48-543, 48-551, 48- 2053 640, 49-531, 53-686, 58-296, 58-297, 59-363, 65-335, 70-289, 77-591, 81-320, 135-416, 140-663, 179-483, 210-565, 277-505, 278-546, 280-541, 305-547, 341-737, 348-981, 352-662, 353-625, 360-829, 371-577, 371-617, 371-698, 373-737, 388-732, 388-823, 390-683, 419- 881, 428-688, 466-728, 485-762, 498-793, 523-887, 542-1081, 571-725, 590-830, 604-1120, 622-841, 624-862, 643-868, 643-888, 645-918, 645-1027, 647-911, 648-1087, 657-934, 657-1189, 691-961, 741-978, 757-986, 758-1021, 766-1044, 780-1052, 795-1201, 808-1081, 828- 1078, 835-1098, 841-1083, 859-1070, 859-1158, 883-1163, 884-1187, 905-1201, 912-1156, 922-1122, 930-1168, 9835-1193, 944-1201, 952- 1162, 983-1201, 986-1177, 991-1189, 1010-1201, 1059-1306, 1202-143,2 1202-1435, 1203-1450, 1204-1482, 1204-1726, 1212-1429, 1213- 1545, 1213-1834, 1240-1529, 1243-1656, 1251-1518, 1257-1878, 1259-1475, 1259-1534, 1262-1533, 1264-1498, 1264-1524, 1266-1561, 1266-1792, 1275-1543, 1277-1540, 1277-1815, 1281-1916, 1288-1529, 1288-1580, 1295-1538, 1302-1777, 1312- 1522, 1312-1547, 1313-1561, 1314-1827, 1316-1567, 1317-1509, 1317-1510, 1317-1518, 1317-1533, 1320-1534, 1320-1542, 1323-1590, 1328-1575, 1328-1610, 1331-1490, 1331-1605, 1335-1641, 1335-1688, 1336-1595, 1341-1632, 1341-1636, 1342-1688, 1344-1884, 1352- 1620, 1361-1517, 1370-1620, 1385-1937, 1397-1608, 1427-1667, 1428-1733, 1429-2046, 1430-1525, 1455-1987, 1456-1729, 1456-1935, 1460-2053, 1462-1720, 1477-1723, 1478-2010, 1479-2002, 1481-1830, 1505-1773, 1517-1988, 1518-1779, 1521-1884, 1533-1716, 1542- 1872, 1546-2013, 1546-2053, 1558-1886, 1564-2053, 1568-2049, 1569-1821, 1572-1983, 1576-2049, 1581-2006, 1581-2049, 1583-2045, 1583-2050, 1584-2053, 1588-2049, 1589-1844, 1592-2048, 1593-2048, 1594-2040, 1594-2048, 1596-1902, 1596-2046, 1597-2050, 1598- 2047, 1601-2053, 1603-2047, 1603-2053, 1604-2007, 1604-2053, 1606-1770, 1611-2043, 1612-2048, 1616-2013, 1616-2046, 1616-2053, 1617-2053, 1618-2046, 1625-1651, 1625-2046, 1625-2053, 1626-1890, 1627-2053, 1631-2049, 1632-2053, 1635-2047, 1636-2046, 1639-2053, 1640- 2046, 1640-2053, 1642-1986, 1645-2046, 1647-2046, 1648-2047, 1648-2053, 1649-1930, 1649-1931, 1653-2053, 1654-1984, 1654-2048, 1657-2053, 1664-1925, 1675-2053, 1683-2043, 1687-1986, 1691-1903, 1701-2046, 1703-2047, 1704-2043, 1704-2046, 1712-2046, 1713- 2046, 1718-2046, 1737-2050, 1749-2046, 1753-2047, 1754-1961, 1755-2032, 1756-2047, 1757-2041, 1760-2005, 1760-2013, 1760-2045, 1771-2021, 1771-2030, 1775-2049, 1805-2039, 1815-2046, 1823-2046, 1825-1993, 1827-2038, 1827-2049, 1827-2052, 1840-2046, 1861- 2048, 1865-2046, 1880-2046, 1949-2046 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 55/7517814CB1/ 1-717, 1-824, 1-904, 2-1665, 151-1094, 200-1093, 570-1461, 571-1495, 729-1666, 794-1666, 949-1666 1666 56/7519828CB1/ 1-209, 1-467, 1-649, 1-69, 1-695, 1-735, 1-736, 1-768, 2-1346, 3-649, 440-1347, 605-1351, 613-1344, 688-1347, 781-1342, 970-1347 1351 57/7519833CB1/ 1-209, 1-597, 2-1488, 23-868, 118-868, 499-1362, 500-1251, 714-1489, 755-1487, 792-1489, 863-1489, 883-1489, 884-1489, 923-1484, 1489 1112-1489 58/7519834CB1/ 1-209, 1-466, 1-561, 1-728, 2-1292, 460-1293, 461-1219, 559-1291, 596-1293, 667-1293, 687-1293, 688-1293, 727-1288, 916-1293 1293 59/7519939CB1/892 1-717, 173-892, 204-889 60/7520133CB1/ 1-751, 1-799, 1-860, 2-1157, 422-1158 1158 61/7520171CB1/872 1-741, 1-749, 2-811, 2-871, 3-872, 4-872, 270-872, 576-872 62/7522453CB1/ 1-3083, 107-574, 199-985, 213-586, 220-586, 222-595, 224-402, 224-438, 224-509, 224-554, 224-560, 224-562, 224-571, 224-572, 224- 3186 574, 224-586, 224-589, 224-595, 225-563, 240-554, 240-586, 339-968, 453-915, 526-640, 571-1129, 595-1232, 666-1294, 670-8798, 673- 1275, 709-991, 709-1187, 722-1223, 735-1316, 735-1332, 755-1389, 802-1437, 811-1397, 812-1434, 832-1452, 875-1461, 910-1412, 928- 1497, 937-1412, 955-1547, 1116-1770, 1129-1746, 1148-1398, 1713-2316, 1739-2316, 1838-2128, 1857-2608, 1900-2139, 1900-2178, 1902-2378, 1907-2151, 1914-2041, 1914-2697, 1941-2214, 1950-2238, 1957-2199, 1991-2459, 1991-2563, 1991-2597, 1991-2610, 1992- 2693, 1996-2564, 2004-2463, 2008-2463, 2013-2591, 2021-2588, 2021-2605, 2046-2395, 2061-2617, 2061-2668, 2067-2677, 2081-2342, 2083-2783, 2091-2608, 2092-2593, 2093-2593, 2108-2678, 2128-2754, 2132-2398, 2139-2289, 2140-2732, 2149-2393, 2190-2380, 2190- 2439, 2203-2410, 2206-2426, 2206-2676, 2230-2565, 2233-2492, 2241-2442, 2241-2452, 2247-2380, 2261-2835, 2262-2589, 2277-2614, 2284-2840, 2311-2549, 2312-2542, 2348-3076, 2351-2604, 2359-3073, 2361-2896, 2366-2829, 2376-2992, 2376-3049, 2395-3019, 2395- 3097, 2401-2641, 2423-2659, 2424-2700, 2428-2896, 2437-2931, 2443-3102, 2453-2759, 2454-3078, 2464-3008, 2469-3090, 2475-3019, 2483-2893, 2485-2606, 2489-2789, 2516-3186, 2518-2784, 2520-3041 Table 4 Polynucleotide Sequence Fragments SEQ ID NO:/ Incyte ID/ Sequence Length 63/7522454CB1/ 1-429, 1-2846, 223-270, 224-554, 224-571, 224-572, 224-574, 225-563, 540-807, 595-1232, 735-1316, 755-1389, 802-1437, 832-1452, 910- 2864 1412, 931-1208, 937-1412, 995-1261, 1006-1582, 1122-1791, 1131-1749, 1148-1349, 1148-1398, 1199-1808, 1228-1791, 1241-1827, 1428- 1515, 1296-1749, 1298-1895, 1324-1576, 1345-1611, 1352-1945, 1355-1931, 1374-1931, 1382-1930, 1418-1922, 1424-1855, 1431-1752, 1431-2047, 1433-1682, 1437-1636, 1461-1827, 1507-1672, 1522-1903, 1550-2181, 1563-2103, 1620-2094, 1632-2168, 1644-1873, 1644- 2115, 1665-2193, 1681-2188, 1687-2347, 1720-1962, 1776-2176, 1846-2546, 1895-2161, 1912-2156, 1969-2439, 1994-2160, 2004-2205, 2004-2215, 2024-2598, 2047-2603, 2074-2312, 2075-2305, 2124-2659, 2129-2592, 2139-2755, 2191-2659, 2216-2522, 2232-2854, 2232- 2857, 2281-2547, 2339-2861, 2346-2783, 2375-2782, 2387-2841, 2393-2842, 2393-2850, 2413-2861, 2469-2848, 2487-2740, 2497-2839, 2499-2666, 2506-2846, 2566-2864, 2590-2792, 2590-2833, 2590-2864 64/7517956CB1/ 1-669, 1-789, 1-1338, 588-1333, 644-1338 1338 65/7517957CB1/978 1-948, 1-978, 181-978, 228-973 66/7519293CB1/ 1-519, 1-744, 2-1003, 235-1004, 526-1014, 563-1014 1014 Table 5 Polynucleotide SEQ Icnyte Project ID: Representative Library ID NO: 34 7511094CB1 LUNGNOT18 35 7511619CB1 NGANNOT01 36 7511720CB1 COLCTUT03 37 7511661CB1 FIBPFEA01 38 7512204CB1 LUNGNOT38 39 7512197CB1 FIBRTXS07 40 7511656CB1 PANCTUT02 41 7512297CB1 TMLR3DT02 42 7512392CB1 SINTFEE02 43 7511589CB1 SCOMDIC01 44 7511699CB1 CARGDIT01 45 7512595CB1 OVARDIN02 46 2803911CB1 BRSTTMC01 47 7511806CB1 FIBPFEN06 48 7511853CB1 TESTNOC01 49 7512306CB1 BONRTUT01 50 7512607CB1 HNT2RAT01 51 7511851CB1 TESTNOC01 52 7512569CB1 SININOT04 53 7512570CB1 BRAINOT09 54 7512846CB1 LUNGNOT02 62 7522453CB1 BRAINOT22 63 7522454CB1 BRAITUT08 Table 6 Library Vector Library Description BONRTUT01 pINCY Library was constructed using RNA isolated from rib tumor tissue removed from a 16-year-old Caucasian male during a rib osteotomy and a wedge resection of the lung. pathology indicated a metastatic grade 3 (of 4) osteosarcoma, forming a mass involving the chest wall. BRAINOT09 pINCY Library was constructed using RNA isolated from brain tissue removed from a Caucasian male fetus, who died at 23 weeks' gestation. BRAINOT22 pINCY Library was constructed using RNA isolated from right temporal lobe tissue removed from a 45-year-old Black male during a brain lobectomy. Pathology for the associated tumor tissue indicated dysembryoplastic neuroepithelial tumor of the right temporal lobe. The right temporal region dura was consistent with calcifying pseudotumor of the neuraxis. Family history included obesity, benign hypertension, cirrhosis of the liver, obesity, hyperlipidemia, cerebrovascular disease, and type II diabetes. BRAITUT08 pINCY Library was constructed using RNA isolated from brain tumor tissue removed from the left frontal lobe of a 47-year-old Caucasian male during excision of cerebral meningeal tissue. pathology indicated grade 4 fibrillary astrocytoma with focal tumoral radionecrosis. patient history included cerebrovascular disease, deficiency anemia, hyperlipidemia, epilepsy, and tobacco use. Family history included cerebrovascular disease and a malignant prostate neoplasm. BRSTTMC01 pINCY This large size-fractionated library was constructed using pooled cDNA from four donors. cDNA was generated using mRNA isolated from diseased breast tissue removed from a 40-year-old Caucasian female (donor A) during a bilateral reduction mammoplasty; from breast tissue removed from a 46-year-old Caucasian female (donor B) during unilateral extended simple mastectomy with breast reconstruction; from breast tissue removed from a 56-year-old Caucasian female (donor C) during unilateral extended simple mastectomy with open breast biopsy; and frm breast tissue removed from a 57-year-old Caucasian female (donor D) during a unilateral extended simple mastectomy. Pathology indicated bilateral mild fibrocystic and proliferative changes (A); deep fascia was negative for tumor (B); non-proliferative fibrocystic change (C); and benign fat replaced breast parenchyma (D). Pathology for the matched tumor tissue (B) indicated invasive grade 3 adenocarcinoma, ductal type, with apocrine features. pathology for the matched tumor tissue (C) indicated invasive grade 3 ductal adenocarcinoma. Pathology for the matched tumor tissue (D) indicated residual microscopic infiltrating grade 3 ductal adenocarcinoma and extensive grade 2 intraductal carcinoma. patient history included breast hypertrophy and pure hypercholesterolemia (A); breast cancer (B); chronic airway obstruction and emphysema (C); and benign hypertension, hyperlipidemia, cardiac dysrhythmia, a benign colon neoplasm, a solitary breast cyst, and a breast neoplasm of uncertain behavior (D). Previous surgeries included open breast biopsy (B). Donor B's medications included Cytoxan and Adriamycin.

Table 6 Library Vector Library Description CARGDIT01 pINCY Library was constructed using RNA isolated from diseased cartilage tissue. Patient history included osteoarthritis. COLCTUT03 pINCY Library was constructed using RNA isolated from cecal tumor tissue removed from a 70-year-old Caucasian female during right hemicolectomy, open liver biopsy, flexible sigmoidoscopy, colonoscopy, and permanent colostomy. Pathology indicated invasive grade 2 adenocarcinoma forming an ulcerated mass 2 cm distal to the ileocecal valve and invading the muscularis propria. One regional lymph node (of 16) was positive for metastatic adenocarcinoma. Patient history included a deficiency anemia, malignant breast neoplasm, type II diabetes, hyperlipidemia, viral hepatitis, an unspecified thyroid disorder, osteoarthritis, a malignant skin neoplasm, and normal delivery. Family history included cardiovascular and cerebrovascular disease, hyperlipidemia, and breast and ovarian cancer. FIBPFEA01 PSPORT1 This amplified library was constructed using RNA isolated from untreated fibroblasts of the prostate stroma removed from a male fetus (Clonetics, Sample #CC-2508) who died after 26 weeks'gestation. FIBPFEN06 pINCY The normalized prostate stromal fibroblast tissue libraries were constructed from 1. 56 million independent clones from a prostate fibroblast library. Starting RNA was made from fibroblasts of prostate stroma removed from a male fetus, who died after 26 weeks'gestation. The libraries were normalized in two rounds using conditions adapted from Soares et al., PNAS (1994) 91 : 9228 and Bonaldo et al., Genome Research (1996) 6 : 791, except that a significantly longer (48- hours/round) reannealing hybridization was used. The library was then linearized and recircularized to select for insert containing clones as follows : plasmid DNA was prepped from approximately 1 million clones from the normalized prostate stromal fibroblast tissue libraries following soft agar transformation. FIBRTXS07 pINCY This subtracted library was constructed using 1. 3 million clones from a dermal fibroblast library and was subjected to two rounds of subtraction hybridization with 2. 8 million clones from an untreated dermal fibroblast tissue library. The starting library for subtraction was constructed using RNA isolated from treated dermal fibroblast tissue removed from the breast of a 31-year-old Caucasian female. The cells were treated with 9CIS retinoic acid. The hybridization probe for subtraction was derived from a similarly constructed library from RNA isolated from untreated dermal fibroblast tissue from the same donor. Subtractive hybridization conditions were based on the methodologies of Swaroop et al., NAR (1991) 19 : 1954 and Bonaldo, et al., Genome Research (1996) 6 : 791. HNT2RAT01 PBLUESCRIPT Library was constructed at Stratagene (STR937231), using RNA isolated from the hNT2 cell line (derived from a human teratocarcinoma that exhibited properties characteristic of a committed neuronal precursor). Cells were treated with retinoic acid for 24 hours.

Table 6 Library Vector Library Description LUNGNOT02 PBLUESCRIPT Library was constructed using RNA isolated from the lung tissue of a 47-year-old Caucasian male, who died of a subarachnoid bemorrhage. LUNGNOT18 pINCY Library was constructed using RNA isolated from left upper lobe lung tissue removed from a 66-year-old Caucasian female. Pathology for the associated tumor tissue indicated a grade 2 adenocarcinoma. patient history included cerebrovascular disease, atherosclerotic coronary artery disease, and pulmonary insufficiency. Family history included a myocardial infarction and atherosclerotic coronary artery disease. LUNGNOT38 pINCY Library was constructed using RNA isolated from diseased lung tissue removed from a 15-year-old Caucasian male who died from a gunshot wound to the head. Serology was positive for cytomegalovirus. patient history included asthma. NGANNOT01 PSPORT1 Library was constructed using RNA isolated from tumorous neuroganglion tissue removed from a 9-year-old Caucasian male during a soft tissue excision of the chest wall. Pathology indicated a ganglioneuroma. Family history included asthma. OVARDIN02 pINCY This normalized ovarian tissue library was constructed from 5.76 million independent clones from an ovary library. Starting RNA was made from diseased ovarian tissue removed from a 39-year-old Caucasian female during total abdominal hysterectomy, bilateral salpingo-oophorectomy, dilation andcurettage, partial colectomy, incidental appendectomy, and temporary colostomy. Pathology indicated the right and left adnexa, mesentery and muscularis propria of the sigmoid colon were extensively involved by endometriosis. Endometriosis also involved the anterior and posterior serosal surfaces of the uterus and the cul-de-sac. The endometrium was proliferative. pathology for the associated tumor tissue indicated multiple (3 intramural, 1 subserosal) leiomyomata. The patient presented with abdominal pain and infertility. Patient history included scoliosis. Family history included hyperlipidemia, benign hypertension, atherosclerotic coronary artery disease, depressive disorder, brain cancer, and type II diabetes. The library was normalized in two rounds using conditions adapted from Soares et al., PNAS(1994) 91:9228 and Bonaldo et al., Genome Research 6 (1996):791, except that a significantly longer (48-hours/round) reannealing hybridization was used. PANCTUT02 pINCY Library was constructed using RNA isolated from pancreatic tumor tissue removed from a 45-year-old Caucasian female during radical pancreaticoduodenectomy. Pathology indicated a grade 4 anaplastic carcinoma. Family history included benign hypertension, hyperlipidemia and atherosclerotic coronary artery disease.

Table 6 Library Vector Library Description SCOMDIC01 PSPORT1 This large size-fractionated library was constructed using RNA isolated from diseased spinal cord tissue removed from the base of the medulla of a 57-year-old Caucasian male, who died from a cerebrovascular accident. Serologies were negative. Patient history included Huntington's disease, emphysema, and tobacco abuse (3-4 packs per day, for 40 years). SININOT04 pINCY Library was constructed using RNA isolated from diseased ileum tissue obtained from a 26-year-old Caucasian male during a partial colectomy, permanent colostomy, and an incidental appendectomy. Pathology indicated moderately to severely active Crohn's disease. Family history included enteritis of the small intestine. SINTFEE02 PCDNA2.1 This 5' biased random primed library was constructed using RNa isolated from small intestine tissue removed from a Caucasian male fetus who died from patau's syndrome (trisomy 13) at 20-weeks' gestation. Serology was negative. TESTNOC01 PBLUESCRIPT This large size fractionated library was constructed using RNA isolated from testicular tissue removed from a pool of eleven, 10 to 61-year-old Caucasian males. TMLR3DT02 PBLUESCRIPT Library was constructed using RNA isolated from non-adherent peripheral blood mononuclear cells, which came from a pool of amle and female donors. The cells were cultured for 72 hours following Ficoll Hypaque centrifugation.

Table 7 Program Description Reference Parameter Threshold ABIFACTURA A program that removes vector sequences and masks Applied Biosystems, Foster City, CA. ambiguous bases in nucleic acid sequences. ABI/PARACEL FDF A Fast Data Finder useful in comparing and Applied Biosystems, Foster City, CA; Mismatch <50% annotating amino acid or nucleic acid sequences. Paracel Inc., Pasadena, CA. ABI AutoAssembler A program that assembles nculeic acid sequences. Applied Biosystems, Foster City, CA. BLAST A Basic Local Alignment Search Tool useful in Altschul, S.F. et al. (1990) J. Mol. Biol. ESTs: probability value = 1.0E- sequence similarity search for amino acid and nucleic 215:403-410; Altschul, S.F. et al. (1997) 8 or less; Full Length sequences: acid sequences. BLAST includes five functions: Nucleic Acids Res. 25:3389-3402. Probability value = 1.0E-10 or blastp, blastn, blastx, tblastn, and tblastx. less FASTA A Pearson and Lipman algorithm that searches for Pearson, W.R. and D.J. Lipman (1988) Proc. ESTs: fasta E value = 1.06E-6; similarity between a query sequence and a group of Natl. Acad Sci. USA 85;2444-2448; Pearson, Assembled ESTs: fasta Identity sequences of the same type. FASTA comprises as W.R. (1990) Methods Enzymol. 183:63-98; = 95% or greater and Match least five functions: fasta, tfasta, fastx, tfastx, and and Smith, T.F. and M.S. Waterman (1981) length = 200 bases or greater; ssearch. Adv. Appl. Math. 2:482-489. fastx E value = 1.0E-8 or less; Full Length sequences: fastx score = 100 or greater BLIMPS A BLocks IMProved Searcher that matches a Henikoff, S. and J.G. Henikoff (1991) Probability value = 1.0E-3 or sequence against those in BLOCKS, PRINTS, Nucleic Acids Res. 19:6565-6572; Henikoff, less DOMO, PRODOM, anD PFAM databases to search J.G. and S. henikoff (1996) methods for gene families, sequence homology, and structural Enzymol. 266:88-105; and Attwood, T.K. et fingerprint regions. al. (1997) J. Chem. Inf. Comput. Sci. 37:417- 424.

Table 7 program Description Reference Parameter Threshold HMMER An algorithm for searching a query sequence against Krogh, A. et al. (1994) J. Mol. Biol. PFAM, INCY, SMART or hidden Markov model (HMM)-based databases of 235:1501-1531; Sonnhammer, E.L.L. et al. TIGRFAM hits: Probability protein family consensus sequences, such as PFAM, (1988) Nucleic Acids Res. 26;320-322; value = 1.0E-3 or less; Signal INCY, SMART and TIGRFAM. Durbin, R. et al. (1998) Our World View, in peptide hits: Score = 0 or greater a Nutshell, Cambridge Univ. Press, pp. 1- 350. ProfileScan An algorithm that searches for structural and Gribskov, M. et al. (1988) CABIOS 4:61-66; Normalized quality score # GCG sequence motifs in protein sequences that match Gribskov, M. et al. (1989) Methods specified "HIGH" value for that sequence patterns defined in prosite. Enzymol. 183:146-159; Bairoch, A. et al. particular Prosite motif. (1997) Nucleic Acids Res. 25:217-221. Generally, score = 1.4-2.1. Phred A base-calling algorithm that examines automated Ewing, B. et al. (1998) Genome Res. 8:175- sequencer traces with high sensitivity and probability. 185; Ewing, B. and P. Green (1998) Genome Res. 8:186-194. Phrap A Phils Revised Assembly Program including Smith, T.F. and M.S. Waterman (1981) Adv. Score = 120 or greater; Match SWAT and CrossMatch, programs based on efficient Appl. Math. 2:482-489; Smith, T.F. and length = 56 or greater implementation fo the Smith-Waterman algorithm, M.S. Waterman (1981) J. Mol. Biol. 147:195- useful in searching sequence homology and 197; and Green, P., University of assembling DNA sequences. Washington, Seattle, WA. Consed A graphical tool for viewing and editing Phrap Gordon, D. et al. (1998) Genome Res. 8:195- assemblies. 202. SPScan A weight matrix analysis program that scans protein Nielson, H. et al. (1997) Protein Engineering Score = 3.5 or greater sequences for the presence of secretory signal 10:1-6; Claverie, J.M. and S. Audic (1997) peptides. CABIOS 12:431-439. TMAP A program that uses weight matrices to delineate persson, B. and P. Argos (1994) J. Mol. Biol. transmembrane segments on protein sequences and 237:182-192; persson, B. and P. Argos determine orientation. (1996) Protein Sci. 5:363-371.

Table 7 Program Description Reference Parameter Threshold TMHMMER A program that uses a hidden Markov model (HMM) Sonnhammer, E.L. et al. (1998) Proc. Sixth to delineate transmembrane segments on protein Intl. Conf. On Intelligent Systems for Mol. sequences and determine orientation. Biol., Glasgow et al., eds., The am. Assoc. for Artificial Intelligence (AAAI) Press, Menlo Park, CA, and MIT Press, Cambridge, MA, pp. 175-182. Motifs a program that searches amino acid sequences for Bairoch, A. et al. (1997) Nucleic Acids Res. patterns that matched those defined in Prosite. 25:217-221; Wisconsin Package Program Manual, version 9, page M51-59, Genetics Computer Group, Madison, WI.

Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 34 7511094 2912073H1 SNP00152978 74 952 T T C noncoding n/a n/a n/a n/a 34 7511094 2912073H1 SNP00152979 115 993 T T C noncoding n/a n/a n/a n/a 34 7511094 4327415H1 SNP00035034 110 511 G G A K93 n/a n/a n/a n/a 35 7511619 108766R6 SNP00074977 137 201 G G C L22 n/a n/a n/a n/a 35 7511619 1234107F6 SNP00074976 52 53 T T G noncoding n/a n/a n/a n/a 35 7511619 1234107F6 SNP00074977 201 202 G G C G23 n/a n/a n/a n/a 35 7511619 1234107H1 SNP00074976 50 52 T T G noncoding n/a n/a n/a n/a 35 7511619 1286212T6 SNP00024376 294 785 T T C noncoding 0.70 n/a n/a n/a 35 7511619 2452652T6 SNP00024376 284 781 C T C noncoding 0.70 n/a n/a n/a 35 7511619 4111882H1 SNP00024376 88 780 T T C noncoding 0.70 n/a n/a n/a 36 7511720 3180653H1 SNP00139165 46 139 C C T L16 n/a n/a n/a n/a 37 7511661 1351208H1 SNP00030003 69 430 T T C I117 n/a n/a n/a n/a 37 7511661 1439303H1 SNP00059786 228 1406 T T G noncoding n/a n/a n/a n/a 37 7511661 1494505H1 SNP00135792 19 1517 T T G noncoding n/a n/a n/a n/a 37 7511661 2539359H1 SNP00049925 242 1386 C C T noncoding n/a n/a n/a n/a 37 7511661 2539359H1 SNP00092786 97 1241 C C T F387 n/a n/a n/a n/a 37 7511661 2721572H1 SNP00058511 134 392 G G A V104 n/d n/a n/a n/a 37 7511661 5135947H1 SNP00073232 52 286 G G A R69 n/a n/a n/a n/a 37 7511661 6282640H1 SNP00148027 261 484 A A G Q135 n/a n/a n/a n/a 37 7511661 7201938H1 SNP00135792 407 1503 T T G noncoding n/d n/a n/a n/a 37 7511661 7664748H1 SNP00030003 422 431 T T C I117 n/a n/a n/a n/a 37 7511661 7664748J1 SNP00030003 334 422 T T C I114 n/a n/a n/a n/a 39 7512197 1472015H1 SNP00016121 91 717 A A G noncoding 0.75 0.81 0.82 0.72 39 7512197 1554824H1 SNP00016118 10 58 G G A noncoding n/a n/a n/a n/a 39 7512197 1959033H1 SNP00060617 130 381 G G A R42 n/a n/a n/a n/a 39 7512197 2074042H1 SNP00016119 153 191 G G C noncoding n/a n/a n/a n/a 39 7512197 2907084T6 SNP00016121 456 734 A A G noncoding 0.75 0.81 0.82 0.72 39 7512197 3033753T6 SNP00016121 426 716 A A G noncoding 0.75 0.81 0.82 0.72 Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 39 7512197 7642449J1 SNP00016119 143 172 G G C noncoding n/a n/a n/a n/a 39 7512197 7642449J1 SNP00060617 333 362 G G A G36 n/a n/a n/a n/a 40 7511656 1456204H1 SNP00004812 240 1689 G G A noncoding n/a n/a n/a n/a 40 7511656 1610835F6 SNP00116398 27 1031 A A G noncoding 0.93 n/d n/d 0.99 40 7511656 1734885T6 SNP00004813 337 1868 G G A noncoding n/a n/a n/a n/a 40 7511656 1734885T6 SNP00106990 76 2129 C C A noncoding n/a n/a n/a n/a 40 7511656 1975423T6 SNP00106990 20 2184 C C A noncoding n/a n/a n/a n/a 40 7511656 2184164T6 SNP00004813 342 1865 G G A noncoding n/a n/a n/a n/a 40 7511656 2184164T6 SNP00106990 81 2126 C C A noncoding n/a n/a n/a n/a 40 7511656 2305376H1 SNP00106990 81 2101 C C A noncoding n/a n/a n/a n/a 40 7511656 2361952H1 SNP00076320 169 914 A A G noncoding n/d n/a n/a n/a 40 7511656 2361952H1 SNP00134154 159 904 T T C noncoding n/a n/a n/a n/a 40 7511656 2453447T6 SNP00004813 273 1924 G G A noncoding n/a n/a n/a n/a 40 7511656 2453447T6 SNP00106990 12 2185 C C A noncoding n/a n/a n/a n/a 40 7511656 3434808T6 SNP00004813 364 1840 G G A noncoding n/a n/a n/a n/a 40 7511656 3443062T6 SNP00004813 279 1923 A G A noncoding n/a n/a n/a n/a 40 7511656 7695212J1 SNP00076320 367 884 A A G noncoding n/d n/a n/a n/a 40 7511656 7695212J1 SNP00134154 357 874 T T C noncoding n/a n/a n/a n/a 41 7512297 2124016F6 SNP00019942 128 1511 A A G R495 0.26 n/a n/a n/a 41 7512297 270876R7 SNP00060071 60 68 G G A A14 0.47 n/a n/a n/a 41 7512297 270876R7 SNP00098937 250 258 C C T P78 0.99 n/a n/a n/a 41 7512297 270876R7 SNP00149794 406 413 G G C E129 n/a n/a n/a n/a 41 7512297 489610R6 SNP00149794 207 412 G G C G129 n/a n/a n/a n/a 42 7512392 1237180F1 SNP00050284 218 5083 T T G noncoding n/d n/a n/a n/a 42 7512392 1237180F6 SNP00050284 206 5095 T T G noncoding n/d n/a n/a n/a 42 7512392 1655794T6 SNP00050284 147 5104 T T G noncoding n/d n/a n/a n/a 42 7512392 1830793T6 SNP00050284 150 5080 T T G noncoding n/d n/a n/a n/a 42 7512392 2210157F6 SNP00124093 257 4296 T T C noncoding n/a n/a n/a n/a Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 42 7512392 2210157T6 SNP00050284 153 5084 T T G noncoding n/d n/a n/a n/a 42 7512392 2398188T6 SNP00050284 50 5188 T T G noncoding n/d n/a n/a n/a 42 7512392 2650389H1 SNP00068897 70 4114 C C A noncoding n/d n/d n/d n/d 42 7512392 7083047H1 SNP00124089 270 1364 G G A noncoding n/d n/a n/a n/a 42 7512392 7172418H1 SNP00124090 324 2465 C C T noncoding n/d n/a n/a n/a 42 7512392 7183216H1 SNP00124092 262 3403 T T C noncoding n/a n/a n/a n/a 42 7512392 7633505J1 SNP00124091 232 2745 C G C noncoding n/d n/d n/d n/d 43 7511589 3899917H1 SNP00060090 153 1445 C G C noncoding n/d n/a n/a n/a 44 7511699 150629H1 SNP00008826 20 2048 A G A noncoding 0.59 0.74 0.45 n/a 44 7511699 150629H1 SNP00099341 156 2184 G G A noncoding n/a n/a n/a n/a 44 7511699 3594763H1 SNP00074861 272 736 G G noncoding 0.65 n/a n/a n/a 44 7511699 6620626H1 SNP00099340 284 436 A A C noncoding n/a n/a n/a n/a 44 7511699 8015628J1 SNP00143096 242 177 G G A E2 n/a n/a n/a n/a 44 7511699 8032909J1 SNP00099340 151 435 A A C noncoding n/d n/d n/d n/d 45 7512595 2991617H1 SNP00098449 176 913 T T C P272 n/a n/a n/a n/a 45 7512595 3597987H1 SNP00098448 241 243 C C T A49 0.96 0.97 0.93 0.86 45 7512595 7730065J1 SNP00098448 243 222 C C T T42 0.96 0.97 0.93 0.86 45 7512595 7741810J1 SNP00098449 301 889 T T C V264 n/a n/a n/a n/a 46 2803911 1442538H1 SNP00067670 42 5730 C C T noncoding n/d n/a n/a n/a 46 2803911 1561619H1 SNP00067671 127 5970 T T C noncoding n/a n/a n/a n/a 46 2803911 1561619H1 SNP00115076 25 5868 A A C noncoding n/a n/a n/a n/a 46 2803911 1651515F6 SNP00008916 168 6413 C T C noncoding n/a n/a n/a n/a 46 2803911 1651515F6 SNP00036667 131 6376 C C T noncoding n/a n/a n/a n/a 46 2803911 1651515T6 SNP00008916 254 6417 C T C noncoding n/a n/a n/a n/a 46 2803911 1669273F6 SNP00036665 153 3909 T C T N1077 0.81 n/a n/a n/a 46 2803911 1669273T6 SNP00008916 260 6414 T T C noncoding n/a n/a n/a n/a 46 2803911 1669273T6 SNP00036667 297 6377 C C T noncoding n/a n/a n/a n/a 46 2803911 1671466T6 SNP00036667 215 6448 C C T noncoding n/a n/a n/a n/a Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 46 2803911 1809936F6 SNP00062422 13 5054 A A C noncoding n/a n/a n/a n/a 46 2803911 1809936T6 SNP00008916 99 6421 C T C noncoding n/a n/a n/a n/a 46 2803911 1809936T6 SNP00036667 136 6384 C C T noncoding n/a n/a n/a n/a 46 2803911 1815491T6 SNP00008916 204 6466 C T C noncoding n/a n/a n/a n/a 46 2803911 1815491T6 SNP00036667 241 6429 C C T noncoding n/a n/a n/a n/a 46 2803911 2803911T6 SNP00067670 88 5732 C C T noncoding n/d n/a n/a n/a 46 2803911 3269335F6 SNP00036666 155 4287 C T C R1203 n/a n/a n/a n/a 46 2803911 3269335T6 SNP00008916 86 6462 T T C noncoding n/a n/a n/a n/a 46 2803911 3269335T6 SNP00036667 123 6425 C C T noncoding n/a n/a n/a n/a 46 2803911 326933T56 SNP00067671 534 6014 T T C noncoding n/a n/a n/a n/a 46 2803911 5353165T8 SNP00095294 351 2772 G A G A698 n/d n/a n/a n/a 46 2803911 6428734H1 SNP00118789 71 912 C C G T78 n/a n/a n/a n/a 46 2803911 7242635F8 SNP00138530 543 2688 C C T L670 n/a n/a n/a n/a 47 7511806 2268132H1 SNP00000909 97 322 T T C noncoding n/a n/a n/a n/a 49 7512306 1907894H1 SNP00027551 10 59 G G A M1 n/a n/a n/a n/a 50 7512607 1274866F1 SNP00044866 170 259 C C T F63 n/a n/a n/a n/a 50 7512607 1348065H1 SNP00076259 14 1226 G G T noncoding n/a n/a n/a n/a 50 7512607 1533120H1 SNP00114946 184 1198 T T C noncoding n/a n/a n/a n/a 50 7512607 2158183H1 SNP00034548 35 1056 G A G noncoding n/a n/a n/a n/a 50 7512607 3694931H1 SNP00098582 141 1033 G G A noncoding n/a n/a n/a n/a 50 7512607 7746385H1 SNP00044866 243 260 C C T H64 n/a n/a n/a n/a 52 7512569 2077665h1 SNP00028750 64 102 C C T noncoding n/D n/a n/a n/a 52 7512569 3075539h1 SNP00107593 6 838 G G A noncoding n/a n/a n/a n/a 52 7512569 3075539h1 SNP00107594 225 1057 C C A noncoding n/d n/d n/d n/d 52 7512569 3493163h1 SNP00107592 158 770 A G A P151 n/d n/a n/a n/a 53 7512570 2077665h1 SNP00028750 64 102 C C T noncoding n/d n/a n/a n/a 54 7512846 004785H1 SNP00006320 94 1553 T T C I483 n/a n/a n/a n/a 54 7512846 1286765H1 SNP00029631 138 1964 C C T noncoding n/a n/a n/a n/a Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 54 7512846 1286765H1 SNP00029632 79 1905 C C T noncoding n/a n/a n/a n/a 54 7512846 1413704F6 SNP00029630 309 965 T T C P287 n/a n/a n/a n/a 54 7512846 1433660H1 SNP00029630 35 964 T T C L287 n/a n/a n/a n/a 54 7512846 1695746T6 SNP00029632 51 1946 C C T noncoding n/a n/a n/a n/a 54 7512846 1904674T6 SNP00029631 24 1965 C C T noncoding n/a n/a n/a n/a 54 7512846 1904674T6 SNP00029632 83 1906 C C T noncoding n/a n/a n/a n/a 54 7512846 2741335T6 SNP00029631 12 1988 C C T noncoding n/a n/a n/a n/a 54 7512846 2741335T6 SNP00029632 71 1929 C C T noncoding n/a n/a n/a n/a 54 7512846 2879851T6 SNP00029631 25 1963 C C T noncoding n/a n/a n/a n/a 54 7512846 2879851T6 SNP00029632 84 1904 C C T noncoding n/a n/a n/a n/a 54 7512846 7124360H1 SNP00063465 205 1651 A A G E516 n/a n/a n/a n/a 54 7512846 7128954H1 SNP00063465 179 1650 G A G E516 n/a n/a n/a n/a 55 7517814 2079106F6 SNP00060957 24 1120 G A G noncoding n/a n/a n/a n/a 55 7517814 3518064H1 SNP00060957 53 1100 A A G noncoding n/a n/a n/a n/a 56 7519828 1219552R7 SNP00015105 126 1146 C C T G380 n/a n/a n/a n/a 56 7519828 1219552R7 SNP00066214 5 1025 T T G V340 n/a n/a n/a n/a 56 7519828 2470766T6 SNP00015105 199 1184 C C T S393 n/a n/a n/a n/a 56 7519828 2470766T6 SNP00066214 320 1063 T T G L353 n/a n/a n/a n/a 56 7519828 2907233H1 SNP00071680 64 159 C C A G51 n/d n/a n/a n/a 56 7519828 2907233H1 SNP00071681 119 214 T T C C70 n/a n/a n/a n/a 56 7519828 876382R6 SNP00071681 406 215 C T C P70 n/a n/a n/a n/a 57 7519833 1219552R7 SNP00015105 126 1288 C C T noncoding n/a n/a n/a n/a 57 7519833 1219552R7 SNP00066214 5 1167 T T G noncoding n/a n/a n/a n/a 57 7519833 2470766T6 SNP00015105 199 1326 C C T noncoding n/a n/a n/a n/a 57 7519833 2470766T6 SNP00066214 320 1205 T T G noncoding n/a n/a n/a n/a 57 7519833 2907233H1 SNP00071680 64 159 C C A G51 n/d n/a n/a n/a 57 7519833 2907233H1 SNP00071681 119 214 T T C C70 n/a n/a n/a n/a 57 7519833 876382R6 SNP00071681 406 215 C T C P70 n/a n/a n/a n/a Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 58 7519834 1219552R7 SNP00015105 126 1092 C C T G362 n/a n/a n/a n/a 58 7519834 1219552R7 SNP00066214 5 971 T T G V322 n/a n/a n/a n/a 58 7519834 2470766T6 SNP00015105 199 1130 C C T S375 n/a n/a n/a n/a 58 7519834 2470766T6 SNP00066214 320 1009 T T G L335 n/a n/a n/a n/a 58 7519834 2907233H1 SNP00071680 64 159 C C A G51 n/d n/a n/a n/a 58 7519834 2907233H1 SNP00071681 119 214 T T C C70 n/a n/a n/a n/a 58 7519834 876382R6 SNP00071681 406 215 C T C P70 n/a n/a n/a n/a 59 7519939 1359604H1 SNP00006194 222 157 G G A P42 0.66 0.70 0.64 0.61 59 7519939 1359604H1 SNP00029418 174 109 C C T T26 n/a n/a n/a n/a 59 7519939 1732361F6 SNP00006194 222 158 A G A I43 0.66 0.70 0.64 0.61 59 7519939 1732361F6 SNP00029418 174 110 C C T L27 n/a n/a n/a n/a 59 7519939 2644970F6 SNP00006194 216 160 G G A L43 0.66 0.70 0.64 0.61 59 7519939 2644970F6 SNP00029418 168 112 C C T F27 n/a n/a n/a n/a 60 7520133 1275278H1 SNP000020695 89 835 C C T A272 n/a n/a n/a n/a 60 7520133 1480416H1 SNP00020694 79 428 A A G A136 0.95 n/a n/a n/a 60 7520133 3016303H1 SNP00136636 173 739 C C A T240 n/d n/a n/a n/a 60 7520133 3088849H1 SNP00020446 65 79 G A G S20 n/d n/d n/d n/a 60 7520133 8020007J1 SNP00020694 480 456 A A G K146 0.95 n/a n/a n/a 60 7520133 8020007J1 SNP00136636 169 767 C C A G249 n/d n/a n/a n/a 61 7520171 2651464F6 SNP00152518 287 802 A A G noncoding n/a n/a n/a n/a 61 7520171 2651464T6 SNP00152518 71 796 A A G noncoding n/a n/a n/a n/a 61 7520171 4825157H1 SNP00152518 52 789 A A G noncoding n/a n/a n/a n/a 62 7522453 1223609H1 SNP00015477 47 2776 C C A noncoding n/d n/d n/d n/d 62 7522453 1273442T6 SNP00015477 238 2783 C C A noncoding n/d n/d n/d n/d 62 7522453 1553729T6 SNP00015477 204 2816 C C A noncoding n/d n/d n/d n/d 62 7522453 2362019T6 SNP00015477 125 2895 C C A noncoding n/d n/d n/d n/d 63 7522454 1223609H1 SNP00015477 47 2539 C C A noncoding n/d n/d n/d n/d 63 7522454 1273442T6 SNP00015477 238 2546 C C A noncoding n/d n/d n/d n/d Table 8 SEQ PID EST ID SNP ID EST CB1 EST Allele Allele Amino Acid Caucasian African Asian Hispanic ID SNP SNP Allele 1 2 Allele 1 Allele 1 Allele 1 Allele 1 NO: frequency frequency frequency frequency 63 7522454 1553729T6 SNP00015477 204 2579 C C A noncoding n/d n/d n/d n/d 63 7522454 2362019T6 SNP00015477 125 2658 C C A noncoding n/d n/d n/d n/d 66 7519293 1299369F6 SNP00012028 243 768 C T C F249 n/a n/a n/a n/a 66 7519293 1466334H1 SNP00098884 22 16 C C T noncoding n/a n/a n/a n/a 66 7519293 2449607F6 SNP00052598 415 431 A A G D137 0.99 n/d n/d n/d 66 7519293 4620808T6 SNP00012028 255 862 T T C noncoding n/a n/a n/a n/a