AU2287101A - Novel proteins and nucleic acids encoding same - Google Patents

Novel proteins and nucleic acids encoding same Download PDF

Info

Publication number
AU2287101A
AU2287101A AU22871/01A AU2287101A AU2287101A AU 2287101 A AU2287101 A AU 2287101A AU 22871/01 A AU22871/01 A AU 22871/01A AU 2287101 A AU2287101 A AU 2287101A AU 2287101 A AU2287101 A AU 2287101A
Authority
AU
Australia
Prior art keywords
fctrx
nucleic acid
polypeptide
amino acid
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU22871/01A
Inventor
Catherine E. Burgess
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CuraGen Corp
Original Assignee
CuraGen Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by CuraGen Corp filed Critical CuraGen Corp
Priority claimed from PCT/US2000/034898 external-priority patent/WO2001046231A2/en
Publication of AU2287101A publication Critical patent/AU2287101A/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

WO 01/46231 PCT/USOO/34898 NOVEL PROTEINS AND NUCLEIC ACIDS ENCODING SAME FIELD OF THE INVENTION The invention generally relates to nucleic acids and polypeptides encoded therefrom. 5 More specifically, the invention relates to nucleic acids encoding novel polypeptides, as well as vectors, host cells, antibodies, and recombinant methods for producing these nucleic acids and polypeptides. SUMMARY OF THE INVENTION The invention is based in part upon the discovery of novel nucleic acid sequences 0 encoding novel polypeptides. Nucleic acids encoding the polypeptides disclosed in the invention, and derivatives and fragments thereof, will hereinafter be collectively designated as "FCTRX" nucleic acid or polypeptide sequences. In one aspect, the invention provides an isolated FCTRX nucleic acid molecule encoding a FCTRX polypeptide that includes a nucleic acid sequence that has identity to the 5 nucleic acids disclosed in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. In some embodiments, the FCTRX nucleic acid molecule can hybridize under stringent conditions to a nucleic acid sequence complementary to a nucleic acid molecule that includes a protein-coding sequence of a FCTRX nucleic acid sequence. The invention also includes an isolated nucleic acid that encodes a FCTRX polypeptide, or a fragment, homolog, analog or 0 derivative thereof. For example, the nucleic acid can encode a polypeptide at least 80% identical to a polypeptide comprising the amino acid sequences of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. The nucleic acid can be, for example, a genomic DNA fragment or a cDNA molecule that includes the nucleic acid sequence of any of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. 5 Also included in the invention is an oligonucleotide, e.g., an oligonucleotide which includes at least 6 contiguous nucleotides of a FCTRX nucleic acid (e.g., SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29) or a complement of said oligonucleotide. Also included in the invention are substantially purified FCTRX polypeptides (SEQ ID NO:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30). In some embodiments, the 0 FCTRX polypeptides include an amino acid sequence that is substantially identical to the amino acid sequence of a huma FCTRX polypeptide. 1 WO 01/46231 PCT/USOO/34898 The invention also features antibodies that immunoselectively-binds to FCTRX polypeptides. In another aspect, the invention includes pharmaceutical compositions which include therapeutically- or prophylactically-effective amounts of a therapeutic and a pharmaceutically 5 acceptable carrier. The therapeutic can be, e.g., a FCTRX nucleic acid, a FCTRX polypeptide, or an antibody specific for a FCTRX polypeptide. In a further aspect, the invention includes, in one or more containers, a therapeutically- or prophylactically-effective amount of this pharmaceutical composition. In a further aspect, the invention includes a method of producing a polypeptide by 0 culturing a cell that includes a FCTRX nucleic acid, under conditions allowing for expression of the FCTRX polypeptide encoded by the DNA. If desired, the FCTRX polypeptide can then be recovered. In another aspect, the invention includes a method of detecting the presence of a FCTRX polypeptide in a sample. In the method, a sample is contacted with a compound that 5 selectively binds to the polypeptide under conditions allowing for formation of a complex between the polypeptide and the compound. The complex is detected, if present, thereby identifying the FCTRX polypeptide within the sample. The invention also includes methods to identify specific cell or tissue types based on their expression of a FCTRX. 0 Also included in the invention is a method of detecting the presence of a FCTRX nucleic acid molecule in a sample by contacting the sample with a FCTRX nucleic acid probe or primer, and detecting whether the nucleic acid probe or primer bound to a FCTRX nucleic acid molecule in the sample. In a further aspect, the invention provides a method for modulating the activity of a 5 FCTRX polypeptide by contacting a cell sample that includes the FCTRX polypeptide with a compound that binds to the FCTRX polypeptide in an amount sufficient to modulate the activity of said polypeptide. The compound can be, e.g., a small molecule, such as a nucleic acid, peptide, polypeptide, peptidomimetic, carbohydrate, lipid or other organic (carbon containing) or inorganic molecule, as further described herein. 0 Also within the scope of the invention is the use of a Therapeutic in the manufacture of a medicament for treating or preventing disorders or syndromes including, e.g., cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. The Therapeutic can be, e.g., a FCTRX nucleic acid, a FCTRX 2 WO 01/46231 PCT/USOO/34898 polypeptide, or a FCTRX-specific antibody, or biologically-active derivatives or fragments thereof. The invention further includes a method for screening for a modulator of disorders or syndromes including, e.g., cancer, neurodegenerative disorders, Alzheimer's Disease, 5 Parkinson's Disorder, immune disorders, and hematopoietic disorders. The method includes contacting a test compound with a FCTRX polypeptide and determining if the test compound binds to said FCTRX polypeptide. Binding of the test compound to the FCTRX polypeptide indicates the test compound is a modulator of activity, or of latency or predisposition to the aforementioned disorders or syndromes. 0 Also within the scope of the invention is a method for screening for a modulator of activity, or of latency or predisposition to an disorders or syndromes including, e.g., cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, by administering a test compound to a test animal at increased risk for the aforementioned disorders or syndromes. The test animal expresses a recombinant 5 polypeptide encoded by a FCTRX nucleic acid. Expression or activity of FCTRX polypeptide is then measured in the test animal, as is expression or activity of the protein in a control animal which recombinantly-expresses FCTRX polypeptide and is not at increased risk for the disorder or syndrome. Next, the expression of FCTRX polypeptide in both the test animal and the control animal is compared. A change in the activity of FCTRX polypeptide in the test 0 animal relative to the control animal indicates the test compound is a modulator of latency of the disorder or syndrome. In yet another aspect, the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a FCTRX polypeptide, a FCTRX nucleic acid, or both, in a subject (e.g., a human subject). The method includes 5 measuring the amount of the FCTRX polypeptide in a test sample from the subject and comparing the amount of the polypeptide in the test sample to the amount of the FCTRX polypeptide present in a control sample. An alteration in the level of the FCTRX polypeptide in the test sample as compared to the control sample indicates the presence of or predisposition to a disease in the subject. Preferably, the predisposition includes, e.g., cancer, 0 neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. Also, the expression levels of the new polypeptides of the invention can be used in a method to screen for various cancers. In a further aspect, the invention includes a method of treating or preventing a pathological condition associated with a disorder in a mammal by administering to the subject 3 WO 01/46231 PCT/USOO/34898 a FCTRX polypeptide, a FCTRX nucleic acid, or a FCTRX-specific antibody to a subject (e.g., a human subject), in an amount sufficient to alleviate or prevent the pathological condition. In preferred embodiments, the disorder, includes, e.g., cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic 5 disorders. In yet another aspect, the invention can be used in a method to identity the cellular receptors and downstream effectors of the invention by any one of a number of techniques commonly employed in the art. These include but are not limited to the two-hybrid system, affinity purification, co-precipitation with antibodies or other specific-interacting molecules. 0 Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references 5 mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. Other features and advantages of the invention will be apparent from the following detailed description and claims. 0 DETAILED DESCRIPTION The invention is based, in part, upon the discovery of novel nucleic acid sequences that encode novel polypeptides. The novel nucleic acids and their encoded polypeptides are referred to individually as FCTR1, FCTR2, FCTR3, FCTR4, FCTR5, FCTR6, FCTR7, FCTR8, FCTR9, FCTR10, FCTR11, FCTR12, FCTR13, and FCTR14. The nucleic acids, and 5 their encoded polypeptides, are collectively designated herein as "FCTRX". The novel FCTRX nucleic acids of the invention include the nucleic acids whose sequences are provided in Tables 1A, 2A, 3A, 4A, 5A, 6A, 7A, 8A, 9A, 10A, 11A, 12A, 13A, and 14A, inclusive ("Tables 1A - 14A"), or a fragment thereof. The invention also includes a mutant or variant FCTRX nucleic acid, any of whose bases may be changed from the 0 corresponding base shown in Tables 1A - 14A while still encoding a protein that maintains the activities and physiological functions of the FCTRX protein fragment, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including complementary nucleic acid fragments. The 4 WO 01/46231 PCT/USOO/34898 invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the 5 chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to 20% or more of the bases may be so changed. The novel FCTRX proteins of the invention include the protein fragments whose 0 sequences are provided in Tables IB, 2B, 3B, 4B, 5B, 6B, 7B, 8B, 9B, 10B, IB, 12B, 13B, and 14B, inclusive ("Tables 1B - 14B"). The invention also includes a FCTRX mutant or variant protein, any of whose residues may be changed from the corresponding residue shown in Tables 1B - 14B while still encoding a protein that maintains its native activities and physiological functions, or a functional fragment thereof. In the mutant or variant FCTRX 5 protein, up to 20% or more of the residues may be so changed. The invention further encompasses antibodies and antibody fragments, such as Fab or (Fab)2, that bind immunospecifically to any of the FCTRX proteins of the invention. FCTR1 (AL031943_A) The novel FCTR1 nucleic acid encoding a C-terminal fragment of a novel FCTR1 0 protein is shown in Table 1A. A "TAA" stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The stop codon is shown in bold letters. This sequence originates in chromosome 6. No ATG start codon was found, indicating that the cDNA extends 5' of the disclosed sequence in Table 1A. Table 1A. FCTR1 (ALO31943_A) nucleotide fragment (SEQ ID NO:1). 5 acccatctttttctcttcttcgtgctcctaaacttaggctaccaagctttgctggggaaagcactccag gtgggtgttactacaaatcaccgtctgctgacccactggtactacctgacagcctttgatatttccaga gtcaatacctgctttccattctccacagcatctaatataagtcatggcttctcatctgtcctgcttccc cgcttcgcgttcaccactgtgctgagatatagggaaaggaatgggaacaaggaagccatcgccggcctc tccagctctggaggcttcacagcttgcctcctccttcgtctgttgagtcatcccacacgcaaccacaac 0 tatgtgggagattctgtgccaggctttggcaactaa The encoded C-terminal fragment of the encoded protein is presented using the one letter code in Table 1B. The protein including the C-terminal fragment disclosed has a high probability of being secreted extracellularly. A signal peptide most likely is cleaved between 5 residues 19 and 20, i.e., at the dash in the amino acid sequence LLG-KAL. 5 WO 01/46231 PCT/USOO/34898 Table 1B. C-terminal fragment of the encoded FCTR1 protein sequence (SEQ ID NO:2). THLFLFFVLLNLGYQALLGKALQVGVTTNHRLLTHWYYLTAFDISRVNTCFPFSTASNISHGFSSVLLP RFAFTTVLRYRERNGNKEAIAGLSSSGGFTACLLLRLLSHPTRNHNYVGDSVPGFGN 5 In a search of sequence databases, no similarities were found to any known expressed nucleic acid or protein. The human genomic fragment HS223B 1, from clone RP 1 -223B 1 on chromosome 6p24.1-25.3, aligned with the FCTR1 nucleotide sequence, as shown in Table 1C. Putative intron and exon information can be construed from this alignment. Table IC. BLASTN alignments of FCTR1 (SEQ ID NO:1) with genomic clone HS223B1 0 Alignment between: HS223B1 Human DNA sequence from clone RPl-223B1 on chromosome 6 p 24 .1- 25
.
3 Contains STSs and GSSs, complete sequence. 5/2000 and (PastedNo.:1-228) Length 126281 Score - 452.0, bits (228.0), Expect = le-125 5 Identities = 228/228 (100%) Strand Plus / Plus Query: 1 acccatctttttctcttcttcgtgctcctaaacttaggctaccaagctttgctggggaaa 60 l II I1111 i i I I I |1 l 11111 i I 11 Il IIi i1 o Sbjct: 1483 acccatctttttctcttcttcgtgctcctaaacttaggctaccaagctttgctggggaaa 1542 Query: 61 gcactccaggtgggtgttactacaaatcaccgtctgctgacccactggtactacctgaca 120 Sbjct: 1543 gcactccaggtgggtgttactacaaatcaccgtctgctgacccactggtactacctgaca 1602 5 Query: 121 gcctttgatatttccagagtcaatacctgctttccattctccacagcatctaatataagt 180 1 1 lI iI li Il lI iI 111111111 I l Ii i11 (11I II I 111111I1lI I| I i Sbjct: 1603 gcctttgatatttccagagtcaatacctgctttccattctccacagcatctaatataagt 1662 0 Query: 181 catggcttctcatctgtcctgcttccccgcttcgcgttcaccactgtg 228 Sbjct: 1663 catggcttctcatctgtcctgcttccccgcttcgcgttcaccactgtg 1710 (SEQ ID NO:34) Alignment between: 5 HS223B1 Human DNA sequence from clone RP1-223B1 on chromosome 6p24.1-25.3 Contains STSs and GSSs, complete sequence. 5/2000 and (PastedNo.:226-381) Length 126281 Score 309.0, bits (156.0), Expect = 5e-82 Identities = 156/156 (100%) 0 Strand Plus / Plus Query: 226 gtgctgagatatagggaaaggaatgggaacaaggaagccatcgccggcctctccagctct 285 Sbjct: 4527 gtgctgagatatagggaaaggaatgggaacaaggaagccatcgccggcctctccagctct 4586 5 Query: 286 ggaggcttcacagcttgcctcctccttcgtctgttgagtcatcccacacgcaaccacaac 345 Sbjct: 4587 ggaggcttcacagcttgcctcctccttcgtctgttgagtcatcccacacgcaaccacaac 4646 0 Query: 346 tatgtgggagattctgtgccaggctttggcaactaa 381 Sbjct: 4647 tatgtgggagattctgtgccaggctttggcaactaa 4682 (SEQ ID NO:35) The nucleic acids and proteins of the invention are potentially useful in the treatment 5 of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 6 WO 01/46231 PCT/USOO/34898 FCTR2 (AL078594_A) The novel nucleic acid encoding a novel protein C-terminal fragment is shown in Table 2A. The initiation codon is at the 5' end, and a "TAG" stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The start and stop codons are shown in 5 bold letters. This sequence originates in chromosome 6, in clone RP1-293L8 at map location q22.2-22.33. Homology of 100% was shown to the human genomic clone HSDJ293L8 obtained from this region, which contains the HEY2 gene for hairy/enhancer-of-split related with YRPW motif 2 (cardiovascular basic helix-loop-helix factor 1, CHF1), ESTs, STSs, GSSs and four putative CpG islands. FCTR2 nucleotide regions 1-213, 214-367, and 366-570 0 correspond 100% to HSDJ293L8 regions 49502-49714, 52745-52898, and 54432-54636, respectively. Table 2A. Nucleotide sequence (SEQ ID NO:3) of FCTR2 (AL078594_A). atgactgtcaaggctcctaaaggtcataaaggtgacataacttctatactgttagttcaaacacttgct cagagctgccatgctgtgaggaggcccaagctagtcagctcagagagagcatctggagaggctctgaag 5 ctacacaactatagagtcctcagctgcacaagccccctgctgttccagctccaaccactgctagactac aaccatatgatactgagtaacttagccccagacgtcagggtgccactgagtatgcagtatgctgactta atcataaaaattaacacctttagtattcaagcagctcatatcactcacaaatttctctttaacaaagaa aggcatgcatttcatacacggggacaattcggtcagattgtttcttcccaatacctctatgagatcaat tgcactgaaggaatgcctatttttactagaagaacgaaggtggaagtcaataattttgaagcatggggt 0 agcttcagaggaggagaggttcggggatcgggtacaagacttggcttgggccaggataaaaatactcag tatgaaaaacctgag tag The encoded FCTR2 polypeptide sequence (SEQ ID NO.:4) is presented using the one letter code in Table 2B. The protein appears not to have a strong probability of secretion. No 5 signal peptide is predicted for this protein. No significant matches were found in a BLASTP search against the FCTR2 polypeptide. Table 2B. Encoded FCTR2 protein sequence (SEQ ID NO:4). MTVKAPKGHKGDITSILLVQTLAQSCHAVRRPKLVSSE RASGEALKLHNYRVLSCTSPLLFQLQPLLDY NHMILSNLAPDVRVPLSMQYADL IIKINTFSIQAAHITHKFLFNKERHAFHTRGQFGQIVSSQYLYEIN 0 CTEGMPIFTRRTKVEVNNFEAWGSFRGGEVRGSGTRLGLGQDKNTQYEKPE The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 5 FCTR3 (AL078595_A) The novel nucleic acid encoding a novel protein C-terminal fragment is shown in Table 3A. The initiation codon is at the 5' end and a TGA stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The start and stop codons are shown in 7 WO 01/46231 PCT/USOO/34898 bold letters. This sequence originates in clone RP3-399J4 on chromosome 6q15-16.3. No significant matches were found in a BLASTN search against the FCTR3 nucleotide sequence. Table 3A. Nucleotide sequence (SEQ ID NO:5) of FCTR3 (AL078595 A). atgccgccactgctggtcctgctcttgctcctgccgccaccacttgcacctcccctcttcagccagtgt 5 ggtggcagcggctgctcccgacagcccaccattcccatcagtaatatggaggggcaaatatgtgtaaag ccttcaggtgccaaagctgctccagaacccctggaagaattatcaaagatgcggtccctctcttcaatt ccatggtatattttgtccttcagttctgcagagcctgcaatcaaacatgctaaagcagagaaatacaat aagagacctatacttgacattagcagaggaagtccagctgtgtacactaattatgataaacatccattc acaatgtctgggaggagactagccacagacctggaaagaggtgaagaaaaacgacaccatgaaaaagga 0 gcaaagtga The encoded protein is presented using the one-letter code in Table 3B. The protein has a high probability of extracellular secretion. A signal peptide is predicted for this protein with a cleavage site between residues 16 and 17, i.e., at the dash in the amino acid sequence 5 PLA-PPL. No significant matches were found in a BLASTP search against the FCTR3 polypeptide. Table 3B. Encoded FCTR3 protein sequence (SEQ ID NO:6). MPPLLVLLLLLPPPLAPPLFSQCGGSGCSRQPTIPISNMEGQICVKPSGAKAAPEPLEELSKMRSLSSI PWYILSFSSAEPAIKHAKAEKYNKRPILDISRGSPAVYTNYDKHPFTMSGRRLATDLERGEEKRHHEKG 0 AK The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 5 FCTR4 (AL109627 A) The novel nucleic acid encoding a novel transforming immortalized mammary oncogene-like protein is shown in Table 4A. An initiation codon is shown at the beginning of the sequence and a TGA stop codon was identified at the 3' end indicating that this sequence is a coding sequence. This sequence originates in chromosome 1 from clone RP4-733M16 at 0 map location p36.11-36.23. Table 4A. Nucleotide sequence (SEQ ID NO:7) of FCTR4 (AL109627 A). atggccagacctcccgtgcccggttcggtggttgtcccaaactggcacgagagtgccgagggcaaggag tacctggcttgcattctgcgcaagaaccgccggcgggtgtttgggctgcttgagcggccagtgctgctg ccgcctgtgtccattgacactgccagctacaagatctttgtgtccgggaagagtggtgtgggcaagacg 5 gcgctggtggccaagctggctggcctggaggtgcctgtggtgcaccacgagaccaccggcatccagacc accgtggtattttggccagccaagctgcaggccagcagccgtgtcgtcatgtttcgttttgagttctgg gactgtggagagtctgcactcaaaaagttcgatcatatgctgctggcttgcatggagaacacagatgcc ttcctcttctttctccttcactgaccgtgcctctttgaagacctcctggacagtggcccgcata gcaggtgaggcccctggtgtcgtcaggatggtcatcggctccaaatttgaccagtacatgcacacggac 0 gtgcccgagcgggacctcacagccttccggcaggcctgggagctgcccctgctacgggtgaagagtgtg ccggggcggcggctggctgatgggcgcacactggacgggcgggctgggctggccgacgttgcccacata 8 WO 01/46231 PCT/USOO/34898 ctcaatggccttgctgagcagctgtggcaccaggaccagacggcgatgacgccaccgacaggacgacga ctgtgtctcgcgccctgcggcggcatttatgtgccggactctaggggtacattttctgagacgggaaaa cctgcattgataaaagtgggacagagcggggtcagaccgctcctaactgtccccctgaccccgcgatgg gttagacttcgtgctcgcctgggaggagaagctgcgacccccgcggcggcgggagagaggcgactccgg 5 cagcggcgctggcgcgagaattttcagcggaacctggaggagggcctctttgaactgcctgggtaccag gtacccggttcagatctcaactcttgccaattgctgtacccatactgggcttgctggggatactggcac aagtaccagcccctggaccagcctttggacaaactgagctgcctctttgaccacccaggaaccgtgttc ttcagcatcttcatgtccttctggggccatggccttcctggagcactggaagcagggagtgccaccttg gcccaccactgggactgcagtgacttccaggaccaggaggcaatgcccagttcagccccccaccactgg 0 gactgcagcgacttccaggaccaggaggtgatgcccagttcagccctccaccactgggactgcagcgac ttccaggaccaggaggagtgcccacatctacagtttgctgccctggccctgcagatgacccagaaccca gtgacaggcttgaaggagccctacttccaaccgcacagctgcctttcccacctactcaccagctctgca gccatcctcactgtgctctgtgtggtgatgattttcctggtatctgtcataatttaccatggcatcatc agcattgcaatgttccacactggcaactctgtgctcatgacccaagcgaatgtcctttggggcaatgga 5 ggccccaaagccctgagtaaggtgctctgtgtctgccaacaacagtgcggtcctggtggctgccacatt caggtcacccagcagctcatcatcatcatggtgggcaaacagctgctcaaccacatggaagaatttgtt gggctgggaggtggccccgggcctgacactccctgcctgccagagctgcagtttgggttcatcaccatc tttgtgggagccttcctgctggcacccctgttcactctgctcaacaaccgggtagagattggactggac gcccacaagttcctgtgcaagtaccagcgaccaatggctgggcgcggctggacatctggatctgactgc 0 tcctgctggaggccatgtgagctgattctgccccggacaaatgcgcggagccggctagggtactggctg aacgggcagggccagattctagggagaaggaggggaggaaatgcggggttcggagtcgagatccgagag cctctccagaccccgcaacccagatacaaggcctctcgcgacgtgggggtgaacctcgccctcttctac tggaagctgctggctgtgcatgtgcatctgggtttcattatcgccttcgagggtttgatgaatcaaact ctttgtctgggtgggatctcccccagccagctgggcagagagagggcttcccctgccggaacagccaaa 5 cagcatcagcagcgggcctgggcccagagagggccaggtgggtggcagagcaaaagaggaatggactgt gggccacctgctaccctccagccccacctgactgggccacctggcactgcccaccaccctgtagcagtg tgccagcaggagagtctgtcctttgcagagctgcccgccctgaagcccccgagcccagtgtgtctggac cttttccctgttgccccagaggagcttcgggctcctggcagccgctggtccctggggacccctgcccct ctccaagggttgctatggccattatccccaggaggctcagatacagagatcaccagcggggggatgcgg 0 cccagcagggctggcagctggccacactgtcctggtgcccagcccccagctctggagggaccctggagt ccccgacacacacagccacagcgccgggccagccacggctcggagaagaagtctgcctggcgcaagatg cgggtgtaccagcgtgaagaggtccccggctgccccgaggcccacgctgtcttcctagagcctggccag gtagtgcaagagcaggccctgagcacagaggagcccagggtggagttgtctgggtccacccgagtgagc ctcgaaggtcctgagcggaggcgcttctcggcatcggagctgatgacccggctgcactcttctctgcgc 5 ctggggcggaattcagcagcccgggcactcatctctgggtcaggcaccggagcagcccgggaagggaaa gcatctggaatggaggctcgaagtgtagagatgagcggggaccgggtgtcgcggccagcccctggtgac tcacgagagggcgattggtccgagcccaggctagacacacaggaagagccgcctttggggtccaggagc accaacgagcggcgccagtctcgattcctccttaactccgtcctctatcaggaatacagcgacgtggcc agcgcccgcgaactgcggcggcagcagcgcgaggaggagggcccgggggacgaggccgagggcgcagag 0 gaggggccggggccgccgcgggccaacctctcccccagcagctccttccgggcgcagcgctcggcgcga ggctccaccttctcgctgtggcaggatatccccgacgtacgcggcagcggcgtcctggccacgctgagc ctgcgggactgcaagctgcaggaggccaagtttgagctgatcacctccgaggcctcctacatccacagc ctgtcggtggctgtgggccacttcttaggctctgccgagctgagcgagtgtctgggggcgcaggacaag cagtggctgttttccaaactgcccgaggtcaagagcaccagcgagaggttcctgcaggacctggagcag 5 cggctggaggcagatgtgctgcgcttcagcgtgtgcgacgtggtgctggaccactgcccggccttccgc agagtctacctgccctatgtcaccaaccaggcctaccaggagcgcacctaccagcgcctgctcctggag aaccccaggttccctggcatcctggctcgcctggaggagtctcctgtgtgccagcgtctgccccttacc tccttccttatcctgcccttccagaggatcacccgcctcaagatgttggtggagaacatcctgaagcgg acagcacagggctctgaagacgaagacatggccaccaaggccttcaatgcgctcaaggagctggtgcag 0 gagtgcaatgctagtgtacagtccatgaagaggacagaggaactcatccacctgagcaagaagatccac tttgagggcaagattttcccgctgatctctcaggcccgctggctggttcggcatggagagttggtagag ctggcaccactgcctgcagcaccccctgccaagctgaagctgtccagcaaggcagtctacctccacctc ttcaatgactgcttgctgctctctcggcggaaggagctagggaagtttgccgttttcgtccatgccaag atggctgagctgcaggtgcgggacctgagcctgaagctgcagggcatccccggccacgtgttcctcctc 5 cagctcctccacgggcagcacatgaagcaccagttcctgctgcgggcccggacggaaagtgagaagcag cgatggatctcagccttgtgcccctccagcccccaggaggacaaggaggtcatcagtgagggggaagat tgcccccaggttcagtgtgttaggacatacaaggcactgcacccagatgagctgaccttggagaagact 9 WO 01/46231 PCT/USOO/34898 gacatcctgtcagtgaggacctggaccagtgacggctggctggaaggggtccgcctggcagatggtgag aaggggtgggtgccccaggcctatgtggaagagatcagcagcctcagcgcccgcctccgaaacctccgg gagaataagcgagtcacaagtgccaccagcaaactgggggaggctcctgtgtga 5 The encoded protein is presented using the one-letter code in Table 4B. The protein has a high probability of sorting into the plasma membrane. No signal peptide is predicted to occur for this protein. Table 4B. Encoded FCTR4 protein sequence (SEQ ID NO:8). MARPPVPGSVVVPNWHESAEGKEYLACILRKNRRRVFGLLERPVLLPPVSIDTASYKIFVSGKSGVGKT 0 ALVAKLAGLEVPVVHHETTGIQTTVVFWPAKLQASSRVVMFRFEFWDCGESALKKFDHMLLACMENTDA FLFLFSFTDRASFEDLPGQLARIAGEAPGVVRMVIGSKFDQYMHTDVPERDLTAFRQAWELPLLRVKSV PGRRLADGRTLDGRAGLADVAHILNGLAEQLWHQDQTAMTPPTGRRLCLAPCGGIYVPDSRGTFSETGK PAL IKVGQSGVRPLLTVPLT PRWVRLRARLGGEAAT PAAAGERRLRQRRWRENFQRNLEEGLFELPGYQ VPGSDLNSCQLLYPYWACWGYWHKYQPLDQPLDKLSCLFDHPGTVFFSIFMSFWGHGLPGALEAGSATL 5 AHHWDCSDFQDQEAMPSSAPHHWDCSDFQDQEVMPSSALHHWDCSDFQDQEECPHLQFAALALQMTQNP VTGLKEPYFQPHSCLSHLLTSSAAILTVLCVVMIFLVSVIIYHGIISIAMFHTGNSVLMTQANVLWGNG GPKALSKVLCVCQQQCGPGGCHIQVTQQLIIIMVGKQLLNHMEEFVGLGGGPGPDTPCLPELQFGFITI FVGAFLLAPLFTLLNNRVEIGLDAHKFLCKYQRPMAGRGWTSGSDCSCWRPCELILPRTNARSRLGYWL NGQGQILGRRRGGNAGFGVEIREPLQTPQPRYKASRDVGVNLALFYWKLLAVHVHLGFI IAFEGLMNQT 0 LCLGGISPSQLGRERASPAGTAKQHQQRAWAQRGPGGWQSKRGMDCGPPATLQPHLTGPPGTAHHPVAV CQQESLSFAELPALKPPS PVCLDLFPVAPEELRAPGSRWSLGTPAPLQGLLWPLS PGGS DTEI TSGGMR PSRAGSWPHCPGAQPPALEGPWS PRHTQPQRRASHGSEKKSAWRKMRVYQREEVPGCPEAHAVFLEPGQ VVQEQALSTEEPRVELSGSTRVSLEGPERRRFSASELMTRLHSSLRLGRNSAARALISGSGTGAAREGK ASGMEARSVEMSGDRVSRPAPGDSREGDWSEPRLDTQEEPPLGSRSTNERRQSRFLLNSVLYQEYSDVA 5 SARELRRQQREEEGPGDEAEGAEEGPGPPRANLSPSSSFRAQRSARGSTFSLWQDIPDVRGSGVLATLS LRDCKLQEAKFELI TSEASYIHSLSVAVGHFLGSAELSECLGAQDKQWLFSKLPEVKSTSERFLQDLEQ RLEADVLRFSVCDVVLDHCPAFRRVYLPYVTNQAYQERTYQRLLLENPRFPGILARLEESPVCQRLPLT SFLILPFQRI TRLKMLVENILKRTAQGSEDEDMATKAFNALKELVQECNASVQSMKRTEELIHLSKKIH FEGKIFPLISQARWLVRHGELVELAPLPAAPPAKLKLSSKAVYLHLFNDCLLLSRRKELGKFAVFVHAK 0 MAELQVRDLSLKLQGIPGHVFLLQLLHGQHMKHQFLLRARTESEKQRWISALCPSSPQEDKEVISEGED CPQVQCVRTYKALHPDELTLEKTDILSVRTWTSDGWLEGVRLADGEKGWVPQAYVEEISSLSARLRNLR ENKRVTSATSKLGEAPV In a search of sequence databases, it was found, for example, that the nucleic acid 5 sequence has 1120 of 1772 bases (63%) identical to human guanine nucleotide exchange factor Rac-GEF cDNA (patn:V99828) (SEQ ID NO: 36), as shown in Table 4C. The terms "percent identities" and "percent positives" are defined below in the Nucleic Acid section. Table 4C. BLASTN identity search of FCTR4 and the hGEF cDNA (SEQ ID NO: 36). >patn:V99828 Human guanine nucleotide exchange factor Rac-GEF cDNA - Homo sapiens, 0 3171 bp. Score = 1856 (278.5 bits), Expect = 7.2e-78, P = 7.2e-78 Identities = 1120/1772 (63%), Positives = 1120/1772 (63%), Strand = Plus / Plus Query: 3042 ATGACCCGGCTGCACTCTTCTCTG-CGCCTGGGGCGGAATTCAGCAGCCCGGGCACT-CA 3099 5 1 1 1 ] i l 1 1 1 1 1 l I1 l 1 i li il1 1 1 Sbjct: 371 ATGAGCCC-CTG-AC-CTTGAATATCCCCTGGAGCAGAATGC--CT--CCTTGCAGAACA 423 Query: 3100 TCTCTGG-GTCAGGCACCGGAGCAGCCCGGGAAGGGAAAGCATCTGGAATGGAGGCTCG- 3157 II i ii I I I iI I iii I iI I i I I I i i I I Ii 0 Sbjct: 424 GCAATGCAGACAGAC-CCAG-G-AGCCCAGGAAATGAGTGAGTC-GTCCTCCACCCCGGG 479 Query: 3158 AAGTGTAGAGATGAGCGGGGACCGGGTGTCGC-GGCC-A--GCCCCTGGTGACTCAC-GA 3212 10 WO 01/46231 PCT/USOO/34898 I i 1i 1 1 11 1 I I l11 1 1111 1 11111 i 11 Sbjct: 480 AAATGGGGCCACGCCCGAGGAGTGGCCGGCCCTGGCCGACAGCCCCACCACGCTCACCGA 539 Query: 3213 GAGG--GCGATTGGTCCGAGCCCAGGCTAGAC-ACACAGGAAGAGCCGCCTTTGGGGTCC 3269 5 1 11 1111 1 1 1 1 1 I 1 11 1 1 1 Sbjct: 540 GGCCCTGCGGATGATCC-ACCCCATTCCCGCCGACTCCTGGAGAAACCTCATTGAACAAA 598 Query: 3270 -AGGAGCACCA--ACGAGCGGCGCC-AG-TC--TCGATTCCTCCTT-AACTCCGTCCTCT 3321 111 11 11 I I 1 11 11 1 II l I 1 1 11Iii I I 0 Sbjct: 599 TAGG-GCTCCTGTATCAGGAATACCGAGATAAATCGACTC-TCCAAGAAATCGAAAC-C- 654 Query: 3322 ATCAGGAATACAGCGACGTGGCCAGCGCCCGCGA-ACTGCGGCGGCAGCAGCGCGAG-GA 3379 I 111 1I I l III iii III 1111111 Sbjct: 655 AGGAGGCA-ACAG-GATGCAGAAATAGAAGACAATACCAATGGGTCCCCGGC-C-AGTGA 710 5 Query: 3380 GGAGGGCCCGGGGGACGAGGCCGA-G-GGCGCAG-AGGAGGGGCCGGG--GCCGCCGCGG 3434 III IIlII 11I1 I I Ii l Il I lI l 1 Hll III I I Sbjct: 711 GGACACCCCGGAGGAGGAAGAAGAAGAGGAGGAGGAGGAGGAGCCGGCCAGCCCACCAGA 770 0 Query: 3435 GCCAACCTCTCCCCCAGCAGCTCCTTCCGGGCGCAGCGCTCGGCGCGAGGCTCCACCTTC 3494 I I 11111 III III II II l ii i I i I I I Sbjct: 771 GAGGAAGACTCTGCCC-CAGATC-TGCCTG-CTCAGTAACCC-C-C-A--CTCAAGGTTC 822 Query: 3495 TCGCTGTGGCAGGATATCCCCGACGTACGCGGCAGCGGCGTCCTGGCCACGCTG-AGCCT 3553 5 11 1111111 H i l 1 1 H I 1 11II 1 1 1 11111 Sbjct: 823 AACCTCTGGCAGGATCTTCCCGAGATCCGGAGCAGCGGGGTGCTTGAGATCCTACAGCCT 882 Query: 3554 GCGG-GACTGCAAGCTGCAGGAGGCCAAGTTTGAGCTGATCACCTCCGAGGCCTCCTACA 3612 1 1 1 1 1 1 I 1 111111 II M 0 Sbjct: 883 GAGGAGATT--AAGCTGCAGGAGGCCATGTTCGAGCTGGTCACTTCCGAGGCGTCCTACT 940 Query: 3613 TCCACAGCCTGTCGGTGGCT-GTGGGCCACTTCTTAG-GCTCTGCCGAGCTGAGCGAGTG 3670 1 1 11 111 1 11 111 11 1 1 1 1 1 1 lI 1 1 1 1 1 1 1 1 Sbjct: 941 ACAAGAGTCTGAACCTG-CTCGTGTCCCACTTCATGGAGAACGAGCG-GATAAGGAAGAT 998 5 Query: 3671 TCTGGGGGCGCAGGACAAGCAGTGGCTGTTTTCCAAACTGCCCGAGGTCAAGAGCAC-CA 3729 IIi I I I 111 I I I Ill 1 1111 II I I I Sbjct: 999 CCTGCACCCGTCCGAGGCGCACATCCTCTTCTCCAACGTCCTGGACGTGCTG-GCTGTCA 1057 0 Query: 3730 GCGAGAGGTTCCTGCAGGACCTGGAGCAGCGGCTGGAGGCAGATG-TGCTGCGCTTCAG- 3787 i ii i | 1 I i i 11 111111 I 1I IM 111 I I 1 I l 1 Sbjct: 1058 GTGAGCGGTTGGTCCTGGAGCTGGAGCACCGGATGGAGG-AGAACATGGTCATCT-CTGA 1115 Query: 3788 CGTGTGCGACGTGGTGCTGGACCACTGCCCGGCCTTCCGCAGA---GTCTACCTGCCCTA 3844 5 llli 1ll lil I II I llil H l II 1111 1111 Sbjct: 1116 CGTGTGTGACATCGTG-T--ACCGTTATGCGGCCGACCACTTCTCTGTCTACATCACCTA 1172 Query: 3845 TGTCACCAACCAGGCCTACCAGGAGCGCACCTACCAGCGCCTGCTCCTGGAGAACCCCAG 3904 i I I Ii I I III 11 l i i ll III 1111111 111111 I II 0 Sbjct: 1173 CGTCAGCAATCAGACCTACCAGGAGCGGACCTATAAGCAGCTGCTCCAGGAGAAGGC-AG 1231 Query: 3905 GTTCCCTGGCA-TCCTGGCTCGCCTGGAGGA-GTCTCCTGTGTGCCAGCGTCTGCCCCTT 3962 11 11 11 1 1 11 1 11 111 III i i i 1 1 i 1 Sbjct: 1232 CTTTCCGGGAGCTGATCGCGCAGCTAGAGCTCGACCCCAA-GTGCAGGGGGCTGCCCTTC 1290 5 Query: 3963 ACCTCCTTCCTTATCCTGCCCTTCCAGAGGATCACCCGCCTCAAGATGTTGGTGGAGAAC 4022 Sbjct: 1291 TCCTCCTTCCTCATCCTGCCTTTCCAGAGGATCACACGCCTCAAGCTGTTGGTCCAGAAC 1350 0 Query: 4023 ATCCTGAAGCGGACAGCACAGGGCTCTGAAGACGAAGA-CATGGCCACCAAGGCCTTCAA 4081 l I l I i I l I I i I I I l i I I I I l l I I I II I Sbjct: 1351 ATCCTGAAGAGGGTAGAAGAGAGGTCTGA-GCGGGAGTGCACTGCTTTGGATGCTCACAA 1409 Query: 4082 TGCGCTCAAGGAGCTGGTGCAGGAGTGCAATGCTAGTGTACAGTCCA-TGAAGAGGACAG 4140 5 1 111 1 1 11111 111 ill I 1 1l 1l 1 1111 1 11 1 Sbjct: 1410 GGAGCTGGAAATGGTGGTGAAGGCATGCAACGAGGGCGT-CAGGAAAATGAGCCGCACGG 1468 Query: 4141 AGGAACTCATCCACCTG-AGCAAGAAGATCCACTTTGAGGGCAAGATTTTCCCGCTGATC 4199 I I 1 1 1 11111111 IIII I III 111111 1 11 1 111 0 Sbjct: 1469 AACAGATGATCAGCATTCAG-AAGAAGATGGAGTTCAAGATCAAGTCGGTGCCCATCATC 1527 11 WO 01/46231 PCT/USOO/34898 Query: 4200 TCTCAGGCCCGCTGGCTGGTTCGGCATGGAGAGTTG--GTAGA-G-CTGGCACCACTGCC 4255 1i i i 11 [Ii1I Ii I 1 I I 1111 II I I I Sbjct: 1528 TCCCACTCCCGCTGGCTGCTGAAGCAGGGTGAGCTGCAGCAGATGTCAGGCCCCAAGACC 1587 5 Query: 4256 TGCAGCACCCCCTGCCAAGCT-GAAGCTGTCCAGCAAGGCAGTCTACCTCCACCTCTTCA 4314 1 1 1 I 1 11 1 I I II I I lH I I I III I I I I I I | Sbjct: 1588 TCCCGGACCC--TGAGGACCAAGAAGCTCTTC--CACGAAATT-TACCTCTTCCTGTTCA 1642 0 Query: 4315 ATGAC-TGCTTGCTGCTCTCTCGGCGGAAGG-AGCTAGGGAAGTTTGCCGTTTTCGTC-C 4371 1 I l 1 1| 11 1111 I l i i 1 i 11 11 1 1 1 Sbjct: 1643 ACGACCTGCTGG-TGATCTGCCGGCAGATTCCAGG-AGACAAGTACCAGGTATTTGACTC 1700 Query: 4372 ATGCCAAGATGGCTGAGCTGCAG-GTGCGGGACCTGAGCCTGAAGCTGCAGGGC-ATCCC 4429 5 I l 1I l 11 illi i ill Il 1111 11 11 111111 1 Sbjct: 1701 A-GCTCCGCGGG--GA-CTGCTGCGTGTGG-AG--GAGC-TGGAGGACCAGGGCCAGACG 1752 Query: 4430 C-GGCCA-CGTGTTCCTCCTCCAGCTCCTCCACGGGCAGCACATGAA-GC-A--CCAGTT 4483 1 11111 1111111 III l I i 1 1 1 1l I I I 1 111 I 0 Sbjct: 1753 CTGGCCAACGTGTTCATCCTGCGGCTGCTGGA-GAAC-GCAGATGACCGGGAGGCCACCT 1810 Query: 4484 CC-TGCTGCGGGCCCGGACGGAAAGTGAGAAGCAGCGATGGATCTCAGCCTTGTGCCCCT 4542 1 1 1 1 1111II I II IIlIIII II I I 1 11ill Sbjct: 1811 ACATGCTAAAGGCGTCCTCTCAGAGTGAGATGAAGCGTTGGATGACCTCACTG-GCCCC- 1868 5 Query: 4543 CCAGCCCCCAGGAGGAC-AAGGAGGT--CAT-CAG-TGAGGGG-GAAG-ATTGCCCCCAG 4595 l I l I l I i l l IiI I l II lii [ | l I I lllIIII I Sbjct: 1869 CAA----C-AGGAGGACCAAGTTTGTTTCGTTCACATCCCGGCTGCTGGACTGCCCCCAG 1923 0 Query: 4596 GTTCAGTGTGTTAGGACATACAAGGCACTGCACCCAGATGAGCTGACCTTGGAGAAGACT 4655 I Iiii iI II I i iii 111111 11 I I i I i I I I I I Sbjct: 1924 GTCCAGTGCGTGCACCCATACGTGGCTCAGCAGCCAGACGAGCTGACGCTGGAGCTCGCC 1983 Query: 4656 GACATCCTGT-CAGTGAGGACCTGGACCAGTGACGGCTGGCTGGAAGGGGTCCGCCTGGC 4714 5 1 1 1 1 1 1 | | i 1 l1 1 i I l l i l 1Il l I Il 1 1 1I li l I Sbjct: 1984 GACATCCTCAACATCCTGGACAAG-ACTGACGACGGGTGGATCTTTGGCGAGCGTCTG-C 2041 Query: 4715 A-GATGGTGAGAAGGGGTGGGTGCCCCAGGCCTATGTG-GAAGAGATCA-GCAGCCTCAG 4771 1 1 1i i I l I IIIIII I llI i H 111111 i i 1 0 Sbjct: 2042 ACGACCAGGAGAGAGGCTGGTT-CCCCAGCTCCATGACTGAGGAGATCTTGAATCCCAAG 2100 Query: 4772 CGCCCGCCTCCGAAACCTCCGGGAGAATAAGCGAGTC-ACAAG 4813 l l i I l I I I II I I I iH Sbjct: 2101 ATCCGGTCCCAGAA-CCTCAAGGAATGTTTCCGTGTCCACAAG 2142 5 FCTR4 has an even higher homology to a probable guanine nucleotide regulatory protein TIM (SEQ ID NO: 37; SWISSPROT-ACC:Q12774), as shown in Table 4D. The full amino acid sequence of the FCTR4 protein was found to have 276 of 517 residues (53%), identical to, and 355 of 517 residues (68%) positive with, the 519 amino acid residue human 0 probable guanine nucleotide regulatory protein TIM (oncogene TIM, P60 TIM, transforming immortalized mammary oncogene) from ptnr: SWISSPROT-ACC:Q12774. TIM has transforming activities in NIH/3T3 fibroblasts. See, e.g., Chan et al., 1994 Oncogene 9: 1057 1063. The 2.3-kb TIM cDNA encodes a predicted protein of 60-kD containing a Dbl homology (DH) domain. See, e.g., Online Mendelian Inheritance in Man database accession 5 number OMIM 600888. The DH motif is shared by several signal transducing molecules that are implicated as regulators of small GTP-binding proteins. See, OMIM 600888. Therefore, 12 WO 01/46231 PCT/USOO/34898 the TIM oncogene is also thought to be involved in the control of cytoskeletal organization through regulation of small GTP-binding proteins. See, e.g., Chan et al., 1994; OMIM 600888. Table 4D. BLASTX identity search of FCTR4 and hTIM protein (SEQ ID NO:37). >ptnr:SWISSPROT-ACC:Q12774 PROBABLE GUANINE NUCLEOTIDE REGULATORY PROTEIN TIM 5 (ONCOGENE TIM) (P60 TIM) (TRANSFORMING IMMORTALIZED MAMMARY ONCOGENE) - Homo sapiens (Human), 519 aa. Score = 1275 (448.8 bits), Expect = 3.5e-129, P = 3.5e-129 Identities = 276/517 (53%), Positives = 355/517 (68%), Frame = +3 0 Query: 3285 RRQSRFLLNS--VLYQEYSDVASARELRRQQREEEGPGDEAEGAEEGPGPPRANLSPSSS 3458 Il 1+ 1+11 +1111111 +I++ 1il i I I I i I I Sbjct: 6 RRCSK-LINSSQLLYQEYSDVVLNKEIQSQQRLESL--SETPGPSS-PRQPRKALVSSES 61 Query: 3459 FRAQRSARGSTFSLWQDIPDVRGSGVLATLSLRDCKLQEAKFELITSEASYIHSLSVAVG 3638 5 + Ii + 1+ 1111+11 i1I I + I I liii 1111+ 1I ++Il Sbjct: 62 Y-LQRLSMASSGSLWQEIPVVRNSTVLLSMTHEDQKLQEVKFELIVSEASYLRSLNIAVD 120 Query: 3639 HFLGSAELSECLGAQDKQWLFSKLPEVKSTSERFLQDLEQRLEADVLRFSVCDVVLDHCP 3818 S ( I I 1 1+ 11111+1 +1+ I I 111+ 1 ++ I 11111+1 1 0 Sbjct: 121 HFQLSTSLRATLSNQEHQWLFSRLQDVRDVSATFLSDLEENFENNIFSFQVCDVVLNHAP 180 Query: 3819 AFRRVYLPYVTNQAYQERTYQRLLLENPRFPGILARLEESPVCQRLPLTSFLILPFQRIT 3998 l l I l I l l i l l l + 1 1+ 1 1 + 1 + 1 1 1 1 1 1 1 1 1 i l 1 1 1 Sbjct: 181 DFRRVYLPYVTNQTYQERTFQSLMNSNSNFREVLEKLESDPVCQRLSLKSFLILPFQRIT 240 5 Query: 3999 RLKMLVENILKRTAQGSEDEDMATKAFNALKELVQECNASVQSMKRTEELIHLSKKIHFE 4178 lil+I++IIIII 11 +1 lIl +11++]H+++11 +1111+111111+1I+11 11 Sbjct: 241 RLKLLLQNILKRTQPGSSEEAEATKAHHALEQLIRDCNNNVQSMRRTEELIYLSQKIEFE 300 0 Query: 4179 GKIFPLISQARWLVRHGELVELAPLPAAPPAKLKLSSKAVYLHLFNDCLLLSRRKELGKF 4358 1II 11111+ 1111+ li1 I 1+1 + 11++ + 1+ 1 1111111111 + 1 + 1 Sbjct: 301 CKIFPLISQSRWLVKSGELTALE-FSASPGLRRKLNTRPVHLHLFNDCLLLSRPREGSRF 359 Query: 4359 AVFVHAKMAELQVRDLSLKLQGIPGHVFLLQLLHG-QHMKHQFLLRARTESEKQRWISAL 4535 5 li 11 + ++ +11 i ++1 1 i I + +1l I 1+111 1111 1 Sbjct: 360 LVFDHAPFSSIRGEKCEMKLHGPHKNLFRLFLRQNTQGAQAEFLFRTETQSEKLRWISAL 419 Query: 4536 CPSSPQEDKEVISEGEDCPQVQCVRTYKALHPDELTLEKTDILSVRTWTSDGWLEGVRLA 4715 + 1+1+ +++ I + 11111+1 11 111 III -t-+ 1 +IIIIHMI+ 0 Sbjct: 420 --AMPREELDLL-ECYNSPQVQCLRAYKPRENDELALEKADVVMVTQQSSDGWLEGVRLS 476 Query: 4716 DGEKGWVPQAYVEEISSLSARLRNLRENKRVTSATSKLGE 4835 ((1+11 1 11 11+ 1 +11+1 1I +1 +1 1 Sbjct: 477 DGERGWFPVQQVEFISNPEVRAQNLKEAHRVKTAKLQLVE 516 5 A multiple sequence alignment for AL109627_A is given in Table 4E, with the FCTR4 protein of the invention being shown on line 2, in a ClustalW analysis comparing the protein of the invention with related protein sequences. Table 4E depicts a ClustalW alignment of the FCTR4 against proteins from a public database. Human oncogene p60 TIM (SEQ ID NO:37; 0 GenBank Acc. No. Q12774) is on line one, FCTR4 (SEQ ID NO:8) is on line two, and an unknown human polypeptide (SEQ ID NO:38; Acc. No. Q99434) is on line three. Based on this alignment, black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); grayed amino acid residues can be mutated to a residue with comparable steric and/or chemical 5 properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted 13 WO 01/46231 PCT/USOO/34898 amino acid residues can potentially be mutated to a much broader extent without altering structure or function. TABLE 4E TIM HUMAN GGFSC<L LNSRQL L N iQSMQI L S-LSOTPIPSS-IRQKA VS 5 AL109627 STNEIQ 7FLLNI-V ASAn RQ E EGPGDRAEIAEEG1GP ANJSP Q99434_Human TIM HUMAN lE-SYI 2ESMA ,GMMW( V NIT IITHEEQMIZEEAS3 V R NI AL109627 RSSFRAESARG FIWD :DGIGGATLSLR C S 1 V 0 Q99434_Human ------------------------------- ---------- M' LW9ES=Q3[ TIM HUMAN EDMQLHTS SNBMQ E DVHATEENFHNIFSHQ N AL109627 E * LEDWR§SD* Q99434_Human LBEEEQHK VTQMEHHN ILELGAHQSFEI HKQVEDLSIILE' 5 TIM HUMAN D T1S IMN NNVIEI S S AL109627 M*. YRIJLEMP RHP 3IZEST Q99434 Human O o TIM HUMAN _AL109627 H Q 99434 Human P1KM~jJ= JJF.J-- TjO A TRFJ 1VK TIM_ HUMAN TGS QPFSSIRGECHM HEPHK --------- NIRJIFRQTA AE F ALlL09627 ELG Q K MAE fRDSLEQII31 G---------H ILQHG-HMKH o Q99434_Human SEED RV Y QLYVNHIQPSELP GGNRSSSVPPQSTENRESESEFEILPFS TIM HUMAN WTEQLMMAMPREEL --- LCYNS 0 PAENjAM ET AL109627 ARKEQPC li (PEIKEV IE E ATlHpTG ISIR Q99434 Human PSD UT H*RKA 55 L SK ] C GA QQYTHT TIM HUMAN -C KIFLIQSRELVA QEKIAHEKrA -FQS- E HLFDSQQA AL109627 H - G H E ALAALRC SK- rYLHLNDCLLLSR Q99434 Human KV-E YSERT ED L RVVEG RMEL TEDV --- T A From these analyses, it is seen that the FCTR4 AL109627A nucleic acid and protein are similar to the TIM oncogene. The transforming gene, designated TIM, encoded a predicted protein species of 60 kDa containing a Dbl-Homology (DH) motif. This motif is also present in other growth regulatory molecules including B~cr, Cdc24, Vav, Ras-grf, and Ect2 which have been implicated as regulators of small GTP-binding proteins. NJH3T3 cells transfected 5 with TIM expression plasmid showed altered growth properties in vitro and were tumorigenic when injected into nude mice. The 6.5 kilobasepair (kb) transcript of the TIM\ gene was found to be expressed mainly in kidney, liver, pancreas, lung, and placenta. Table 4F:BLAST alignment of FCTR4 BLAST alignment file included sequences: o Line 2 > gilll42O36l1reflXP_004812.11 Oncogene TIM [Homo sapiens] (SEQ ID NO:39) Line 3 > gil48856331refIN9_005426.11 Oncogene TIM [Homo sapiens] (SEQ ID NO: 40) Line 4 > gi198452771ref1NP_063920.11 neuronal guanine nucleotide exchange 5 factor [Mus musculus] (SEQ ID NO:41) 14 WO 01/46231 PCT/USOO/34898 1030 1040 1050 1060 1070 1080 5 FCTR4 ALISGSGTGAAREGKASGMEARSVEMSGDRVSR"G SREGDWSEP DT IEPPLGSR Line 2 -------------------- EGSSDSRGPAVEKH G S-------TVFKKPKEVk Line 3 ----------------------------------------------------------- M Line 4 ------------------------------ MIH5IAjSWRNLIEQIG LYQYRDKST o 1090 1100 1110 1120 1130 1140 FCTR4 STNE QjRF LN- S ISZ L RS2QIE EGPGDIAIAEEG.GP N SP Line 2 GGFSCKLDIS LN SSQLS--LS TPPS- RQ S Line 3 GGFS CKLINQS LN SQIL S--LS TP P S- RQ S Line 4 QEIET ----- RQQDAEISGN DSQVG DAG EE EEEGE EEL P--PE PQ 1150 1160 1170 1180 1190 1200 FCTR4 SSF SESRG P D TiLSLRC T o Line 2 *-SYLSL6 SG T i Line 3 I-SYLS SG SEMT Line 4 IC --- LLSNPH RFN DL E GS DILQP E) 1 Cons M I e 5 1210 1220 1230 1240 1250 1260 FCTR4 H LGIAEISEC L P ST E QPRL DVLR S Line 2 QL TSRT SN H Q D T SQ ENF F eI il Q IN [F PS Line 3 QL TS T SN H Q S,,D T SQ Line 4 ENER KI EPS I RL ER LE E ISD IR 1270 1280 1290 1300 1310 1320 FCTR4 HCPY YER LE E TSLESIL 5 Line 2 HD YYN Q S KSFLILPFQ Line 3 HPF RV Y LPYVTNTYQER S KSFLILPFQ Line 4 ANDHFSVYTYS Y QE PSA LS'K GPS 1330 1340 1350 1360 1370 1380 FCTR4 RIR QLEK DNIK FE Line 2 RITRLKLLRDILKRTP Line 3 RI LL QL KIRRD N Line 4 RIRE Q RE EE TMVKE E QS 1390 1400 1410 1420 1430 1440 FCTR4 GKIFPISWLVH PLP PA S LHF LRR Line 2 CS T FS- G lI LL P 0 Line 3 C S FS- GH I HH I Line 4 IKSVISHSRW'Q QQSGPK IRTT KLFE Y F L LCRQI 1450 1460 1470 1480 1490 1500 5 FCTR4 G KME LVRDLS IPGEN V HG-1HMH L I Line 2 S 1 L FSSI C H E RFRTGAJEFTES L Line 3 S L FSPHILI F R T GA IEF[ E Line 4 D RGL L Q-TL I L DDR L S O 0 1510 1520 1530 1540 1550 1560 FCTR4 CPSSPQEDKEVIS GEC ..... T LHPT TISC RTWSDWE Line 2 P---RE LD LCYN1S VMT|QQS Line 3 P--- EELDLL CY}T NS TQQ SS L 5 Line 4 TS PNRRTKFVSFTSRLLDC H P L LIIEDGWGE 1570 1580 1590 1600 1610 FCR4 ' -QAYIE 6§,L ~ jTATS GJPV ---------- Lin 2 STQ F KQA----------- 15 WO 01/46231 PCT/USOO/34898 Line 3 IS QQ ' *F VIAAH"KVI2LQUVRQA---------- Line 4 IQE SSMT E L "S C " HKMDPQRSVNKDRRKLGSRNRQ FCTR4 was found to have high homology to the domains shown in Table 4G. 5 Table 4G: CD domain analysis of FCTR4 Sequences producing significant alignments: bits) value Guanine nucleotide exchange factor for Rho/Rac/Cdc42-like GTPa... 110 7e-25 RhoGEF, RhoGEF domain 69.3 le-12 ras, Ras family 61.2 4e-10 Rab subfamily of small GTPases; Rab GTPases are implicated in ... 51.2 4e-07 SH3, SH3 domain 49.7 le-06 Rho (Ras homology) subfamily of Ras-like small GTPases; Member... 44.7 4e-05 Src homology 3 domains; Src homology 3 (SH3) domains bind to t... 43.9 6e-05 Ras subfamily of RAS small GTPases; Similar in fold and functi... 38.9 0.002 arf, ADP-ribosylation factor family 38.1 0.003 The AL1 09627_A nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in various cancers, tumors and similar neoplastic diseases. 0 For example, a cDNA encoding the transforming immortalized mammary oncogene-like protein may be useful in gene therapy, and the transforming immortalized mammary oncogene-like protein may be useful when administered to a subject in need thereof. The novel nucleic acid encoding transforming immortalized mammary oncogene-like protein, and the transforming immortalized mammary oncogene-like protein of the invention, or fragments 5 thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These materials are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. FCTR5 (AL109913_A) 0 The novel FCTR5 nucleic acid encoding a C-terminal fragment of a novel FCTR5 protein is shown in Table 5A. This sequence contains no initiation codon. A TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The stop codon is shown in bold letters. This sequence originates in chromosome X, clone RP11-183K14, and is found at map location q26.3-27.3. 5 Table 5A. FCTR5 (AL109913_A) C-terminal nucleotide fragment (SEQ ID NO:9). natgatgatgagcaaaacatgatttcaatattgagcctggtgtctgtgaccattgctgtgttcatccca gttgcctgtgacagtcatgatcaacaagtctgcaccatgaccttctcatctccatatccagtgcccaag ttattcctttccccaactgcaggccccccaacaggatgtgggcagcctgcatctccgctggactggagc caaaatgccaaagcacagcaccttcgagttccatgcctccagaagggcttgtccctgcgcactgggatg 16 WO 01/46231 PCT/USOO/34898 gtgcttgtttgcaaggttatagatgagaaaactgctgccttgtcggaaggaaaggtgctgtttggtctc ttcgctggcatccccatctttaggaattccagcccaaacaagccgccttccaattag The encoded C-terminal fragment of the encoded protein is presented using the one 5 letter code in Table 5B. The C-terminal fragment disclosed has a very high probability of being secreted extracellularly. A signal peptide most likely is cleaved between residues 28 and 29, i.e., at the dash in the amino acid sequence CDS-HDQ. Table 5B. Encoded FCTR5 polypeptide sequence (SEQ ID NO: 10). XDDEQNMISILSLVSVTIAVFIPVACDSHDQQVCTMTFSSPYPVPKLFLSPTAGPPTGCGQPASPLDWS 0 QNAKAQHLRVPCLQKGLSLRTGMVLVCKVIDEKTAALSEGKVLFGLFAGIPIFRNSSPNKPPSN In a search of sequence databases, no similarities were found to any currently disclosed nucleic acid or protein. The nucleic acids and proteins of the invention are potentially useful in the treatment 5 of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. FCTR6 (AL109928_A) A novel nucleic acid encoding a novel transmembrane protein is shown in Table 6A. It was identified in chromosome 20 clone RP4-551D2 at map location ql3.2-13.33. An 0 initiation codon is shown at the beginning of the sequence and a TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. These are shown in bold face in Table 6A. Table 6A. Nucleotide sequence (SEQ ID NO: 11) of FCTR6 (AL109928_A). atgagatccgggaggcacccctcgctgctgctgcttctagtgctgctgctgtggctgctgcaggtcagt 5 atcattgacagtgttcaacaggaaacagatgatcttactaagcaaacaaagtgtcactataagttccag gaaaagatctaccagcctctacggcgatccaagagaagatgggttatcaccaccttggagctggaggag gaagacccgggaccctttcccaaactcattggtgagctgttcaataatatgtcttataacatgtcacta atgtatctaatcagtggacctggtgtggatgaatatccagagattggtttgttttctctagaagatcat gagaacggaaggatatatgttcaccgccctgtcgatcgagaaatgacaccatctttcacgagctggaca 0 gcaagggtgccttcctccagggcttccgcggggatgagcagaggccatctacgggaagggctggtgctg gtttattttgatgttgtggagcgctcaacaggaaaaattgtggatacatccttgattttcaacattagg atcagtgatgtgaatgatcatgcaccccagtttccagagaaggaatttaacatcactgtgcaagaaaac caatctgcagggcaacctatttttcagatgttagcagtcgatttggatgaagaaaacactccaaattct caagtcctttacttcctcatttctcaaacaccattactgaaagaaagtggtttccgggttgatcgcctt 5 agtggagaaatacgactctctggctgcttagattatgagaccgctcctcagtttacactgctaatcaga gccagggactgtggagaaccgtcactgtcatccacgaccaccgttcacgtggatgtgcaagaaggcaac aaccacaggcctgcatttacccaggagaactataaggttcagattcctgaaggccgagccagccagggc gtgttgcgtctcctggttcaagatcgagattctccatttacatcagcttggagagcaaaattcaacata ttgcatggcaatgaagaggggcattttgacatttcgactgaccctgagaccaacgaagggatattaaat 0 gttatcaagcctttggattatgagactcgcccagcgcaaagcctcatcattgtcgtggagaatgaggag aggctcgtcttctgtgagagaggaaagcttcagccgccaaggaaggcagcagccagcgccactgtgagt gtgcaggtgacagacgccaacgacccaccagcctttcacccccagagcttcattgtcaataaagaggag ggcgccaggcctgggaccctgttgggaacttttaatgccatggatccagacagccagataagatatgaa 17 WO 01/46231 PCT/USOO/34898 ctggttcatgacccagcaaattgggtcagcgtcgacaaaaactccggagtggtcatcaccgtggagcca attgaccgagaatcccctcatgtaaataacagtttttatgtaatcatcattcacgctgttgatgatggc ttcccaccgcagactgctacagggaccctaatgctcttcctgtctgacatcaatgacaacgtcccgact ctccggccacgttcccgctacatggaggtctgtgagtctgctgtgcatgagcccctccacatcgaggca 5 gaggatccggacctggagccgttctctgacccatttacatttgaattggacaatacctggggaaatgcg gaggacacatggaagttggggagaaattggggaaactctcctcatcagggggtaggaggctgctgggag tccctgagacatattcttgcatctggcaagaagggtgtttccagggaagctccaggattgacgtcactg tttggcctgggtcaatcagttgaacttttaaccttgagaagcctgccacgtggtaattacttggtgcca ctcttcattggagacaaacagggactttcccagaagcaaactgtccatgtaaggatctgcccctgtgcc 0 agtgggctcacatgtgtggagcttgcagatgcagaagtggggcttcatgtgggggccctgttccctgtc tgtgcagcatttgtggctctggcagtggctctgctttttctgttgcgatgctattttgtgcttgaacct aagaggcatggatgctctgtatccaatgatgaaggccaccaaacactggtcatgtataatgcggagagc aaaggcacttcagcccagacatggtcagatgttgaaggccagaggccggctctgctcatctgcacagct gcagcaggacccacgcagggagttaaggggagggaaccaaagcctccaccttctaggttttggtgtatc 5 tctgggttcccttcagtgtcctgcaaatattgtagatctcgaggaagtgcctccatctgcagcgagtca gtcagcccaagcacgctgtgctctggggagctggatagcacagagacccagatccacagacatgggcca gatgagcaggagactgccagcagcccatcatgggaaacaatgggcagccctgcagaatgggtgctgcct ggcacctgcttcaagacaacacagacatcttctccgggcctagaagctttgcctaaaagcaggcaagcc aggctcctgcagaagggggctgtgtacccacagactcagggctgcagggcccttccccaggtcctgact 0 gctgaactggaaatggggctggaggacagagaaagaacagaggctcttggggaggctttcatggccagg ctggctgccgacctgaagggggactatctgcagagcttgggaagggaggcatccacagtggaatcctgt gttggaaggagccagagtccctcacactggcaggccaaaaaggcctggatccccaaacttttacaaaag agaaataaattcaacaacgtagcacctatagtcaacaacgtagcatctatagtcaacaacatagcacct atagtcaacaacgtagcacctatagtcaacaacgtagcatctatagtcaacaacgtagcacctatagtc 5 aacaacgtagcacctatagtcaacaacatagcacctatagtcaacaacgtagcatctatagtcaacaat gcacttcaacattttactttaagtgctaggatacatgtgcagaaggtgcagtctaaagagagaaatcgc ttcagcctcagcaggggctgcatcatcccccagggaagagccacagctgggcgaggattgccacaagac atttacaaggagatgatgccacggagactaacgcagactggtaaacggaaacacggggctttggctcga acaccctctttcaagaaagttgtttatgaccacaaggaagtgtctctcatctgttgggtacaaacatcc 0 ccagaagatcccccgccacacattccctggatcagaacccatcagtggttccctagtgcctgggaattt ccattcaatggcctccgaaccatgagcctgccttttctgcctgaagcccaaaaccccagctacagatct ttaccccagagaccatcttgggcctccctccaggcttttgcttactctgtgccctcatcctggagtcct gtccccacccctatctacagaaactccaccagccctcctggctgccccgatggtcctcgcacagggaga cttgtctacctcccgaggtcacgtgtgggctctggtcctcttgccatcatggcagagattttgctgtat 5 ctccccctggctgctggtgctctgcttacctcctccagagttgttaacaaagagctgaggatgctgagc tgcccagggacttggctgcaggtggcatag The encoded protein is presented using the one-letter code in Table 6B. The protein has a high probability of sorting into the plasma membrane. Cleavage of a signal peptide is 0 predicted to occur between residues 27 and 28, i.e., at the dash in the sequence IDS-VQQ. Table 6B. Encoded FCTR6 protein sequence (SEQ ID NO: 12). MRSGRHPSLLLLLVLLLWLLQVSIIDSVQQETDDLTKQTKCHYKFQEKIYQPLRRSKRRWVITTLELEE EDPGPFPKLIGELFNNMSYNMSLMYLISGPGVDEYPEIGLFSLEDHENGRIYVHRPVDREMTPSFTSWT ARVPSSRASAGMSRGHLREGLVLVYFDVVERSTGKIVDTSLIFNIRISDVNDHAPQFPEKEFNITVQEN 5 QSAGQPIFQMLAVDLDEENTPNSQVLYFLISQTPLLKESGFRVDRLSGEIRLSGCLDYETAPQFTLLIR ARDCGEPSLSSTTTVHVDVQEGNNHRPAFTQENYKVQ IPE GRASQGVLRLLVQDRDSPFTSAWRAKFNI LHGNEEGHFDI STDPETNEGILNVIKPLDYETRPAQSLI IVVENEERLVFCERGKLQPPRKAAASATVS VQVTDANDPPAFHPQS FIVNKEEGARPGTLLGTFNAMDPDSQIRYELVHDPANWVSVDKNSGVVI TVEP IDRESPHVNNSFYVI I IHAVDDGFPPQTATGTLMLFLSDINDNVPTLRPRSRYMEVCESAVHEPLHIEA 0 EDPDLEPFSDPFTFELDNTWGNAEDTWKLGRNWGNSPHQGVGGCWESLRHILASGKKGVSREAPGLTSL FGLGQSVELLTLRSLPRGNYLVPLFIGDKQGLSQKQTVHVRICPCASGLTCVELADAEVGLHVGALFPV CAAFVALAVALLFLLRCYFVLEPKRHGCSVSNDEGHQTLVMYNAESKGTSAQTWS DVEGQRPALLICTA AAGPTQGVKGREPKPPPSRFWCISGFPSVSCKYCRSRGSASICSESVSPSTLCSGELDSTETQIHRHGP 18 WO 01/46231 PCT/USOO/34898 DEQETAS S PSWE TMGS PAEWVL PGTCFKTTQTSS PGLEAL PKSRQARLLQKGAVYPQTQGCRAL PQVLT AELEMGLEDRERTEALGEAFMARLAADLKGDYLQSLGREASTVESCVGRSQS PSHWQAKKAWIPKLLQK RNKFNNVAPIVNNVASIVNNIAPIVNNVAPIVNNVAS IVNNVAPIVNNVAPIVNNIAPIVNNVASIVNN ALQHFTLSARIHVQKVQSKERNRFSLSRGCIIPQGRATAGRGLPQDIYKEMMPRRLTQTGKRKHGALAR 5 TPSFKKVVYDHKEVSLICWVQTSPEDPPPHIPWIRTHQWFPSAWEFPFNGLRTMSLPFLPEAQNPSYRS LPQRPSWASLQAFAYSVPS SWS PVPTPIYRNSTS PPGCPDGPRTGRLVYLPRSRVGSGPLAIMAEILLY LPLAAGALLT SSRVVNKELRML SCPGTWLQVA In a search of sequence databases, it was found, for example, that the nucleic acid 0 sequence has 225 of 381 bases (59%) identical to human cadherin-13 coding sequence (patn: :T85405) (Table 6C). Table 6C. BLASTN identity search of FCTR6 and hCAD-13 (SEQ ID NO:42). >patn:T85405 Human cadherin-13 coding sequence - Homo sapiens, 2690 bp. Score = 323 (48.5 bits), Expect = 6.4e-05, P = 6.4e-05 5 Identities = 225/381 (59%), Positives = 225/381 (59%), Strand = Plus / Plus Query: 804 TCCTCAGTTTACACTGCTAATCAGAG-C-CAGGGACTG--TGGA--GAACCGTC-ACTGT 856 III ilI il I I I ) i IlII 11 1111 11 I i i Sbjct: 1416 TCCCAAGTATGAACTGATCATC-GAGGCTCAAGATATGGCTGGACTGGATGTTGGATTAA 1474 0 Query: 857 CATCCACGACCACCGTTCACGTGGATGTGCAA-GAAGGCAACAACCACAGGCCTGCATTT 915 liI I lii [Ii i I I l II l i|I I i II i ii ii I I I Sbjct: 1475 CAGGCACGGCCACAGC-CACGATCATGATCGATGACAAAAATGATCACTCACCAAAATTC 1533 5 Query: 916 ACCCAGGAGAACTATAAGGTTCAGATTCCTGAAGGCCGAGCCAGCCAGGGCGTG-TTG-C 973 Sbjct: 1534 ACCAAGAAAGAGTTTCAAGC-CACAGTCGAGGAAG--GAGCT-GT--GGGAGTTATTGTC 1587 Query: 974 G-TCTCCTGGTTCAAGATCGAGATT-CTCCATTTACATCAGCTTGGAGAGCAAAATTCAA 1031 0 i ill 11111 1 M1 ill i 11 11111 1 I ii Sbjct: 1588 AATTTGACAGTTGAAGATAAGGATGACCCCACC-ACAGGTGCATGGAGGGCTGCCTACAC 1646 Query: 1032 CATATTGCATGGCAATGAAGAGGGGCATTTTGACATTTCGACTGACCCTGAGACCAACGA 1091 I II I I I lI I 1 1 1 I I 11 I 11 l I I iIIiI I II| 5 Sbjct: 1647 CATCATCAACGGAAACCCCGGGCAGAGCTTTGAAATCCACACCAACCCTCAAACCAACGA 1706 Query: 1092 AGGGATATTAAATGTTATCAAGCCTTTGGATTATGAGACT-CGCCCAGCGCAAAGCCTCA 1150 1111I I I I iiiiII I I I I IIII i i i 11 i I I i II i i Sbjct: 1707 AGGGATGCTTTCTGTTGTCAAACCATTGGACTATGAAATTTCTGCCTTC-CACACCCTGC 1765 0 Query: 1151 TCATTGTCGTGGAGAATGAGGAGAGGCTCGT 1181 Sbjct: 1766 TGATCAAAGTGGAAAATGAAGACCCACTCGT 1796 5 The full amino acid sequence of the protein was found to have 155 of 413 residues (37%), identical to, and 233 of 413 residues (56%) positive with, human neural-cadherin precursor (n-cadherin) having a total of 906 amino acid residues (SWISSPROT ACC:P19022) (Table 6D). Table 6D. BLASTX comparison of FCTR6 and human N-cadherin (SEQ ID NO:43). 0 >ptnr:SWISSPROT-ACC:P19022 NEURAL-CADHERIN PRECURSOR (N-CADHERIN) - Homo sapiens (Human), 906 aa. Score = 706 (248.5 bits), Expect = 1.3e-87, Sum P(3) = 1.3e-87 Identities = 155/413 (37%), Positives = 233/413 (56%), Frame = +1 5 Query: 514 GKIVDTSLIFNIRISDVNDHAPQFPEKEFNITVQENQSAGQPIFQMLAVDLDEENTPNSQ 693 I 1+ + I + 1+11+ 1+1 + +1 1I I I + + 1+1 1+ I 1 Sbjct: 244 GNQVENPIDIVINVIDMNDNRPEFLHQVWNGTVPEGSKPGTYVMTVTAIDADDPNALNGM 303 19 WO 01/46231 PCT/USOO/34898 Query: 694 VLYFLISQTPLLKESG-FRVDRLSGEI-RLSGCLDYETAPQFTLLIRARDC-GEPS--LS 858 + I I I I I +1+1 ++ II 1 1+11+1+1 I 1 1+ 1I Sbjct: 304 LRYRIVSQAPSTPSPNMFTINNETGDIITVAAGLDREKVQQYTLIIQATDMEGNPTYGLS 363 5 Query: 859 STTTVHVDVQEGNNHRPAFTQENYKVQIPEGRASQGVLRLLVQDRDSPFTSAWRAKFNIL 1038 +1 1 + I + I++ I II + ++II I I I 1 1+1 1 1 Il I + I Sbjct: 364 NTATAVITVTDVNDNPPEFTAMTFYGEVPENRVDIIVANLTVTDKDQPHTPAWNAVYRIS 423 Query: 1039 HGNEEGHFDISTDPETNEGILNVIKPLDYETRPAQSLIIVVENEERLVFCERGKLQPPRK 1218 0 1+ I I I lii +1+1++ 1+11+1+11 1 + 11+ i +1 11+ Sbjct: 424 GGDPTGRFAIQTDPNSNDGLVTVVKPIDFETNRMFVLTVAAENQVPLA---KGIQHPPQ- 479 Query: 1219 AAASATVSVQVTDANDPPAFHPQSFIVNKEEGARPGTLLGTFNAMDPD---- SQIRYELV 1386 ++11111 1 I 1+ 1 1 l++ 11 11+1 I1 I il III + 5 Sbjct: 480 --STATVSVTVIDVNENPYFAPNPKIIRQEEGLHAGTMLTTFTAQDPDRYMQQNIRYTKL 537 Query: 1387 HDPANWVSVDKNSGVVITVEPIDRESPHVNNSFYVIIIHAVDDGFPPQTATGTLMLFLSD 1566 11111+ +1 +1 + 1+ +11111+1 1+ 1 1 1+1 11 + li lI ++ I I Sbjct: 538 SDPANWLKIDPVNGQITTIAVLDRESPNVKNNIYNATFLASDNGIPPMSGTGTLQIYLLD 597 0 Query: 1567 INDNVPTLRPRSRYMEVCESAVHEPLHIEAEDPDLEPFSDPFTFELDNTWGNAEDTWKLG 1746 lli I1 + I+ I 11+ ++1 I I 1++1 + II 1+1 + + I + Sbjct: 598 INDNAPQVLPQEA--ETCETPDPNSINITALDYDIDPNAGPFAFDLPLSPVTIKRNWTIT 655 5 Query: 1747 R 1749 Sbjct: 656 R 656 A multiple sequence alignment for FCTR6 AL109928_A is given in Table 6E, with the 0 protein of the invention being shown on line 4, in a ClustalW analysis comparing the protein of the invention with related protein sequences. Table 6E. BLASTX comparison of FCTR6 and human pre-N-cadherin (SEQ ID NO:44). >ptnr:SWISSPROT-ACC:P19022 NEURAL-CADHERIN PRECURSOR (N-CADHERIN) - Homo sapiens (Human), 906 aa. 5 Score = 151 (53.2 bits), Expect = 1.3e-87, Sum P(3) = 1.3e-87 Identities = 31/82 (37%), Positives = 49/82 (59%), Frame = +1 Query: 157 LRRSKRRWVITTLELEEEDPGPFPKLIGELFNNMSYNMSLMYLISGPGVDEYPEIGLFSL 336 1+1 11I I + I I II + + + ++ 1+11 I ++11I 1+ 1 1+1 + 0 Sbjct: 154 LQRQKRDWVIPPINLPENSRGPFPQELVRIRSDRDKNLSLRYSVTGPGADQ-PPTGIFII 212 Query: 337 EDHENGRIYVHRPVDREMTPSF 402 +l++ 1 +1+111 I Sbjct: 213 NPI-SGQLSVTKPLDREQIARF 233 5 The FCTR6 nucleotide sequence has two regions (nucleotides 1315-1757 and 1875 2305) identical to (100%) the 1808 bp human cadherin-like protein VR20 mRNA (VR20) (GenBank AF169690). Table 6F shows a partial BlastN alignment of FCTR6 with VR20. Table 6G. BlastN alignment of FCTR6 nucleotide with VR20 mRNA (SEQ ID NO:45). 0 >Homo sapiens cadherin-like protein VR20 mRNA, partial cds (GenBank AF169690) 1270 1280 1290 1300 1310 1320 FCTR6 AACGACCCACCAGCCTTTCACCCCCAGAGCTTCATTGTCAATAAAGAGGAGGGCGCA 5 hVR20 ------------------------------------------------------- CA 1330 1340 1350 1360 1370 1380 FCTR6 ACAA AAT ATA AGACAGAGATAA A 0 hVR20 C CT GA A CAACAGCAGAAAA 20 WO 01/46231 PCT/USOO/34898 1390 1400 1410 1420. 1430 1440 FCTR6 CC A CaG AAAG ATA 5 hVR20 TAT ACAAAATG G AAAAACA ACAC 1450 1460 1470 1480 1490 1500 ....I ....|I ....|I ....I ....I ....I ....I ....I ....I ....I ....I ....I FCTR6 GACAATGTAAAAATAATATA 0 hVR20 AGAATACAGAACATAAAAACAG ATAATATA 1510 1520 1530 1540 1550 1560 ....I ....I ....I ....I ....I ....I ....I ....I ....I ....I ....I ....I FCTR6 CAC A A CAGC ACAGGAC 5 hVR20 T AATACAGAGAAGA A 1570 1580 1590 1600 1610 1620 ....I ....|I ....I ....I ....I ....Il ....I ....I ....I ....I ....I ....I FCTR6 TCTGACAT CAT A 1AAC ACACACATA 0 hVR20 ACAAATCA AAA C ACACACAT A 1630 1640 1650 1660 1670 1680 FCTR6 AGAGA CACATAGAGAGAT AC A 5 hVR20 GAGTCTGCTGTGCATGA CACAAGAGAGAGAC A 1690 1700 1710 1720 1730 1740 FCTR6 ACATACATAATACAATAC AAAT AGACACA 0 hVR20 ACATACATAATACAATAC AAAT AGACACA 1750 1760 1770 1780 1790 1800 FCTR6 T AGAAAT CTCTCCTCATCAGGGGGTAGGAGGCTGCTGGGAGTCCCTG 5 hVR20 TTGGGGAGAATGGGG- -------------------- 1810 1820 1830 1840 1850 1860 FCTR6 AGACATATTCTTGCATCTGGCAAGAAGGGTGTTTCCAGGGAAGCTCCAGGATTGACGTCA o hVR20 ----------- 1870 1880 1890 1900 1910 1920 ....I ....I ....I ....I ....I ....|I ....I ....I ....|I ....I ....I ....I FCTR6 CTGTTTGGCCTGGGCAATAGTAATAA AGAAG A 5 hVR20 --------------- TAACAGGAACTAAC TA AAG CACGA 1930 1940 1950 1960 1970 1980 FCTR6 AC AC ATAGACAAACAGAC AGAAGCA A 0 hVR20 AAC A TAGA AAACAGA CAAAAAAC A 1990 2000 2010 2020 2030 2040 FCTR6 A A AT A 5 hVR20 GTAAGATG C CAG TACAT GAGTAGAT A 2050 2060 2070 2080 2090 2100 FCTR6 GGGCTTCATGTGGGGGCCTGTTCCTTCTGTAGCATGA o hVR20 CTATACAT A 2110 2120 2130 2140 2150 2160 ....I ....I ....I ....I ....I ....I ....I ....I ....lI ....I ....I ....I FCTR6 CA ATAACAAGAGATATC 5 hVR20 ATATAAC AAAGATA A 2170 2180 2190 2200 2210 2220 FCTR6 TAATATAAGAAAACAC ATATAACAGAGAAAGGAC 0 hVR20 TAATGAT G AAAACAC AATAAT AAGAAAGA 21 WO 01/46231 PCT/USOO/34898 2230 2240 2250 2260 2270 2280 FCTR6 AGACAT AGAT AAG AGAGAT ACAG C 5 hVR20 AGACAT AGAT AAG AGAGAT ACAG C 2290 2300 2310 2320 2330 2340 FCTR6 GCAGGACCCACGCAGGGAGTTAAGGGGAGGG A AGC TCECCTTCTEGTTTTGG ... o hVR20 GAGACACAGA AAGCTT---iCGTGO AATGCAC'ACAACTC Table 6G shows a ClustalW alignment of FCTR6 with related proteins found in public databases. FCTR6 polypeptide is on line 5, human CAD2 (SEQ ID NO:46) is on line 1, bovine CAD2 (SEQ ID NO:47) is on line 2, mouse CAD2 (SEQ ID NO:48) is on line 3, and chicken 5 CAD2 (SEQ ID NO:49) is on line 4. Based on this alignment, black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); grayed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted amino acid residues can potentially be mutated 0 to a much broader extent without altering structure or function. Table 6G. ClustalW alignment including FCTR6 (AL109928_A) protein. CAD2 HUMAN ALRT ----- ILEL SVE NG IA SDHGA CAD2 BOVIN ---------------------------------- s3CTPDAV 5f LGQ CAD2_MOUSE CRA GRGT ----- ALI SVE 5G IA AiMMfPN >HGP 5 CAD2 CHICK M TPPR I PLALM Q A P K jCDM 3EDMHV PS GIQ AL109928_genscan2 MISG-----RHI --------------------- SMLLLLVLLLWLLQISIIESVQQETD CAD2 HUMAN DG YVRFP Sj-E AQKTE CAD2_BOVIN L F G S -HS KFIAQ 0 CAD2 MOUSE LNVKSN RKE ADK TYASPJ- Q 3 o*dIMIE.61iRlli CAD2 CHICK ER QSDEN Y FGG SS E G iE S] Q A!-PTEEVS PR, KEI E AL109928_genscan 2 DLTEQTKH--YMF EKIYQi--LRSKR--RVTfLE EE2DPGPjPKLIGELFNNMS CAD2 HUMAN QVAVL K T ESAMVFS CH QRQRWI 5 CAD2_BOVIN Q LK AR PVST N Q CAD2 MOUSE NjWQ RE1T8 PEI P RCL . I A CAD2 CHICK IN KI PEI AFGA E MDQKK DEIEWEQYEDES HKDWVIPPINLPENSRG AL109928 genscan 2 YNMS&vYLIS --- GPGD1YPE1G--ASLEDHENG------- IUHR VDR- MTPS 0 CAD2 HUMAN ?FQLVISDKNSRVTPAD GQS R F CAD2_BOVIN ?FQLRRSRKLLRMTPADPGI IQSK R : CAD2 MOUSE ?PEVISDRKLLRSTPADPGI I T L : CAD2 CHICK ?FPQELVRIRSDRD S LS ADPI NS V Q:S AL109928_genscan 2 - TSWTA2VP SM ---------- ASAEMSR---------- H ---- REGLVl4VY D 5 CAD2 HUMAN LAHAVINVPDIVMD ELQNT GS TY VAID' CAD2 BOVIN LAAVINEIVVMDPL WG ESKT VAID CAD2 MOUSE LAAVINVPDIIM RELQ G PG GYTAIDA CAD2_CHICK * NQEIVNDDRFHWTPSGY VAIA o AL109928 genscan 2 VVERST--- jKL PTSLIFN SEVHAE PEKE I QSAMQP FQOLMV!L CAD2 HUMAN l. . SQ AS TSP N MFT G AAGRE IAT CAD2 BOVIN 3DPNA LN GMLRYRILAP ST 3 PN IEIITAAGL D RE KVQYTL AT 3 CAD2 MOUSE 5 CAD2 CHICK 3DPNA Q AL109928genscan_2 lETPMSQVL F i TELLK-ESGJRVDRLAPER- SNCJY TAP F LBR R C 22 WO 01/46231 PCT/USOO/34898 CAD2_HUMAN 3 II 0 CAD2_HOVIN 3 35 5333 CAD2 MOUSE .5 S30 CAD2_CHICK i*mm 5 AL109928 gens can 2 -EM -ISTVVJ GNPAQNKQTGAQj~jJMla CAD2 HUMAN 5.*-eem~e= m RQ I =A" CAD2 BOVIN =A ' 3 CAD2 MOUSE L3 3 S 5 o CAD2_CHICK =RflQMTeQTlL I" = ALlO 992 8genscan_2 E~AKFB~N-EIDOEMilNloIMPQI'VEEREC CAD2 HUMANs -- e *03 CAD2_BOVIN @- 5*0I 5 CAD2_MOUSE *--** Ljj CAD2 CHICK M0=01 --- BISVLV=LM~LSNfiiR nio AU 09928_gens can_2 PKQIKAA QTPDPHFIVNKARPILGNiM-- CAD2_HUMAN66-3 3 * 35 KI o CAD2 BOVIN 6 " 3 CAD2_MOUSE Io I OWN OR Sa 10 UPQ I3n 0aD; CAD2 CHICK 0 " 3 e *3 IIE AL109928_genscan_2 -SoMELVHNVSVKNSVVIiVEP3JHNSFVIIH.FVDFSQ t~ 5 CAD2_HUMAN* -- . i33 CAD2_BOVIN S IS -7O*3300339 CAD2_MOUSE .DIWTr --- 6 -E 3 CAD2 CHICK 03 6IKI--TLQMIAVM FI-eE ALl09928 gens can_2 BEMLSIZN:RRRMElSAHPH~nE~i EFSDMETEUUNTW 0 'I CAD2 HUMAN ----- -- -- - - - - - - -I U I D93 - - - --i CAD2_HOVIN -- iiw.. --------------------- 3 mawam--- CAD2_-MOUSE ----- ----------------- IN Rg3eT ff0* --- CAD2 CHICK --- DHP S------------------------------- IVELSEHS--------- j 5 AL109 92 8_genscan 2 GNAEDTW KGMGNSPHQGVGGCWESLRHILASGKKGVSMEAPGLTSIFGLGQSVELIT CAD2_HUMAN *qr"V~f~~ * i I CAD2 BOVIN S.e i CAD2 MOUSE .- 3 3 o CAD2_CHICK 3 3 ALl 0992 8genscan_2 L-SPIEMF~K-GIKTHIMLJtLEM~VJLP CAD2_HUMAN -- -- 3 ------- CAD2_HOVIN -5-- m *'* ' ---- ------- 5 CAD2_MOUSE -- 53 *' ---- ------- CAD2 CHICK - R 0 4 1 k3 13----ANT1 1a ----------- ALl 09928_genscan_2 CA~~AA ILRYLPIGSSJGSLMNEKTATSIG CAD2_HUMAN ---- B0 e ------ r l *~ o CAD2_BOVIN ---- ome 003 ----- om mfl CAD2__MOUSE 30 a 035 kiIao5IP31----s alw---am I----- m CAD2 CHICK --.-- 0r --03---- AL109928genscan 2 NPAl ICTAA, PTQGVKHPIPKPPPSRF.WC *GFPSVSCKYCRSRGSSICSZS, SIST 5 CAD2 HUMAN -M- . . .~" CAD2_BOVIN --a---NRIPT- -----*S5 3 *33 CAD2_MOUSE -u- - '' CAD2_CHICK --..... ' "' AL109928 genscan_2 , SEETTBRGDQTSEWT]S~tVLICKTTILAI CAD2_HUMAN - - -* 3 CAD2_BOVIN RR --. Te--- Polk,~ CAD2_MOUSE-- - -i3 3 . 3 3 CAD2_CHICK N- - *' ~ 5 5 AL109928_genscan 2 SRALQIVPEIRLQIALMLZ .ZTAMAIALALEY 23 WO 01/46231 PCT/USOO/34898 CAD2 HUMAN -------- CAD2 BOVIN -----------------
----
CAD2_MOUSE ------------- ------------ CAD2 CHICK ------------- - -- 5 AL109928 genscan 2 QSLGREASTVESCVGRSQSPSHWQAKKAWIPKLLQKRNKFNNVAPIVNNVASIVNNIAPI CAD2 HUMAN -------- ------- CAD2 BOVIN ------------ CAD2 MOUSE ---------------- 0 CAD2_CHICK ---- ----------- AL109928_genscan_2 VNNVAPIVNNVASIVNNVAPIVNNVAPIVNNIAPIVNNVASIVNNALQHFTLSARIHVQK CAD2 HUMAN -------------------- CAD2 BOVIN ----------------- 5 CAD2~MOUSE ------- -------- CAD2 CHICK ---------------- AL109928_genscan_2 VQSKERNRFSLSRGCIIPQGRATAGRGLPQDIYKEMMPRRLTQTGKRKHGALARTPSFKK CAD2 HUMAN --------------------------------------------------------- 0 CAD2 BOVIN -------------------------------------------- CAD2 MOUSE ------------------
--
CAD2 CHICK ------------ ---- --- --- AL109,928_genscan_2 VVYDHKEVSLICWVQTSPEDPPPHIPWIRTHQWFPSAWEFPFNGLRTMSLPGLPEAQNPS 5 CAD2 HUMAN ------- --- -- CAD2 BOVIN ------------- CAD2 MOUSE -------------------- CAD2 CHICK ------------------
-------
AL109928Bgenscan2 YRSLPQRPSWASLQAFAYSVPSSWSPVPTPIYRNSTSPPGCPDGPRTGRLVYLPRSRVGS CAD2 HUMAN -------- CAD2 BOVIN ----- CAD2 MOUSE ------------ CAD2 CHICK ---------- 5 AL109928_genscan_2 GPLAIMAEILLYPLAAGALLTSSRVVNKELRMLSCPGTWLQVA From these analyses, it is seen that the FCTR6 AL109928_A nucleic acid and protein a weak resemblance to neural cadherin, and a strong resemblance across a portion of FCTR6 with human cadherin-like VR20. Cadherins are calcium dependent cell adhesion proteins. 0 They preferentially interact with themselves in a homophilic manner in connecting cells; cadherins may thus contribute to the sorting of heterogeneous cell types. N-cadherin may be involved in neuronal recognition mechanism. They are type I membrane proteins. Finally, FCTR6 was found to have high homology to the domains shown in Table 6H. Table 6H: CD domain analysis of FCTR4 Score E Sequences producing significant alignments: (bits) value cadherin, Cadherin domain 73.9 5e-14 cadherin, Cadherin domain 57.0 6e-09 cadherin, Cadherin domain 44.3 4e-05 cadherin, Cadherin domain 40.4 6e-04 Cadherin repeats.; Cadherins are glycoproteins involved in Ca2... 56.6 8e-09 Cadherin repeats.; Cadherins are glycoproteins involved in Ca2... 49.3 le-06 5 24 WO 01/46231 PCT/USOO/34898 The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. FCTR7 (AL109953_A) 5 The novel FCTR7 nucleic acid encoding a novel secreted FCTR7 protein is shown in Table 7A1. This sequence contains an initiation codon at the 5' end, and a TGA stop codon was identified at the 3' end indicating that this sequence is a coding sequence. An alternative novel FCTR7A nucleic acid encoding a novel secreted protein is shown in Table 7A2. This sequence contains an initiation codon at the 5' end, a frameshift at position 61, and a TAA 0 stop codon indicating that this sequence is a coding sequence. The start and stop codons for both sequences are shown in bold letters. These sequences originate in chromosome 20 clone RP4-746H2. Table 7A1. FCTR7 (AL109953_A) nucleotide sequence (SEQ ID NO:13). atgggatgcagactgctgaccctgctgtgtttcctacaacctgcttccagctcctcgtggctctttggc 5 tcccaatccagagctttcgcgaacaccagagcccctgtgcctctccctgcagctggctgggagttccag ggcattaacacagacagtctttgcccatcagccagtgactgtatggagcttggatgtgaatacacagct cctgcatccctccgaggcatctccacaccgtctcccagagaatgtctcgtaaaagctgctcctcttggg gaggctctgggctttggagagagcacctggaattccccactagaaaagcccaaaaactga 0 Table 7A2. Alternative FCTR7A (AL109953_A) nucleotide sequence (SEQ ID NO:29). atgggatgcagactgctgaccctgctgtgtttcctacaacctgcttccagctcctcgtggtctttggct cccaatccagagctttcgcgaacaccagagcccctgtgcctctccctgcagctggctgggagttccagg gcattaacacagacagtctttgcccatcagccagtgactgtatggagcttggatgtgaatacacagctc ctgcatccctccgaggcatctccacaccgtctcccagagaatgtctcgtaaaagctgctcctcttgggg 5 aggctctgggctttggagagagcacctggaattccccactagaaaagcccaaaaactga The encoded FCTR7 protein is presented using the one-letter code in Table 7B 1. The FCTR7 protein has a low probability of being secreted extracellularly, although a signal peptide most likely is cleaved between residues 17 and 18, i.e. at the dash in the sequence 0 ASS-SSW. The encoded FCTR7A protein is presented using the one-letter code in Table 7B2. Table 7B. FCTR7 protein sequence (SEQ ID NO:14) encoded by SEQ ID NO:13. MGCRLLTLLCFLQPAS S S SWLFGSQSRAFANTRAPVPLPAAGWEFQGINTDSLCPSASDCMELGCEYTA PASLRGISTPS PRECLVKAAPLGEALGFGESTWNSPLEKPKN 5 Table 7B. FCTR7A protein sequence (SEQ ID NO:30) encoded by SEQ ID NO:29. MGCRLLTLLCFLQPAS S S SWSLAPNPELSRTPEPLCLSLQLAGS SRALTQTVFAHQPVTVWSLDVNTQL LHPSEASPHRLPENVS 25 WO 01/46231 PCT/USOO/34898 In a search of sequence databases, no similarities were found to known nucleic acid or protein. The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune 5 disorders, and hematopoietic disorders. FCTR8 (AL110115_A) The novel nucleic acid encoding a novel secreted protein is shown in Table 8A. This sequence contains an initiation codon at the 5' end, and a TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. This sequence originates in 0 chromosome 20 clone RP3-324017. Table 8A. FCTR8 (ALl 10115 A) nucleotide sequence (SEQ ID NO:15). atgaagctccttcttctgcttttgactgttactctgctcctggcccaggtcaccccaggtctgccagcc atgaaacttctttacctgtttcttgccatccttctggccatagaagaaccagtgatatcagtagagtgt tggatggatggacactgccggttgttgtgcaaagatggtgaagacagcatcatacgctgccgaaatcgt 5 aaacggtgctgtgttcctagtcgttatttaacaatccaaccagtaacaattcatggaatccttggctgg accactcctcagatgtccacaacagctccaaaaatgaagacaaatataactaatagatag The encoded protein is presented using the one-letter code in Table 8B. The protein has a moderate probability of sorting to the plasma membrane. A signal peptide most likely is 0 cleaved between residues 43 and 44, i.e. at the dash in the sequence VIS-VEC. Table 8B. Encoded FCTR8 protein sequence (SEQ ID NO:16). MKLLLLLLTVTLLLAQVTPGLPAMKLLYLFLAILLAIEEPVISVECWMDGHCRLLCKDGEDSIIRCRNR KRCCVPSRYLTIQPVTIHGILGWTTPQMSTTAPKMKTNITNR 5 In a search of sequence databases, the BLASTN comparison revealed 91 of 129 bases (70%), out of a total of 413 bases, are identical to an unidentified human secreted protein. No similarities of significance were identified at the amino acid level. Table 8C. BLASTN of FCTR8 with (SEQ ID NO:50). Query: 28 ATGAAGCTCCTTCTTCTGCTTTTGACTGTTACT-CTGCTCCTGGCCCAGGTCACCCCAGG 86 0 1 1 1 1 1 1 1 1 1 1 1 1 I lIi 1 1 1 1 1 1 1 1 1 i 1 1 1 1 1 1 1 I l1 1 1i I 1i 1 1 1 1 1 Sbjct: 43 ATGAAGCTCCTTTTGCTGACTTTGACTGTG-CTGCTGCTCTTATCCCAGCTGACTCCAGG 101 Query: 87 TCTGCCAGCCATGAAACTTCTTTACCTGTTTCTTGCCA-T-CC-TT-CTG--GC-CATAG 139 I || 1 II lI 1 I IIII II ilI I III l i I II I iI 5 Sbjct: 102 TG-GC-ACCCAA-AGA-TGCTGGAA-TCTTTATGGCAAATGCCGTTACAGATGCTCCAAG 156 Query: 140 AAG-AACCAGTGATAT 154 I l l I l I l i l Sbjct: 157 AAGGAAAGAGTC-TAT 171 0 26 WO 01/46231 PCT/USOO/34898 The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. FCTR9 (AL117336_A) 5 The novel nucleic acid encoding a novel secreted protein is shown in Table 9A. This sequence contains an initiation codon at the 5' end, and a TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The start and stop codons are indicated in bold type. This sequence originates in chromosome 10 clone RP11-324122. Table 9A. FCTR9 (AL1 17336_A) nucleotide sequence (SEQ ID NO: 17). 0 atggcaaaggaggggccccaggagcccttgagaccgctgggcttgctgcctccccgcattctggcccag tgctgcttggtcactctggctgtgcctccagcaggcccagctctcaacgctggctgcacggtcaagacc tag The encoded protein is presented using the one-letter code in Table 9B. The protein 5 has a moderate probability of sorting to the plasma membrane. A signal peptide most likely is cleaved between residues 43 and 44, i.e., the dash in the amino acid sequence GCT-VKT. Table 9B. Encoded FCTR9 protein sequence (SEQ ID NO:18). MAKEGPQE PLRPLGLL PPRILAQCCLVTLAVPPAGPALNAGCTVKT 0 In a search of sequence databases no similarities of significance were identified at either the nucleic acid or the amino acid level. The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 5 FCTR1O (AL118509_A) The novel nucleic acid encoding a novel secreted protein is shown in Table 10A. This sequence contains an initiation codon at the 5' end, and a TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. The start and stop codons are shown in bold type. This sequence originates in chromosome 20 clone RP4-770C23. 0 Table 10A. FCTR10 (AL118509_A) nucleotide sequence (SEQ ID NO:19) atgcactcactgcggttcctactgcttttgtggttgctgtttcctctgtcactgctatccttctcttcc cctacagtagggtttctggactgcggcacagttgtcacttcagaccaggtaagggctctattaattatg ttctatgaatcacaatcagatttaaaaacaaacaaaaataaaacaaaacaaaaaaaaaaagagaaggg aaggagcggtctgtgaacgttaacaaatggaaatccactggggatcagcctctgtcagaactaagctcc 5 aggaaggaggaggttcagccagttgaggagccagtatcattatcagaagggaatttaggaaaaagcaag aaggtgatgaagaatgagagggaggaagaaaagaaggaaaaggaacaaacttccagcttctcacaattc ccttctgaaagacgtacactgcccatggcaaggcacgctggatatgggttaagtaaccccaatctgaaa 27 WO 01/46231 PCT/USOO/34898 atccaaaatccaaaatgctacaacatcccaaatgttttgagtgccaatgtgatgatcaatggaaatgtt cactag The encoded protein is presented using the one-letter code in Table 10B. The protein 5 has a high probability of being secreted extracellularly. A signal peptide most likely is cleaved between residues 27 and 28, i.e. at the dash in the sequence TVG-FLD. Table 10B. Encoded FCTR10 protein sequence (SEQ ID NO:20). MHSLRFLLLLWLLFPLSLLSFSSPTVGFLDCGTVVTSDQVRALLIMFYESQSDLKTNKNKTKQKQKREG KERSVNVNKWKSTGDQPLSELSSRKEEVQPVEEPVSLSEGNLGKSKKVMKNEREEEKKEKEQTSSFSQF 0 PSERRTLPMARHAGYGLSNPNLKIQNPKCYNIPNVLSANVMINGNVH In a search of sequence databases, the BLASTN comparison (see Table 10C) revealed 90 of 117 bases (76%), in a large genomic fragment originating on chromosome 6q23.1-24.3, are identical to a human DNA sequence containing the MEKK5 (ASK1, iAPKKK5) gene for 5 MAP/ERK kinase kinase 5 (Mitogen Activated Protein kinase kinase kinase 5), as well as ESTs, GSSs and a putative CpG island. No similarities of significance were identified at the amino acid level. Table 10C. BLASTN of FCTR10 with MEKK5. >gb:GENBANK-ID:HS325F22Iacc:AL024508 Human DNA sequence from clone 325F22 on 0 chromosome 6q23.1-24.3. Contains the MEKK5 (ASK1, MAPKKK5) gene for MAP/ERK kinase kinase 5 (Mitogen Activated Protein kinase kinase kinase 5), ESTs, GSSs and a putative CpG island, complete sequence - Homo sapiens, 154788 bp. Score = 330 (49.5 bits), Expect = 1.9e-10, Sum P(2) = 1.9e-10 Identities = 90/117 (76%), Positives = 90/117 (76%), Strand = Plus / Plus 5 Query: 444 AAGGCACGCTGGATATGG--GTTAAGTAACCCCAATCTGAAAATCCAAAATCCAAAATGC 501 1 1 1 1 1 1 l I 1 1 1 1 1 1 1 1 1 1 1 1 1 1 111111 1 1 1 1 1 1111111 Sbjct: 145356 AAGGCACACTGTAAATACAAGTTGAGTAACCCTAATAAAAAAATCTGAAATCTAAAATGC 145415 0 Query: 502 TACAACATCCCAAATGTTTTGAGTGCCAATGTGATGATCAATGGAAATGTTCACTAG 558 l iI i l iil I I I I i l I ii l l I I I ll l i i i i lIIl II ii i | Sbjct: 145416 TCCAAAATCCAAAACTTTTTGAGTGCCAACATGATGCTCAAAGGAAATGCTCATTGG 145472 (SEQ ID NO:51) 5 Score = 163 (24.5 bits), Expect = 1.9e-10, Sum P(2) = 1.9e-10 Identities = 69/96 (71%), Positives = 69/96 (71%), Strand = Plus / Plus Query: 145 GAATCACAATCAGATTTAAAAACAAACAAAAATAAAA-CAAAA-CAAAAACAAAAAAGAG 202 0 11 I 1 1 | II I I i 1i ll 111111111 lill 11111 i i I iiI il Sbjct: 29978 GAGTGAGACTCCG-TCTCAAAA-AAACAAAAACAAAAACAAAAACAAAAACAAAAACAAG 30035 Query: 203 AAGGGAAGGAGCGGTCTGTGAACGTTAACAAATGGAAA 240 i i I I I I I 1 I 1 1 1 1 1 I M I II 5 Sbjct: 30036 AA---ATGCATCCATAT-T-AAC-TTC-CAAATGCAAA 30066 (SEQ ID NO:52) Score = 121 (18.2 bits), Expect = 1.4e-08, Sum P(2) = 1.4e-08 Identities = 57/86 (66%), Positives = 57/86 (66%), Strand = Plus / Plus 0 Query: 94 GGCACAGTTGTCACTTCAGACCAGGT-A-AGGGCTCTATT-AATTATGTTCTATGAATCA 150 11111 11 1111 11 iii 1 1 111 I I I i Sbjct: 13513 GGCACTATTTT-ACTTT-GAGGTGATTACATTGCTTTACTCAAAGAACTTGGTGGAATGG 13570 5 Query: 151 CAATCAGATTTAAAAACAAACAAAAATAA 179 28 WO 01/46231 PCT/USOO/34898 Sbjct: 13571 CTAA-AGTTTTAAAAACAAACAAAACTAA 13598 (SEQ ID NO:53) 5 Score = 117 (17.6 bits), Expect = 2.0e-08, Sum P(2) = 2.0e-08 Identities = 27/30 (90%), Positives = 27/30 (90%), Strand = Plus / Plus Query: 171 CAAAAATAAAACAAAACAAAAACAAAAAAG 200 0 Sbjct: 101741 CAAAAATAAAACAAAACAAAAG-AATAAAG 101769 (SEQ ID NO:54) The nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 5 FCTR11 (AL118522_AEXT) The novel nucleic acid encoding a novel K+ channel-like protein is shown in Table 11 A. This sequence contains an initiation codon at the 5' end, and a TAA stop codon was identified near the 3' end indicating that this sequence is a coding sequence. The start and stop codons are shown in bold type and a putative 3'UTR is underlined. This sequence 0 originates in chromosome 20 and was assembled as a consensus extension using the 8 sequences FCTR1 1 AL 118522_genscan_2+, est:gb AA283204+, est:gb_AI091631-, est:gb_AI097455+, est:gb_A690321+, est:gb_A1739096+, est:gb_A1968607+, and est:gbAW073155+. Table 11A. FCTR1 1 (ALl 18522_A_EXT) nucleotide sequence (SEQ ID NO:21). 5 ATGCGGAGGCCGAGCGTGCGCGCGGCCGGGCTGGTCCTGTGCACCCTGTGTTACCTGCTGGTGGGCGCT GCTGTCTTCGACGCGCTCGAGTCCGAGGCGGAAAGCGGCCGCCAGCGACTGCTGGTCCAGAAGCGGGGC GCTCTCCGGAGGAAGTTCGGCTTCTCGGCCGAGGACTACCGCGAGCTGGAGCGCCTGGCGCTCCAGGCT GAGCCCCACCGCGCCGGCCGCCAGTGGAAGTTCCCCGGCTCCTTCTACTTCGCCATCACCGTCATCACT ACCATCGAGTACGGCCACGCCGCGCCGGGTACGGACTCCGGCAAGGTCTTCTGCATGTTCTACGCGCTC 0 CTGGGCATCCCGCTGACGCTGGTCACTTTCCAGAGCCTGGGCGAACGGCTGAACGCGGTGGTGCGGCGC CTCCTGTTGGCGGCCAAGTGCTGCCTGGGCCTGCGGTGGACGTGCGTGTCCACGGAGAACCTGGTGGTG GCCGGGCTGCTGGCGTGTGCCGCCACCCTGGCCCTCGGGGCCGTCGCCTTCTCGCACTTCGAGGGCTGG ACCTTCTTCCACGCCTACTACTACTGCTTCATCACCCTCACCACCATCGGCTTCGGCGACTTCGTGGCA CTGCAGAGCGGCGAGGCGCTGCAGAGGAAGCTCCCCTACGTGGCCTTCAGCTTCCTCTACATCCTCCTG 5 GGGCTCACGGTCATTGGCGCCTTCCTCAACCTGGTGGTCCTGCGCTTCCTCGTTGCCAGCGCCGACTGG CCCGAGCGCGCTGCCCGCACCCCCAGCCCGCGCCCCCCGGGGGCGCCCGAGAGCCGTGGCCTCTGGCTG CCCCGCCGCCCGGCCCGCTCCGTGGGCTCCGCCTCTGTCTTCTGCCACGTGCACAAGCTGGAGAGGTGC GCCCGCGACAACCTGGGCTTTTCGCCCCCCTCGAGCCCGGGGGTCGTGCGTGGCGGGCAGGCTCCCAGG CTTGGGGCCCGGTGGAAGTCCATCTGACAACCCCACCCAGGCCAGGGTCGAATCTGGAATGGGAGGGTC 0 TGGCTTCAGCTATCAGGGCACCCTCCCCAGGGATTGGAAACGGATGACGGGCCTTTAGGCGGTTTTTTG CCACGAGCAGTTTTTCATTACTGTCTGTGGCTAAGTCCCCTCCCTCCTTTCCAAAAATATATTACAGTC ACCCCATAAGCCCAA 29 WO 01/46231 PCT/USOO/34898 The encoded protein is presented using the one-letter code in Table 1 1B. The protein has a high probability of being sorted to the plasma membrane. A signal peptide most likely is cleaved between residues 23 and 24, i.e. at the dash in the sequence VGA-AVF. Table 11B. Encoded FCTR11 protein sequence (SEQ ID NO:22). 5 MRRPSVRAAGLVLCTLCYLLVGAAVFDALESEAESGRQRLLVQKRGALRRKFGFSAEDYRELERLALQA EPHRAGRQWKFPGSFYFAITVITTIEYGHAAPGTDSGKVFCMFYALLGIPLTLVTFQSLGERLNAVVRR LLLAAKCCLGLRWTCVSTENLVVAGLLACAATLALGAVAFSHFEGWTFFHAYYYCFITLTT IGFGDFVA LQSGEALQRKLPYVAFSFLYILLGLTVIGAFLNLVVLRFLVASADWPERAARTPSPRPPGAPESRGLWL PRRPARSVGSASVFCHVHKLERCARDNLGFS PPSS PGVVRGGQAPRLGARWKSIXQPHPGQGRIWNGRV 0 WLQLSGHPPQGLETDDGPLGGFLPRAVFHYCLWLSPLPPFQKYITVTP In a search of sequence databases, the BLASTN comparison (see Table 11 C) revealed 641 of 854 bases (75%), in a complete coding sequence of 2590 bases, are identical to a human mRNA encoding TWIK-related acid-sensitive K+ channel (TASK) (GenBank 5 ID:AF006823). In a BLASTX comparison it was found that the full amino acid sequence of the protein has 168 of 258 residues (65%), identical to, and 200 of 258 residues (77%) positive with, mouse CTBAK having a total of 409 amino acid residues (SPTREMBL-ACC:0351 11) (Table 11D). Table 11C. BLASTN of FCTR1 1 with TWIK (SEQ ID NO:55). 0 >gb:GENBANK-ID:AF006823lacc:AF006823 Homo sapiens TWIK-related acid-sensitive K+ channel (TASK) mRNA, complete cds - Homo sapiens, 2590 bp (RNA). Score = 2097 (314.6 bits), Expect = 4.2e-89, P = 4.2e-89 Identities = 641/854 (75%), Positives = 641/854 (75%), Strand = Plus / Plus 5 Query: 1 ATGCGGAGGCCGAGCGTGCGCGCGGCCGG-GCTGGTCCTGTGCACCCTGTGTTACCTGCT 59 I l I I lI l l I I11 1 11I 111i i I i I I IIIlIII l IIlIIIII Sbjct: 126 ATGAAGCGGCAGAACGTGCGCACG-CTGGCGCTCATCGTGTGCACCTTCACCTACCTGCT 184 0 Query: 60 GGTGGGCGCTGCTGTCTTCGACGCGCTCGAGTCCGAGGCGGAAA-G--CGGCCGCCAGCG 116 i iIlIii iiIiii IlIII III i ii i I I I 111111I Sbjct: 185 GGTGGGCGCCGCGGTCTTCGACGCGCTGGAGTCGGAGCCCGAGCTGATCGAGCGGCAGCG 244 Query: 117 ACTGCTGGTCCAGAAGCGGGGCGCTCTCCGGAGGAAGTTCGGCTTCTCGGCC-GAG-GAC 174 5 ill I I1 1i 1I I ll 1 iI I I I I IlI lI l I I I I Sbjct: 245 GCTGGAGCTGCGGCAGCAGGA-GCTG--CGGGCGCGCTACAAC--CTCAGCCAGGGCGGC 299 Query: 175 TACCGCGAGCTGGAGCGCCTGGCGCTCCAGGCTGA-GCCCCACCGCGCCGGCCGCCAGTG 233 0 Sbjct: 300 TACGAGGAGCTGGAGCGCGTCGTGCTGC-GCCTCAAGCCGCACAAGGCCGGCGTGCAGTG 358 Query: 234 GAAGTTCCCCGGCTCCTTCTACTTCGCCATCACCGTCATCACTACCATCGAGTACGGCCA 293 I I li I I I I l I iiiiiI I iIl I II l II I I lII ii IIlllI I 111II lI1l Sbjct: 359 GCGCTTCGCCGGCTCCTTCTACTTCGCCATCACCGTCATCACCACCATCGGCTACGGGCA 418 5 Query: 294 CGCCGCGCCGGGTACGGACTCCGGCAAGGTCTTCTGCATGTTCTACGCGCTCCTGGGCAT 353 I I I II ii I 111 IIIiiii IiIIIlI1 I IiilI | Sbjct: 419 CGCGGCACCCAGCACGGATGGCGGCAAGGTGTTCTGCATGTTCTACGCGCTGCTGGGCAT 478 0 Query: 354 CCCGCTGACGCTGGTCACTTTCCAGAGCCTGGGCGAACGGCTGAACGCGGTGGTGCGGCG 413 I I I I I I 1 1 1 111 1 1 IIIIi II Sbjct: 479 CCCGCTCACGCTCGTCATGTTCCAGAGCCTGGGCGAGCGCATCAACACCTTGGTGAGGTA 538 Query: 414 CCTCCTGTTGGCG-GCCAAGTGCTGCCTGGGCCTGCGGTG-GACGTGCGTGTCCACGGAG 471 5 Ill 111 Ii 111111 I 111111 lill I I il 11111111 il 30 WO 01/46231 PCT/USOO/34898 Sbjct: 539 CCTGCTGCAC-CGCGCCAAGAAGGGGCTGGGCATGCGGCGCGCCGA-CGTGTCCATGGCC 596 Query: 472 AACCTGGTGGTGGCCGGGCTGCTGG-CGTGTGCCGCCACCCTGGCCCTCGGGGCCGTCGC 530 Ii i I I 111111 1 1111 iii III 5 Sbjct: 597 AACATGGTGCTCATCGG-CTTCTTCTCGTGCATCAGCACGCTGTGCATCGGCGCCGCCGC 655 Query: 531 CTTCTCGCACTTCGAGGGCTGGACCTTCTTCCACGCCTACTACTACTGCTTCATCACCCT 590 Sbjct: 656 CTTCTCCCACTACGAGCACTGGACCTTCTTCCAGGCCTACTACTACTGCTTCATCACCCT 715 0 Query: 591 CACCACCATCGGCTTCGGCGACTTCGTGGCACTGCAGAGCGGCGAGGCGCTGCAGAGGAA 650 l I l i i I I I I i i i I I I i I i i iI I I I I I I I i | I i iI I I I I I I I Sbjct: 716 CACCACCATCGGCTTCGGCGACTACGTGGCGCTGCAGAAGGACCAGGCCCTGCAGACGCA 775 5 Query: 651 GCTCCCCTACGTGGCCTTCAGCTTCCTCTACATCCTCCTGGGGCTCACGGTCATTGGCGC 710 II I i II II iiii I iiiiIi i i I I I III I ii I l i III I Ii IliI iiil 11111 Sbjct: 776 GCCGCAGTACGTGGCCTTCAGCTTCGTCTACATCCTTACGGGCCTCACGGTCATCGGCGC 835 Query: 711 CTTCCTCAACCTGGTGGTCCTGCGCTTCCTCGTTGCCAGCGCCGACTGGCCCGA-GCGCG 769 0 11Il i I llI l 11111 111111111 i I lli I 1 I 1| 11111 Sbjct: 836 CTTCCTCAACCTCGTGGTGCTGCGCTTCATGACCATGAACGCCGAG-GACGAGAAGCGCG 894 Query: 770 CTGCCC-GCACC-C-C-CAGCCCGCGCCCCCCGGG--GGCGCCCGAGAGCCGTGGCCTCT 823 III I I I I I I II I 11 I l Ii | I 11 1 1 1 5 Sbjct: 895 ACGCCGAGCACCGCGCGCTGCTCACGCGCAACGGGCAGGCGGGCGGCGGCGGAGGGGG-T 953 Query: 824 GGCTGCCCCGC-CGCC-CGG-C-CCGC 846 1I I 11 1 11 1 1 1 I Sbjct: 954 GGCAGCGC-GCACACTACGGACACCGC 979 0 Strong homology was found between FCTR1 1 and the human mRNA encoding the two pore potassium channel KT3.3, as shown in Table 1 1D. The FCTR1 1 nucleic acid is on line 1, the KT3.3 mRNA (GenBank gill16412741refl INM_022358.1 Homo sapiens two pore potassium channel KT3.3 (LOC64181),mRNA) is on line 2, and the KT3.3 complete mRNA 5 (GenBank gil1 12286851gblAF257081.1 IAF257081 Homo sapiens two pore potassium channel KT3.3 mRNA, complete) is on line 3. Table I1D. BLASTN of FCTR1 1 nucleotide with human KT3. Line 2 > gi1116412741ref INM_022358.11 KT3.3 (LOC64181),mRNA Line 3 > gil11228685gblAF257081.1lAF257081 channel KT3.3 mRNA, complete 0 10 20 30 40 50 60 FCTR11 --------- Line 2 GGAGCCGCGG GACA CGAG AGGTGGGC A TA Line 3 AGACACAGAG ACA 5 70 80 90 100 110 120 FCTR11 ---- ACAG Line 2 AT AG AGACA 0 Line 3 C AT AG G AG CAC AC 130 140 150 160 170 180 FCTRll GCTG 5 Line 2 GGGC G TTACGC GCTCAG TCCAGGG GAAAG C CAG Line 3 GGGC GGT ACGCGCTCGAGTCCGAGGCGGAAAGCGCCAG 190 200 210 220 230 240 0 FCTRl ^ C Line 2 A CAAAGAGAAGAGA A Line 3 A C T AGAAGGAGGAAGCAGGAC A 31 WO 01/46231 PCT/USOO/34898 250 260 270 280 290 300 5 Line 2 AG AGAG AG A A A Line 3 CCGCGLjeyACTGACCTGGCCAGTACCCCGGCGCGCGG 310 320 330 340 350 360 0 FCTR11 GTCCGCCTCTCTGCTACGCTATCACGGAGCAG Line 2 AC ACA CACACACAT AC A Line 3 AC ATAC A TA TAC AT G A A 370 380 390 400 410 420 5 FCTR11 Line 2 AC AC AAGAT ACA Line 3 AC AC AAGAT ACA 0 430 440 450 460 470 480 ....|I ....I .... I .... I .... I .... I .... I .... |I .... |I .... I .... I .... I FCTR11 GTGC~~ TG~TCAC*PTOVMML[ieeO~~TTCAGGC TGGGCGACGT GAACGCGGTGGTGCGGCGCCTre Line 2 ACAC AGA Line 3 GCTGACGCTGGTCACT C CAGAGCCTGGGCGAACGGCTGAACGCGGTG C C C 5 490 500 510 520 530 540 ....I .... I .... I .... |I .... I .... I .... I .... I .... I .... I .... I ....|I FCTRl1CTGAGAG Line 2 AAC TAC A A o Line 3 CCTTGGCGGCCAAGTGCTGGCCT CGCTGGACGT GGTGACGGAGAACCT 550 560 570 580 590 600 FCTR11 GG 5 Line 2 GTGGCGGTCGCT CAC Line 3 A 610. 620 630 640 650 660 0 FCTR11 GA A Line 2 AC AG ACAC ACACAC ATAC ACA Line 3 GA AG AC T A ATACA TACAC ACA 670 680 690 700 710 720 5 .... FCTR11CTGCTGCATCGGCCGAACGGGGGTCGGAGTC Line 2 ATACAC AGAG AGAGA Line 3 ATACAC AGAG AGAGA 0 730 740 750 760 770 780 ....I .... I .... I .... I .... |I .... |I .... I .... I .... I .... |I .... I ....|I FCTR11 CTACGT GGCCT AGCTT CCTCA ATCCTC CT TCACGG AT TGGCGCCTTCCT Line 2 A CAG AAA A Line ACAGACATAC A 5 790 800 810 820 830 840 FCTR11 C4iiAACTeGGGG~rTCCGGTTCCTGCACCCATGCCACCGTCC Line 2 AAC GAG ACA O Line 3 AAT AG A C A 850 860 870 880 890 900 FCTR1l AC AGAG 5 Line AGAGA Line 3 AGAGA 910 920 930 940 950 960 0 FCTR11 CA ACAAGAGA 32 WO 01/46231 PCT/USOO/34898 Line 2 CACGTGCACAAGCTGGAGAGG Line 3 CCGA AAAGCT GA AGGT 970 980 990 1000 1010 1020 5 ....| . . | . . . . . . | . . . . 1 . . . . . . . . . . . .. . . .I FCTR11 A Line 2 C ACATAGA Line 3 CGCCCGCGA AAC CTGGGCTTTTCGCCCCCCTCAGCCC GGGGCGTGGTGGGGGA 0 1030 1040 1050 1060 1070 1080 FCTR1l T Line 2 AGAATAT ACAAC ACAG A Line 3 AGAATAT ACAAC ACAG A 5 1090 1100 1110 1120 1130 1140 FCTR11GG Line 2 AAGAGAGATAG AC AGA 0 Line 3 GA A GAGATACAG ACAGA 1150 1160 1170 1180 1190 1200 FCTR11 GGATACGGGCCTT G TTTTTCTCCCIGC-G TTTTTCATTACTGTCTGTGGC 5 Line 2 ATA CAG A AAG T A Line 3 GATGAC G CAGA CGAGCAAGT TTCAT TAC 1210 1220 1230 1240 1250 1260 0 FCTR11 T CCC Y Line 2 AA AAAAATATA ACA AC A AAAAAAAAAAAAAAA Line 3 AAGAAAAATATA ACA AC A AAAAAAAAAAAAAAA 1270 1280 5 .... _.. ._. .. ... . Line 2 (SEQ ID NO:56) Line 3 A (SEQ ID NO:57) 0 A BlastP search against the FCTR1 1 protein also identified FCTR1 1 as having high homology to the potassium channel proteins TASK and KT3.3, as shown in Table 11E. Line 1 shows the FCTR1 1 polypeptide (SEQ ID NO:22), line 2 is the human TASK protein (gil109442751 emblCAC14068.11 (AL118522) dJ781B1.1 (A novel protein similar to the acid sensitive potassium channel protein TASK (KCNK3)) [Homo sapiens])(SEQ ID NO:58), line 5 3 is the human KT3.3 protein (gil112286861 gblAAG33127.1|AF257081_1 (AF257081) two pore potassium channel KT3.3 [Homo sapiens]) (SEQ ID NO:59), and line 4 is the guinea pig TASK3 protein (gil75468391gblAAF63706.llAF212827_1 (AF212827) potassium channel TASK3 [Cavia porcellus])(SEQ ID NO:60). Table I1E: BlastP search of FCTR1 1 protein 0 10 20 30 40 50 60 FCTRIl MRRPSVAAVLCT LCYLLVAAFDA LESEAE n SSG RQRLV ALRR KFGFAE D Line 2 MP AAGGAAVDALEAESG RAL 5 Line 3 MRRPSVAALV L C T AAV FALEAESG KARFGA Line 4 MKQN RTLS A CFTYLAAVFASHE RE9E 1LKAZEIRRG NISTEY 70 80 90 100 110 120 33 WO 01/46231 PCT/USOO/34898 FCTR11 E LV E YAAP G K V FA Line 2 E AA AGI E Line 3 SGY Line 4 MLLIL P Q IWVTTIG 5 130 140 150 160 170 180 FCTR11 SLAGACAATALAVA Line 2 T Q NA0 eAAKC C LGRWCV VAG LA A A A o Line 3 S LAR R L A K CA AAA Line 4 TLMFQLMNTF RYRK' 190 200 210 220 230 240 5 FCTR11 FG FAYG AL ALR AFAF Line 2 FG FAYG ALG AL YAFAF Line 3 FWTFF Y I G F V QE RKLTPYVA FFYILLGLTVI GLN Line 4 CESFAYC LT G ALQSKGA YA L AFN 0 250 260 270 280 290 300 FCTRllLVRLAAWEARTSRPAERLLRPASGAVCVKEC Line 2 LVVRFVA AWERAARTPSPRPGAPERGLWPRRNNPARSGAVCVKEC Line 3 L RVS ERS A 5 Line 4 LR FL ETM SDIE-ERGEGEEGAALP PSS-V YTHISEE A QVRQRYRGE.GDLQOSVCR 310 320 330 340 350 360 FCTR11 03 55 NOW40AS XQPHPGQGRIWNGRVWLQLSGHPPQGLETD 0 Line 2 -- ---------------------- Line 3 P ----------------------------- Line 4 CACYRSQNFGATAPQPLHS IE SP----STLKNSLFPSPISSVSPGLHSFG 370 380 390 5 . . . . I . . . . I . . . . I . . . . I . . . . I . . . FCTR11 DGPLGGFLPRAVFHYCLWLSPLPPFQKYITVTP Line 2 --------------------------------- (SEQ ID NO:58) Line 3 --------------------------------- (SEQ ID NO:59) Line 4 DNHRLMLRRKSV--------------------- (SEQ ID NO:60) 0 Table 11F. BLASTX of FCTR1 1 nucleotide with CTBAK. >ptnr:SPTREMBL-ACC:035111 CTBAK - MUS MUSCULUS (MOUSE), 409 aa. Score = 832 (292.9 bits), Expect = 1.0e-85, Sum P(2) = 1.0e-85 Identities = 168/258 (65%), Positives = 200/258 (77%), Frame = +1 5 Query: 1 MRRPSVRAAGLVLCTLCYLLVGAAVFDALESEAES-GRQRLLVQKRGALRRKFGFSAEDY 177 1+1 +11 i++II I1 Ili I .II + ++ I I Sbjct: 1 MKRQNVRTLALIVCTFTYLLVGAAVFDALESEPEMIERQRLELRQL-ELRARYNLSEGGY 59 0 Query: 178 RELERLALQAEPHRAGRQWKFPGSFYFAITVITTIEYGHAAPGTDSGKVFCMFYALLGIP 357 I I Il I+ 1+ + 1 1+ 1 1 1 I+ 1 1 1 1 1 1 1iI i i i i i i Sbjct: 60 EELERVVLRLKPHKAGVQWRFAGSFYFAITVITTIGYGHAAPSTDGGKVFCMFYALLGIP 119 Query: 358 LTLVTFQSLGERLNAVVRRLLLAAKCCLGLRWTCVSTENLVVAGLLACAATLALGAVAFS 537 5 liH 1111111+1 il li il 11+1 li 1+1+ 1 ++I +11 +11 ill Sbjct: 120 LTLVMFQSLGERINTFVRYLLHRAKRGLGMRHAEVSMANMVLIGFVSCISTLCIGAAAFS 179 Query: 538 HFEGWTFFHAYYYCFITLTTIGFGDFVALQSGEALQRKLPYVAFSFLYILLGLTVIGAFL 717 + + 1 1 1 1 1 I i I I I I I l l + 1 1 1 + 1 I I i I + 1 1 I i I I i 1 1I I 0 Sbjct; 180 YYERWTFFQAYYYCFITLTTIGFGDYVALQKDQALQTQPQYVAFSFVYILTGLTVIGAFL 239 Query: 718 NLVVLRFLVASADWPERAA 774 1II)Il|+ +1+ +1 1 Sbjct: 240 NLVVLRFMTMNAEDEKRDA 258 (SEQ ID NO:61) 5 Score 52 (18.3 bits), Expect = 1.Oe-85, Sum P(2) = 1.Oe-85 Identities = 17/35 (48%), Positives = 20/35 (57%), Frame = +2 34 WO 01/46231 PCT/USOO/34898 Query: 941 SCVAGRLPGLGPGGSPSDNPTQAR----VESGMGGSGF 1042 1 1++ 1 I I I I I 1+ 1 11 11 Sbjct: 277 SCLSG---SLGDGVRPRDPVTCAAAAGGVGVGVGGSGF 311 (SEQ ID NO:62) 5 Score = 40 (14.1 bits), Expect = 1.9e-84, Sum P(2) = 1.9e-84 Identities = 13/39 (33%), Positives = 16/39 (41%), Frame = +2 0 Query: 941 SCVAGRLPGLGPGGSPSDNPTQARVESGMGGSGFSYQGT 1057 +11 II 111 + +11 I I 1 Sbjct: 353 TCVEHSHSSPGGGGRYSDTPSHPCLCSGTQRSAISSVST 391 (SEQ ID NO:63) Potassium channels are ubiquitous multisubunit membrane proteins that regulate 5 membrane potential in numerous cell types. One family of mammalian K+ channels is characterized by the presence of 4 transmembrane domains and 2 P domains per subunit; this family includes TASK, TWIK (KCNK1; OMIM 601745) and TREK (KCNK2; OMIM 603219). See, Duprat et al., 1997 EMBO J. 16: 5464-5471. The human cDNA, designated TASK, encodes a 394-amino acid polypeptide with 85% identity to the mouse ortholog. See, 0 Duprat et al., 1997. The sequence contains consensus sites for N-linked glycosylation and for phosphorylation at the C-terminal. Northern blot analysis showed that TASK is expressed in a variety of human tissues, with highest levels in pancreas and placenta. See, Duprat et al., 1997. Expression of the TASK cDNA revealed that the functional protein creates currents that are K(+)-selective, instantaneous, and noninactivating. See, OMIM 603220. These currents 5 showed an outward rectification when external K+ was low, but evinced absence of activation and inactivation kinetics as well as voltage independence, characteristics of so-called leak or background conductances. See, OMIM 603220. TASK currents were very sensitive to small changes in extracellular pH, suggesting that TASK has a role in cellular responses to changes in extracellular pH. See, OMIM 603220. 0 Finally, FCTR1 1 was found to have high homology to the domains shown in Table 11G. Table 11G: CD domain analysis of FCTR1 1 Score E Sequences producing significant alignments: (bits) value TWIK_channel, TASK K+ channel 284 5e-78 CNGmembrane, Transmembrane region cyclic Nucleotide Gated Cha... 35.8 0.004 The nucleic acids and proteins of the invention are potentially useful in the treatment 5 of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 35 WO 01/46231 PCT/USOO/34898 FCTR12 (AL121574_A) The novel nucleic acid encoding a novel protein C-terminal fragment is shown in Table 12A. A TAG stop codon was identified at the 3' end indicating that this sequence is a coding sequence. This sequence originates in clone RP3-441A12 of chromosome 6. 5 Table 12A. FCTR12 (AL121574_A) nucleotide sequence (SEQ ID NO:23). natcagactctattgaccgccactctaacgttgtcaggcattgtggcaattgtgtccttgtggctttgg gcatttaagcttcactacttgacctctatagttttggcatcttctcatacacatgactatcagcaagct aaattatttactgactgtcctgctccccgcactccgcctttgaggcgcggaacgaagtggcacgcccgg atcccagctgatcagcggctgggctttggcgttggctcccccgggcgagaccattgtgactcctcggga 0 ggggcgcacgccggggagggggcggagcggccattgtccggtcagcgcagcctccgggggaggggacgg tgttacggagacagcagggcccggggcttcagagcggccgctgcgactccggagccggcggggggctcc ggtccttccctgcgccaccgcacaggacatctctctggctggggagcggcggtgagacccgccgagggc gtctgtgtccctcctcccccgcggtcctcgagcggggcccgggcccagccgccgccaccgctgccgccg ccgagctccgccgccgccgagcaccatgggagacgctgggagcgagcgcagcaaagcgcccagcctgcc 5 gcctcgctgtccctgcggcttctggggactaacggcagttcctttaggattgctgctctttcgagtgac ttaggctgcaggacttgctgcccagcattgcccagtcaggacactaatcagtgtggctcggttgaatag The encoded C-terminal fragment of the encoded protein is presented using the one letter code in Table 12B. The C-terminal fragment disclosed has a very high probability of 0 being sorted to the plasma membrane. No cleavage site for a signal peptide was detected. Table 12B. Encoded FCTR12 protein sequence (SEQ ID NO:24). XQTLLTATLTLSGIVAIVSLWLWAFKLHYLTS IVLASSHTHDYQQAKLFTDCPAPRTPPLRRGTKWHAR IPADQRLGFGVGSPGRDHCDSSGGAHAGEGAERPLSGQRSLRGRGRCYGDSRARGFRAAAATPEPAGGS GPSLRHRTGHLSGWGAAVRPAEGVCVPPPPRS S SGARAQPPPPLPPPSSAAAEHHGRRWERAQQSAQPA 5 ASLSLRLLGTNGSSFRIAALSSDLGCRTCCPALPSQDTNQCGSVE In a search of sequence databases, no similarities were found to any known nucleic acid or protein. The nucleic acids and proteins of the invention are potentially useful in the treatment 0 of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. FCTR13 (AL121723_A) A novel nucleic acid encoding a novel secreted morphogenic protein is shown in Table 13A. It was identified in chromosome 20 clone RP5-854E16. An initiation codon is shown at 5 the beginning of the sequence and a TGA stop codon was identified at the 3' end indicating that this sequence is a coding sequence. These are shown in bold face in Figure 13A. Table 13A. Nucleotide sequence (SEQ ID NO:25) of FCTR13 (AL121723_A). atgcggcatccgctggtcctgctgctgctcctctctgccctggtgacctccttcactgcagcctctatc cacgatgctcatgcccaagagagctccttgggtcttacaggcctccagagcctactccaaggcttcagc 0 cgacttttcctgaaagatgacctgcttcggggcatagacagcttcttctctgcccccatggacttccgg 36 WO 01/46231 PCT/USOO/34898 ggcctccctaggaactaccaacaagaggagaacgaggagcaccagctgaggaacaacaccctctccagc cacctccatattgacaaggtgaccgacaataagacaggagaggtgctgatctccgagaaggtggtggca tccatccagccggcggaggggagcttcgagggtaactggaaggcggcggccctggtgtccatccggaag gctatggacaacttccatgcagagctccatccccgggtggccttttggatcatgaagctgccacggtgg 5 aggtcccaccacaatgtcctggagggcggccgctggctcagtgagaagcgacaccgcctgcaggccatc caggatgggctccacgaggggacccgcgaggacgtcctaaaagaggggacccagggctcctcccactcc gggctgtcctccgaaagacccacttactgtacatcttcaggctttcctggcagctataggggttgggac cggggagcacctgcaagctgggttggtgtctgggtcagcgtatcaaagggcctggcacatggacccaca gggttgggcctggagcctggatccagtgggatagactttgtgaatgcgttcatggagggctacagtaac 0 caaaacatcatggtactagtacaaaaacggatacatagaccaatgcaacagaacagagaggccagaaat aaggccacacacctacaaccatctgatcttcgacaaagctga The encoded protein is presented using the one-letter code in Table 13B. The protein has a very high probability of secreted extracellularly. Cleavage of a signal peptide is 5 predicted to occur between residues 28 and 29, i.e. at the dash in the sequence AHA-QES. Table 13B. Encoded FCTR13 protein sequence (SEQ ID NO:26). MRHPLVLLLLLSALVTSFTAASIHDAHAQESSLGLTGLQSLLQGFSRLFLKDDLLRGIDSFFSAPMDFR GLPRNYQQEENEEHQLRNNTLSSHLHIDKVTDNKTGEVLISEKVVASIQPAEGSFEGNWKAAALVSIRK AMDNFHAELHPRVAFWIMKLPRWRSHHNVLEGGRWLSEKRHRLQAIQDGLHEGTREDVLKEGTQGSSHS 0 GLSSERPTYCTSSGFPGSYRGWDRGAPASWVGVWVSVSKGLAHGPTGLGLEPGSSGIDFVNAFMEGYSN QNIMVLVQKRIHRPMQQNREARNKATHLQPS DLRQS In a search of sequence databases, it was found, for example, that the nucleic acid sequence has 356 of 388 bases (91%) identical to human cysteine-rich secreted protein-like-N 5 cDNA (patn: : V07910) (Table 13C). The full amino acid sequence of the protein was found to have 166 of 218 residues (76%), identical to, and 181 of 218 residues (83%) positive with, human dickkopf-1 (dkk-1) having a total of 242 amino acid residues. This protein (soggy-1 protein) is a member of a novel family of secreted proteins and functions in head induction during Xenopus embryogenesis, acting as a potent inhibitor of Wnt signaling. 0 (TREMBLNEW-ACC:AAF02678) (Table 13D). Table 13C. BLASTN of FCTR13 with GenBank V07910 >patn:V07910 Human cysteine-rich secreted protein-like-N cDNA - Homo sapiens, 928 bp. Score = 1652 (247.9 bits), Expect = 1.6e-123, Sum P(2) = 1.6e-123 5 Identities = 356/388 (91%), Positives = 356/388 (91%), Strand = Plus / Plus Query: 1 ATGCGGCATCCGCTGGTCCTGCTGCTGCTCCTCTCTGCCCTGGTGACCTCCTTCACTGCA 60 Sbjct: 105 AGGCGGCATCTGCTGGTCCTGCTGCTGCTCCTCTCTACCCTGGTGATCCCCTCCGCTGCA 164 0 Query: 61 GCCTCTATCCACGATGCTCATGCCCAAGAGAGCTCCTTGGGTCTTACAGGCCTCCAGAGC 120 I I 1 1 1i 1 1 1 I I I I I I I I I I I I I I I l Sbjct: 165 GCTCCTATCCATGATGCTGACGCCCAAGAGAGCTCCTTGGGTCTCACAGGCCTCCAGAGC 224 5 Query: 121 CTACTCCAAGGCTTCAGCCGACTTTTCCTGAAAGATGACCTGCTTCGGGGCATAGACAGC 180 Iii I IIIIiiIIiIiIiiIIIIiiIII II 11 1 l [ 1 1 111 111 I I IiIi i Il II I 1 I | II l iIi Sbjct: 225 CTACTCCAAGGCTTCAGCCGACTTTTCCTGAAAGGTAACCTGCTTCGGGGCATAGACAGC 284 Query: 181 TTCTTCTCTGCCCCCATGGACTTCCGGGGCCTCCCTAGGAACTACCAACAAGAGGAGAAC 240 0 , I IIl IIllll I I II I llill l Ill ll lll1 Sbjct: 285 TTATTCTCTGCCCCCATGGACTTCCGGGGCCTCCCTGGGAACTACCACAAAGAGGAGAAC 344 37 WO 01/46231 PCT/USOO/34898 Query: 241 GAGGAGCACCAGCTGAGGAACAACACCCTCTCCAGCCACCTCCATATTGACAAGGTGACC 300 Sbjct: 345 CAGGAGCACCAGCTGGGGAACAACACCCTCTCCAGCCACCTCCAGATCGACAAGATGACC 404 5. Query: 301 GACAATAAGACAGGAGAGGTGCTGATCTCCGAGAAGGTGGTGGCATCCATCCAGCCGGCG 360 Sbjct: 405 GACAACAAGACAGGAGAGGTGCTGATCTCCGAGAATGTGGTGGCATCCATTCAACCAGCG 464 0 Query: 361 GAGGGGAGCTTCGAGGGTAACTGGAAGG 388 Sbjct: 465 GAGGGGAGCTTCGAGGGTGATTTGAAGG 492 (SEQ ID NO:64) 5 Score = 1244 (186.7 bits), Expect = 1.6e-123, Sum P(2) = 1.6e-123 Identities = 282/322 (87%), Positives = 282/322 (87%), Strand = Plus / Plus Query: 384 GAAGGCGGCGGCCCTGGTGTCCATCCGGAAGGCTATGGACAACTTCCATGCAGAGCTCCA 443 o Sbjct: 506 GGAGAAGGAGGCCCTGGTACCCATCCAGAAGGCCACGGACAGCTTCCACACAGAACTCCA 565 Query: 444 TCCCCGGGTGGCCTTTTGGATCATGAAGCTGCCACGGTGGAGGTCCCACCACAATGTCCT 503 Sbjct: 566 TCCCCGGGTGGCCTTCTGGATCATTAAGCTGCCACGGCGGAGGTCCCACCAGGATGCCCT 625 5 Query: 504 GGAGGGCGGCCGCTGGCTCAGTGAGAAGCGACACCGCCTGCAGGCCATCCAGGATGGGCT 563 Sbjct: 626 GGAGGGCGGCCACTGGCTCAGCGAGAAGCGACACCGCCTGCAGGCCATCCGGGATGGACT 685 0 Query: 564 CCACGAGGGGACCCGCGAGGACGTCCTAAAAGAGGGGACCCAGGGCTCCTCCCACTCCGG 623 Sbjct: 686 CCGCAAGGGGACCCACAAGGACGTCCTAGAAGAGGGGACCGAGAGCTCCTCCCACTCCAG 745 Query: 624 GCTGTCCTCC-GAAAGACCCACTTACTGTACATCTTCAGGCTTTCCTGGCAGCTATAGGG 682 5 1 1 1 1 1 1 1 1 i 1111 1 1 1 1 1 1 1 1 1 1 1 1 1 111111 i i 1 1 1 1 1 1 1 lI l1 Sbjct: 746 GCTGTCCCCCCGAAAGACCCACTTACTGTACATCCTCAGGCCCTCTCGGCAGCTGTAGGG 805 Query: 683 GTTGGGACCGGGGAGCACCTGC 704 iI l I l i i I l 111 I i i1 I I 0 Sbjct: 806 GTGGGGACCGGGGAGCACCTGC 827 (SEQ ID NO:65) Table 13D. BLASTX of FCTR13 with Soggy-1. >ptnr:TREMBLNEW-ACC:AAF02678 SOGGY-1 PROTEIN - Homo sapiens (Human), 242 aa. 5 Dickkopf-i (dkk-1) is member of a novel family of secreted proteins and functions in head induction during Xenopus embryogenesis, acting as a potent inhibitor of Wnt signaling. Score = 813 (286.2 bits), Expect = 6.3e-84, Sum P(2) = 6.3e-84 Identities = 166/218 (76%), Positives = 181/218 (83%), Frame = +1 0 Query: 4 RHPLVLLLLLSALVTSFTAASIHDAHAQESSLGLTGLQSLLQGFSRLFLKDDLLRGIDSF 183 II iiIIIIII 11 11 1 I il I I i i IlIII(iI ii I II I lII i II lilI i+11I1II Ii Sbjct: 12 RHLLVLLLLLSTLVIPSAAAPIHDADAQESSLGLTGLQSLLQGFSRLFLKGNLLRGIDSL 71 5 Query: 184 FSAPMDFRGLPRNYQQEENEEHQLRNNTLSSHLHIDKV-TDNKTGEVLISEKVVASIQPAE 363 Sbjct: 72 FSAPMDFRGLPGNYHKEENQEHQLGNNTLSSHLQIDKMTDNKTGEVLISENVVASIQPAE 131 Query: 364 GSFEGNWKAA-----ALVSIRKAMDNFHAELHPRVAFWIMKLPRWRSHHNVLEGGRWLS 525 0 111 1 + 1 ill 1+ 11 1+ 11 11111111| + 1111 lii + iI i III Sbjct: 132 GSFEGDLKVPRMEEKEALVPIQKATDSFHTELHPRVAFWIIKLPRRRSHQDALEGGHWLS 191 Query: 526 EKRHRLQAIQDGLHEGTREDVLKEGTQGSSHSGLSSER 639 iI I II I +l+lI+ +1I1+111+ 1i i l + 5 Sbjct: 192 EKRHRLQAIRDGLRKGTHKDVLEEGTESSSHSRLSPRK 229 (SEQ ID NO:66) 38 WO 01/46231 PCT/USOO/34898 Score = 54 (19.0 bits), Expect = 6.3e-84, Sum P(2) = 6.3e-84 Identities = 12/15 (80%), Positives = 12/15 (80%), Frame = +3 Query: 633 RKTHLLYIFRLSWQL 677 5 i l l l i l l i 1 1 1 i Sbjct: 228 RKTHLLYILRPSRQL 242 (SEQ ID NO:67) A multiple sequence alignment for FCTR13 AL121723_A A is given in Table 13E in a ClustalW analysis comparing the protein of the invention with related protein sequences. The 0 FCTR13 polypeptide is shown on line 1, the human Soggy-1 protein (gil7657554| ref] NP_055234.11 soggy-1 gene [Homo sapiens])(SEQ ID NO:68) on line 2, mouse Soggy-i protein (gil106445671 gblAAG21340.11 AF274312_1 (AF274312) soggy precursor [Mus musculus]) (SEQ ID NO:69) on line 3, and mouse Soggy-1 protein (gij106445691 gblAAG21341.1 AF274313_1 (AF274313) soggy precursor [Mus musculus]) (SEQ ID 5 NO:70) on line 4. Table 13E depicts a ClustalW alignment of FCTR13 against proteins from a public database. Based on this alignment, black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties); grayed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); 0 non-highlighted amino acid residues can potentially be mutated to a much broader extent without altering structure or function. Table 13E. ClustalW alignment of the FCTR13. 10 20 30 40 50 60 5 FCTR13 ---------- RHPI ISALVT FI TI S * L LT LS lGRF] Line 2 MGEASPPAPARRH ILL STLF:P ES$ L LT (S G RF Line 3 ----------- C F PLA L QNTS FL R S Line 4 ----------- CR L R PLA F ,4S FL L R eLS 0 70 80 90 100 110 120 FCTR13 G1iF F G QR Line 2 G0 G, D. LFAMFG it l Qg NLSQIKTNTGLS Line 3 D D D Line 4 SDD D 130 140 150 160 170 180 FCTR13 Q EG'F -ALS FLHPi AFWI SH 0 Line 2 Q EGSF DL PRMEEKEALjPTQ T SF LHPEVAFWII H Line 3 E S ER-P PKVEAKEPP P L PR- AFWI TQ Line 4 E E R ----------- --------------- 190 200 210 220 230 240 5 .... I ....I ....I ....I . ... I . . . . I . . ..I . . . . I . . FCTR13 HNVL R S 3(HETR]EKHLQITQGSS Line 3 PDVQ3 (I]IEKH I R S HIP QARPVR Line 4 0 250 260 270 280 290 300 FCTR13 SYRGWDRGAPASWVGVWVSVSKGLAHGPTGLGLEPGSSGIDFVNAFMEGYSNQNIMVLVQ Line 2 ---- ---------------- 39 WO 01/46231 PCT/USOO/34898 Line 3 ------------------------ Line 4 ------ - --- -- 310 320 5 ... FCTR13 KRIHRPMQQNREARNKATQDLI S (SEQ ID NO:26) Line 2 -------------- THILLYII-- L (SEQ ID NO:68) Line 3 -------------- THFLYIR-- L (SEQ ID NO:69) Line 4 ------- - --- (SEQ ID NO:70) 0 From these analyses, it is seen that the FCTR13 AL121723_A A nucleic acid and protein have a strong similarity with human soggy-1 protein. The nucleic acids and proteins of the invention are potentially useful in the treatment 5 of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. FCTR14 (AL121756_A) The novel nucleic acid encoding a novel secreted protein is shown in Table 14A. This sequence contains an initiation codon at the 5' end, and a TGA stop codon was identified at 0 the 3' end indicating that this sequence is a coding sequence. The start and stop codons are shown in bold type. This sequence originates in chromosome 20 clone RP4-726C3. Table 14A. FCTR14 (AL121756_A) nucleotide sequence (SEQ ID NO:27). atgctgcggatcctgtgcctggcactctgcagcctgctgactggcacgcgagctgaccctggggcactg ctgcggttgggcatggacatcatgaaccgtgaggtccagagcgccatggatgagagtcatatcctggag 5 aagatggcagccgaggcaggcaagaaacagccagggatgaaacctatcaagggcatcaccaatttgaag gtgaaggatgtccagctgcccgtcatcacactgaactttgtacctggagtgggcatcttccaatgtgtg tccacaggcatgaccgtcactggcaagagcttcatgggagggaacatggagatcatcgtggccctgaac atcacagccaccaaccggcttctgcgggatgaggagacaggcctccccgtgttcaagagtgagggctgt gaggtcatcctggtcaatgtgaagactaacctgcctagcaacatgctccccaagatggtcaacaagttc 0 ctggacagcaccctgcacaaagtcctccctgggctgatgtgtcccgccatcgatgcagtcctggtgtat gtgaacaggaagtggaccaacctcagtgaccccatgcctgtgggccagatgggcaccgtcaaatatgtt ctgatgtccgcaccagccaccacagccagctacatccaactggacttcagtcctgtggtgcagcagcaa aagggcaaaaccatcaagcttgctgatgccggggaggccctcacgttccctgagggttatgccaaaggc tcgtcgcagctgctgctcccagccaccttcctctctgcagagcttgcccttctgcagaagtcctttcat 5 gtgaatatccaggatacaatgattggtgagctgcccccacaaaccaccaagaccctggctcgcttcatt cctgaagtggctgtagcttatcccaagtcaaagcccttgacgacccagatcaagataaagaagcctccc aaggtcactatgaagacaggcaagagcctgctgcacctccacagcaccctggagatgttcgcagctcgg tggcggagcaaggctccaatgtcccttttctcctagaagtgcacttcaatctgaaggtccagtactca gtgcatgagaaccagctgcagatggccacttctttggacagattactgagcttgtcccggaagtcctca 0 tcgattggcaacttcaatgagagggaattaactggcttcatcaccagctatctcgaagaagcctacatc ccagttgtcaatgatgtgcttcaagtggggctcccactcccggactttctggccatgaattacaacctg gctgagctggacatagtagagcttgggggcatcatggaacctgccgacatatga The encoded protein is presented using the one-letter code in Table 14B. The protein 5 has a moderate probability of being sorted to the plasma membrane. A signal peptide most likely is cleaved between residues 18 and 19, i.e., at the dash in the amino acid sequence TRA DPG. 40 WO 01/46231 PCT/USOO/34898 Table 14B. Encoded FCTR14 protein sequence (SEQ ID NO:28). MLRILCLALCSLLTGTRADPGALLRLGMDIMNREVQSAMDESHILEKMAAEAGKKQPGMKPIKGITNLK VKDVQLPVITLNFVPGVGIFQCVSTGMTVTGKSFMGGNMEIIVALNITATNRLLRDEETGLPVFKSEGC EVILVNVKTNLPSNMLPKMVNKFLDSTLHKVLPGLMCPAIDAVLVYVNRKWTNLSDPMPVGQMGTVKYV 5 LMSAPATTASYIQLDFSPVVQQQKGKTIKLADAGEALTFPEGYAKGSSQLLLPATFLSAELALLQKSFH VNIQDTMIGELPPQTTKTLARFIPEVAVAYPKSKPLTTQIKIKKPPKVTMKTGKSLLHLHSTLEMFAAR WRSKAPMSLFLLEVHFNLKVQYSVHENQLQMATSLDRLLSLSRKS S S IGNFNERELTGFITSYLEEAYI PVVNDVLQVGL PL PDFLAMNYNLAELDIVELGGIME PADI 0 A BLASTN search of sequence databases for the FCTR14 nucleic acid sequence identified significant similarities to the human genomic clone HSDJ726C3, isolated from human DNA sequence from clone RP4-726C3 on chromosome 20. In a BLASTX comparison, it was found that the full FCTR14 amino acid sequence has 130 of 391 residues (33%), are identical to, and 229 of 391 residues (58%) positive with, rat potential ligand 5 binding protein RY2G5 having a total of 409 amino acid residues (SPTREMBL ACC:Q05704) (SEQ ID NO:71). The BLASTX alignment is shown in Table 14C. Table 14C. BLASTX alignment of FCTR14 >ptnr:SPTREMBL-ACC:Q05704 POTENTIAL LIGAND-BINDING PROTEIN RY2G5 - RATTUS NORVEGICUS (RAT), 470 aa (fragment). o CC -!- TISSUE SPECIFICITY: SUBREGIONS OF THE OLFACTORY MUCOSA. Score 579 (203.8 bits), Expect = 2.0e-55, P = 2.0e-55 Identities = 130/391 (33%), Positives = 229/391 (58%), Frame = +1 5 Query: 175 MKPIKGITNLKVKDVQLPVITLNFVPGVGIFQCVSTGMTVTGKSFMGGNMEIIVALNITA 354 + ++1I1 I1++ ++ 1 l +++ +l111++ + ] + + I1I +] ++1 I +11 i Sbjct: 73 LSTVQGITGLRIVELTLPRVSVRLLPGVGVYLSLYTRVAINGKSLIGF-LDIAVEVNITA 131 Query: 355 TNRLLRDEETGLPVFKSEGCEVILVNVKTNLPSNMLPKMVNKFLDSTLHKVLPGLMCPAI 534 0 I i I I i 1 1+ +1 +1 I +11 +1+ ++ I III L+H1 + Sbjct: 132 KVRLTMDR-TGYPRLVIERCDTLLGGIKVKLLRGLLPNLVDNLVNRVLANVLPDLLCPIV 190 Query: 535 DAVLVYVNRKWTNLSDPMPVGQMGTVKYVLMSAPATTASYIQLDFSPVVQQQKGKTIKLA 714 1 II 11 + + +1+1 +1+ +1 I I +++.11 + +1 + 1 1 5 Sbjct: 191 DVVLGLVNDQLGLVDSLVPLGILGSVQYTFSSLPLVTGEFLELDLNTLVGEAGGDLIDYP 250 Query: 715 DAGEALT----FPEGYAKG---SSQLLLPATFLSAELALLQK~--SFHVNIQDTMIGELPP 867 1+ ll 1 +111 + I II1 + 1 +111 + ++I I I +111 Sbjct: 251 LGRPAMLPRPQMPELPPMGDNTNSQLAISANFLSSVLTMLQKQGALDIDITDGMFEDLPP 310 0 Query: 868 QTTKTLARFIPEVAVAYPKSKPLTTQIKIKKPPKVTMKTGKSLLHLHSTLEMFAARWRSK 1047 i I1 11+1 11+1+111 +1++ 1 I I1++ 1+1+ + + II+ ++ + Sbjct: 311 LTTSTLGALIPKVFQQYPESRPLTIRIQVPNPPTVTLQKDKALVKVFATSEVVVSQ-PND 369 5 Query: 1048 APMSLFLLEVHFNLKVQYSVHENQLQMATSLDRLLSLSRKSSSIGNFNERELTGFITSYL 1227 ++ i++I +1 +11 ++1 + 11+ 11+ ++)++III+ I + Sbjct: 370 VETTICLIDVDTDLLASFSVEGDKLMIDAKLDKT-SLNLRTSNVGNFDVFILEMLVEKIF 428 Query: 1228 EEAYIPVVNDVLQVGLPLPDFLAMNYNLAELDIVE 1332 0 + 1++1 +1 +1 1+111 I ++++ 1++l++] Sbjct: 429 DLAFMPAMNAILGSGVPLPKILNIDFSNADIDVLE 463 A multiple sequence alignment for FCTR14 AL121756_A is given in Table 14D, with the protein of the invention being shown on line 3, in a ClustalW analysis comparing the 5 protein of the invention with related protein sequences. Table 14D depicts a ClustalW 41 WO 01/46231 PCT/USOO/34898 alignment of FCTR13 with proteins from the public database. The alignment is presented against Q05704 - POTENTIAL LIGAND-BINDING PROTEIN RY2G5 (FRAGMENT) (SEQ ID NO:71) and Q05701 - POTENTIAL LIGAND-BINDING PROTEIN RYA3 (SEQ ID NO:72). Based on this alignment, black outlined amino acid residues indicate regions of 5 conserved sequence (i.e., regions that may be required to preserve structural or functional properties); grayed amino acid residues can be mutated to a residue with comparable steric and/or chemical properties without altering protein structure or function (e.g. L to V, I, or M); non-highlighted amino acid residues can potentially be mutated to a much broader extent without altering structure or function. 0 TABLE 14D ClustalW alignment of FCTR14 (AL121756 _A) protein Q05704 RY2G5 RAT RLHRRE RPGE PAVZGi -BbANGILnGGGf ---------- Q05701_RYA3 RAT AL121756_A --- ILRLCfCSLTGTRADIURLHMDMNRVQSfMDEIHJLEK 5 Q05704 RY2G5 RAT --- G0 GVL IVIGEEILH I Q RUVRVRYI Q05701 RYA3_RAT SVNQ A----L!SYEFLiEE tSE TE1]KQI AL121756 A MAAEA1KKQTI----------NK- 1 Q05704 RY2G5_RAT Y 0 Q05701RYA3 RAT HJiKI;LHISGP" VQLIQJVSSAGSP-RITiIII -- HISJTI AL121756_ AIRVJFGN9IA NI~E~LVFSGFVIN,-I~~ Q05704 RY2G5_RAT NlDNNR VIAE0II VGItm DQEL VDIQEFSE Q05701 RYA3 RAT TP FGEQ EC VUSESVE LATL PErFATI 5 AL121756 A NMEEMNKFLDSEHMAI@AUVYRKWTN -S T Q05704 RY2G5_RAT W Q05701_RYA3_RAT *ISNQIETUKS AL121756 A 1ATjSEQEFSvQQQKKTKLADrG ------- EAI;TFJEGYAKGSL T 0 Q05704 RY2G5 RAT lSMTA4Q IT GMFEDA SJGKIFQQEER RIQIPNE Q05701_RYA3RAT NTJIGTN B PEMVPRNIT0AALGKLH PGQHILSTRJMISI AL121756 A AT 5 Q05704 RY2G5 RAT T Q05701 RYA3 RAT E AL121756A KMKTGISI HHSla4FAARWRSKIMSLEHFNKVQYHENQIQMAT Q05704 RY2G5 RAT -TMNIRTNVVFIMLVEfDAM 0jS.DIA 0 Q05701 RYA3 RAT F AL121756 A - , DL Q05704 RY2GS RAT IDLES--- (SEQ ID NO:71) Q05701_RYA3 RAT ENVEVP- (SEQ ID NO:72) 5 AL121756_A ALGGIMEP*DI (SEQ ID IO:28) Finally, FCTR14 was found to have high homology to the domains shown in Table 14D. Table 141): CD domain analysis of FCTR14 Sequences producing significant alignments: (ie v e BPJ/LBP/CETP C-terminal domain; Bactericidal permeability-incr ... 72.8 4e-14 BPJ/LBP/CETP N-terminal domain; Bactericidal permeability-incr ... 60.8 le-lO 42 WO 01/46231 PCT/USOO/34898 LBPBPICETP, LBP / BPI / CETP family 45.8 5e-06 The FCTR14 nucleic acids and proteins of the invention are potentially useful in the treatment of cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders. 5 FCTRX Nucleic Acids and Polypeptides One aspect of the invention pertains to isolated nucleic acid molecules that encode FCTRX polypeptides or biologically-active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify FCTRX encoding nucleic acids (e.g., FCTRX mRNAs) and fragments for use as PCR primers for the 0 amplification and/or mutation of FCTRX nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double 5 stranded DNA. An FCTRX nucleic acid can encode a mature FCTRX polypeptide. As used herein, a "mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide or precursor form or proprotein. The naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full length 0 gene product, encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an open reading frame described herein. The product "mature" form arises, again by way of nonlimiting example, as a result of one or more naturally occurring processing steps as they may take place within the cell, or host cell, in which the gene product arises. Examples of such processing steps leading to a "mature" form 5 of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an open reading frame, or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a 0 mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+1 to residue N remaining. Further as used herein, a "mature" form of a polypeptide or protein may arise from a step of post-translational modification other than a 43 WO 01/46231 PCT/USOO/34898 proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristoylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them. 5 The term "probes", as utilized herein, refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use. Probes are used in the detection of identical, similar, or complementary nucleic acid sequences. Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and 0 much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies. The term "isolated" nucleic acid molecule, as utilized herein, is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. 5 Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3'-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated FCTRX nucleic acid molecules can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.). Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or of chemical precursors or other chemicals when chemically 5 synthesized. A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or a complement of this aforementioned nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a 0 portion of the nucleic acid sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29 as a hybridization probe, FCTRX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et al., (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2nd Ed., Cold Spring Harbor Laboratory 44 WO 01/46231 PCT/USOO/34898 Press, Cold Spring Harbor, NY, 1989; and Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993.) A nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard 5 PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to FCTRX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer. As used herein, the term "oligonucleotide" refers to a series of linked nucleotide 0 residues, which oligonucleotide has a sufficient number of nucleotide bases to be used in a PCR reaction. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue. Oligonucleotides comprise portions of a nucleic acid sequence having about 10 nt, 50 nt, or 5 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes. 0 In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of an FCTRX polypeptide). A nucleic acid molecule that is 5 complementary to the nucleotide sequence shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, is one that is sufficiently complementary to the nucleotide sequence shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, that it can hydrogen bond with little or no mismatches to the nucleotide sequence shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, thereby fonning a stable duplex. 0 As used herein, the term "complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding" means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct 45 WO 01/46231 PCT/USOO/34898 or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates. 5 Fragments provided herein are defined as sequences of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, respectively, and are at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid 0 sequence of choice. Derivatives are nucleic acid sequences or amino acid sequences formed from the native compounds either directly or by modification or partial substitution. Analogs are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound but differs from it in respect to certain components or side chains. Analogs may be synthetic or from a different evolutionary origin and may have a 5 similar or opposite metabolic activity compared to wild type. Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species. Derivatives and analogs may be full length or other than full length, if the derivative or analog contains a modified nucleic acid or amino acid, as described below. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, o molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of 5 hybridizing to the complement of a sequence encoding the aforementioned proteins under stringent, moderately stringent, or low stringent conditions. See e.g. Ausubel, et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993, and below. A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or 0 amino acid level as discussed above. Homologous nucleotide sequences encode those sequences coding for isoforms of FCTRX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for an FCTRX polypeptide of 46 WO 01/46231 PCT/USOO/34898 species other than humans, including, but not limited to: vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms. Homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein. A homologous nucleotide sequence 5 does not, however, include the exact nucleotide sequence encoding human FCTRX protein. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, as well as a polypeptide possessing FCTRX biological activity. Various biological activities of the FCTRX proteins are described below. 0 An FCTRX polypeptide is encoded by the open reading frame ("ORF") of an FCTRX nucleic acid. An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide. A stretch of nucleic acids comprising an ORF is uninterrupted by a stop codon. An ORF that represents the coding sequence for a full protein begins with an ATG "start" codon and terminates with one of the three "stop" codons, namely, TAA, TAG, or 5 TGA. For the purposes of this invention, an ORF may be any part of a coding sequence, with or without a start codon, a stop codon, or both. For an ORF to be considered as a good candidate for coding for a bonafide cellular protein, a minimum size requirement is often set, e.g., a stretch of DNA that would encode a protein of 50 amino acids or more. The nucleotide sequences determined from the cloning of the human FCTRX genes 0 allows for the generation of probes and primers designed for use in identifying and/or cloning FCTRX homologues in other cell types, e.g. from other tissues, as well as FCTRX homologues from other vertebrates. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 5 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29; or an anti-sense strand nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29; or of a naturally occurring mutant of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. Probes based on the human FCTRX nucleotide sequences can be used to detect 0 transcripts or genomic sequences encoding the same or homologous proteins. In various embodiments, the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis express an FCTRX protein, such as by measuring a level of an FCTRX-encoding nucleic acid 47 WO 01/46231 PCT/USOO/34898 in a sample of cells from a subject e.g., detecting FCTRX mRNA levels or determining whether a genomic FCTRX gene has been mutated or deleted. "A polypeptide having a biologically-active portion of an FCTRX polypeptide" refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a 5 polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a "biologically active portion of FCTRX" can be prepared by isolating a portion of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, that encodes a polypeptide having an FCTRX biological activity (the biological activities of the FCTRX proteins are described below), 0 expressing the encoded portion of FCTRX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of FCTRX. FCTRX Nucleic Acid and Polypeptide Variants The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences shown in SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 5 and 29, due to degeneracy of the genetic code and thus encode the same FCTRX proteins as that encoded by the nucleotide sequences shown in SEQ ID NO NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. 0 In addition to the human FCTRX nucleotide sequences shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the FCTRX polypeptides may exist within a population (e.g., the human population). Such genetic polymorphism in the FCTRX genes may exist among individuals within a population 5 due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding an FCTRX protein, preferably a vertebrate FCTRX protein. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the FCTRX genes. Any and all such nucleotide variations and resulting amino acid polymorphisms in the FCTRX polypeptides, 0 which are the result of natural allelic variation and that do not alter the functional activity of the FCTRX polypeptides, are intended to be within the scope of the invention. Moreover, nucleic acid molecules encoding FCTRX proteins from other species, and thus that have a nucleotide sequence that differs from the human sequence of SEQ ID NOS:1, 48 WO 01/46231 PCT/USOO/34898 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the FCTRX cDNAs of the invention can be isolated based on their homology to the human FCTRX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a 5 hybridization probe according to standard hybridization techniques under stringent hybridization conditions. Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 0 13, 15, 17, 19, 21, 23, 25, 27, and 29. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, or 2000 or more nucleotides in length. In yet another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 5 60% homologous to each other typically remain hybridized to each other. Homologs (i.e., nucleic acids encoding FCTRX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning. 0 As used herein, the phrase "stringent hybridization conditions" refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5C lower than the 5 thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium. Typically, stringent conditions will be those in 0 which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30'C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60C for longer probes, primers and 49 WO 01/46231 PCT/USOO/34898 oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide. Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. 5 (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA 0 at 65'C, followed by one or more washes in 0.2X SSC, 0.01% BSA at 50'C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequences of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in 5 nature (e.g., encodes a natural protein). In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency 0 hybridization conditions are hybridization in 6X SSC, 5X Denhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55'C, followed by one or more washes in IX SSC, 0.1% SDS at 37 0 C. Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel ,et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990; GENE TRANSFER AND 5 EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY. In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequences of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions 0 are hybridization in 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40'C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS at 50 C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, 50 WO 01/46231 PCT/USOO/34898 e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981. Proc Natl Acad Sci USA 78: 6789-6792. 5 Conservative Mutations In addition to naturally-occurring allelic variants of FCTRX sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences of SEQ ID NO NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, thereby leading to changes in the amino acid sequences of the encoded 0 FCTRX proteins, without altering the functional ability of said FCTRX proteins. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the FCTRX proteins without altering their biological activity, 5 whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the FCTRX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known within the art. Another aspect of the invention pertains to nucleic acid molecules encoding FCTRX 0 proteins that contain changes in amino acid residues that are not essential for activity. Such FCTRX proteins differ in amino acid sequence from SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 45% homologous to the amino acid 5 sequences of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; more preferably at least about 70% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; still more preferably at least about 80% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 0 20, 22, 24, 26, 28 and 30; even more preferably at least about 90% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; and most preferably at least about 95% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. An isolated nucleic acid molecule encoding an FCTRX protein homologous to the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, can be created 51 WO 01/46231 PCT/USOO/34898 by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. 5 Mutations can be introduced into SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side 0 chain. Families of amino acid residues having similar side chains have been defined within the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, 5 tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted non-essential amino acid residue in the FCTRX protein is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of an FCTRX coding sequence, such as by saturation mutagenesis, 0 and the resultant mutants can be screened for FCTRX biological activity to identify mutants that retain activity. Following mutagenesis of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16,18, 20, 22, 24, 26, 28 and 30, the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined. The relatedness of amino acid families may also be determined based on side chain 5 interactions. Substituted amino acids may be fully conserved "strong" residues or fully conserved "weak" residues. The "strong" group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other. Likewise, the "weak" group of conserved residues may be any one 0 of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, VLIM, HFY, wherein the letters within each group represent the single letter amino acid code. In one embodiment, a mutant FCTRX protein can be assayed for (i) the ability to form protein:protein interactions with other FCTRX proteins, other cell-surface proteins, or biologically-active portions thereof, (ii) complex formation between a mutant FCTRX protein 52 WO 01/46231 PCT/USOO/34898 and an FCTRX ligand; or (iii) the ability of a mutant FCTRX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g. avidin proteins). In yet another embodiment, a mutant FCTRX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release). 5 Antisense Nucleic Acids Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or fragments, analogs or derivatives thereof. An "antisense" nucleic acid comprises a nucleotide 0 sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an nRNA sequence). In specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire FCTRX coding strand, or to only a portion thereof. Nucleic acid molecules 5 encoding fragments, homologs, derivatives and analogs of an FCTRX protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; or antisense nucleic acids complementary to an FCTRX nucleic acid sequence of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, are additionally provided. In one embodiment, an antisense nucleic acid molecule is antisense to a "coding 0 region" of the coding strand of a nucleotide sequence encoding an FCTRX protein. The term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues. In another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the FCTRX protein. The term "noncoding region" refers to 5' and 3' sequences 5 which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions). Given the coding strand sequences encoding the FCTRX protein disclosed herein, antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing. The antisense nucleic acid molecule can be complementary 0 to the entire coding region of FCTRX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of FCTRX mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of FCTRX mRNA. An antisense oligonucleotide can be, for example, 53 WO 01/46231 PCT/USOO/34898 about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally-occurring nucleotides or 5 variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex fonned between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used). Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, 0 xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl 2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, .5 beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5'oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the 0 antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection). The antisense nucleic acid molecules of the invention are typically administered to a 5 subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an FCTRX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation). The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in 0 the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface 54 WO 01/46231 PCT/USOO/34898 (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens). The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient nucleic acid molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong 5 pol II or pol III promoter are preferred. In yet another embodiment, the antisense nucleic acid molecule of the invention is an a-anomeric nucleic acid molecule. An ca-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual p-units, the strands run parallel to each other. See, e.g., Gaultier, et al., 1987. Nucl. Acids Res. 15: 0 6625-6641. The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (see, e.g., Inoue, et al. 1987. Nucl. Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (see, e.g., Inoue, et al., 1987. FEBS Lett. 215: 327-330. Ribozymes and PNA Moieties Nucleic acid modifications include, by way of non-limiting example, modified bases, 5 and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In one embodiment, an antisense nucleic acid of the invention is a ribozyme. ZO Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591) can be used to catalytically cleave FCTRX mRNA transcripts to thereby inhibit translation of FCTRX mRNA. A ribozyme having 5 specificity for an FCTRX-encoding nucleic acid can be designed based upon the nucleotide sequence of an FCTRX cDNA disclosed herein (i.e., SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an FCTRX-encoding mRNA. See, e.g., U.S. Patent 30 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al. FCTRX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al., (1993) Science 261:1411-1418. Alternatively, FCTRX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the FCTRX nucleic acid (e.g., the 55 WO 01/46231 PCT/USOO/34898 FCTRX promoter and/or enhancers) to form triple helical structures that prevent transcription of the FCTRX gene in target cells. See, e.g., Helene, 1991. Anticancer Drug Des. 6: 569-84; Helene, et al. 1992. Ann. N.Y Acad. Sci. 660: 27-36; Maher, 1992. Bioassays 14: 807-15. In various embodiments, the FCTRX nucleic acids can be modified at the base moiety, 5 sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al., 1996. Bioorg Med Chemn 4: 5-23. As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g., DNA mimics) in which the deoxyribose phosphate backbone is replaced by 0 a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al., 1996. supra; Perry-O'Keefe, et al., 1996. Proc. Natl. Acad. Sci. USA 93: 14670-14675. 5 PNAs of FCTRX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of FCTRX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in 0 combination with other enzymes, e.g., Si nucleases (see, Hyrup, et al., 1996.supra); or as probes or primers for DNA sequence and hybridization (see, Hyrup, et al., 1996, supra; Perry-O'Keefe, et al., 1996. supra). In another embodiment, PNAs of FCTRX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the 5 formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of FCTRX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA 30 chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (see, Hyrup, etal., 1996. supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et al., 1996. supra and Finn, et al., 1996. Nucl Acids Res 24: 3357-3363. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and 56 WO 01/46231 PCT/USOO/34898 modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al., 1989. Nucl Acid Res 17: 5973-5988. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., 5 Finn, et al., 1996. supra. Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al., 1975. Bioorg. Med. Chem. Lett. 5: 1119-11124. In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across 10 the cell membrane (see, e.g., Letsinger, et al., 1989. Proc. Nati. Acad. Sci. U.S.A. 86: 6553-6556; Lemaitre, et al., 1987. Proc. Nati. Acad. Sci. 84: 648-652; PCT Publication No. W088/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134). In addition, oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al., 1988. BioTechniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988. [5 Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like. FCTRX Polypeptides A polypeptide according to the invention includes a polypeptide including the amino 0 acid sequence of FCTRX polypeptides whose sequences are provided in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residues shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, while still encoding a protein that maintains its FCTRX activities and physiological functions, or a functional 5 fragment thereof. In general, an FCTRX variant that preserves FCTRX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting one or more residues 0 from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above. 57 WO 01/46231 PCT/USOO/34898 One aspect of the invention pertains to isolated FCTRX proteins, and biologically active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-FCTRX antibodies. In one embodiment, native FCTRX proteins can be isolated from cells or tissue sources by an 5 appropriate purification scheme using standard protein purification techniques. In another embodiment, FCTRX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, an FCTRX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques. An "isolated" or "purified" polypeptide or protein or biologically-active portion thereof 0 is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the FCTRX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of FCTRX proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced. In 5 one embodiment, the language "substantially free of cellular material" includes preparations of FCTRX proteins having less than about 30% (by dry weight) of non-FCTRX proteins (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-FCTRX proteins, still more preferably less than about 10% of non-FCTRX proteins, and most preferably less than about 5% of non-FCTRX proteins. When the FCTRX protein or !0 biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the FCTRX protein preparation. The language "substantially free of chemical precursors or other chemicals" includes 5 preparations of FCTRX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. In one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of FCTRX proteins having less than about 30% (by dry weight) of chemical precursors or non-FCTRX chemicals, more preferably less than about 20% chemical precursors or 0 non-FCTRX chemicals, still more preferably less than about 10% chemical precursors or non-FCTRX chemicals, and most preferably less than about 5% chemical precursors or non-FCTRX chemicals. Biologically-active portions of FCTRX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the 58 WO 01/46231 PCT/USOO/34898 FCTRX proteins (e.g., the amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30) that include fewer amino acids than the full-length FCTRX proteins, and exhibit at least one activity of an FCTRX protein. Typically, biologically-active portions comprise a domain or motif with at least one activity of the FCTRX protein. A 5 biologically-active portion of an FCTRX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length. Moreover, other biologically-active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native FCTRX protein. 0 In an embodiment, the FCTRX protein has an amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. In other embodiments, the FCTRX protein is substantially homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, and retains the functional activity of the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, yet differs in amino acid sequence due to 5 natural allelic variation or mutagenesis, as described in detail, below. Accordingly, in another embodiment, the FCTRX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, and retains the functional activity of the FCTRX proteins of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30. 0 Determining Homology Between Two or More Sequences To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at 5 corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity"). 0 The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch, 1970. JMol Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension 59 WO 01/46231 PCT/USOO/34898 penalty of 0.3, the coding region of the analogous nucleic acid sequences referred to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence shown in SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. 5 The term "sequence identity" refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of .0 nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term "substantial identity" as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 5 percent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region. Chimeric and Fusion Proteins The invention also provides FCTRX chimeric or fusion proteins. As used herein, an :0 FCTRX "chimeric protein" or "fusion protein" comprises an FCTRX polypeptide operatively linked to a non-FCTRX polypeptide. An "FCTRX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to an FCTRX protein (SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30), whereas a "non-FCTRX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to a protein that is not substantially 5 homologous to the FCTRX protein, e.g., a protein that is different from the FCTRX protein and that is derived from the same or a different organism. Within an FCTRX fusion protein the FCTRX polypeptide can correspond to all or a portion of an FCTRX protein. In one embodiment, an FCTRX fusion protein comprises at least one biologically-active portion of an FCTRX protein. In another embodiment, an FCTRX fusion protein comprises at least two 0 biologically-active portions of an FCTRX protein. In yet another embodiment, an FCTRX fusion protein comprises at least three biologically-active portions of an FCTRX protein. Within the fusion protein, the term "operatively-linked" is intended to indicate that the FCTRX polypeptide and the non-FCTRX polypeptide are fused in-frame with one another. The 60 WO 01/46231 PCT/USOO/34898 non-FCTRX polypeptide can be fused to the N-terminus or C-terminus of the FCTRX polypeptide. In one embodiment, the fusion protein is a GST-FCTRX fusion protein in which the FCTRX sequences are fused to the C-terminus of the GST (glutathione S-transferase) 5 sequences. Such fusion proteins can facilitate the purification of recombinant FCTRX polypeptides. In another embodiment, the fusion protein is an FCTRX protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of FCTRX can be increased through use of a heterologous 0 signal sequence. In yet another embodiment, the fusion protein is an FCTRX-immunoglobulin fusion protein in which the FCTRX sequences are fused to sequences derived from a member of the immunoglobulin protein family. The FCTRX-immunoglobulin fusion proteins of the invention can be incorporated into pharmaceutical compositions and administered to a subject 5 to inhibit an interaction between an FCTRX ligand and an FCTRX protein on the surface of a cell, to thereby suppress FCTRX-mediated signal transduction in vivo. The FCTRX immunoglobulin fusion proteins can be used to affect the bioavailability of an FCTRX cognate ligand. Inhibition of the FCTRX ligand/FCTRX interaction may be useful therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. 0 promoting or inhibiting) cell survival. Moreover, the FCTRX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-FCTRX antibodies in a subject, to purify FCTRX ligands, and in screening assays to identify molecules that inhibit the interaction of FCTRX with an FCTRX ligand. An FCTRX chimeric or fusion protein of the invention can be produced by standard 5 recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In 0 another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al. (eds.) CURRENT PROTOCOLS IN MOLECULAR 61 WO 01/46231 PCT/USOO/34898 BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). An FCTRX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the FCTRX protein. 5 FCTRXAgonists and Antagonists The invention also pertains to variants of the FCTRX proteins that function as either FCTRX agonists (i.e., mimetics) or as FCTRX antagonists. Variants of the FCTRX protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the FCTRX protein). An agonist of the FCTRX protein can retain substantially the same, or a subset of, 0 the biological activities of the naturally occurring fonn of the FCTRX protein. An antagonist of the FCTRX protein can inhibit one or more of the activities of the naturally occurring form of the FCTRX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the FCTRX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one 5 embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring fonn of the FCTRX proteins. Variants of the FCTRX proteins that function as either FCTRX agonists (i.e., mimetics) or as FCTRX antagonists can be identified by screening combinatorial libraries of 0 mutants (e.g., truncation mutants) of the FCTRX proteins for FCTRX protein agonist or antagonist activity. In one embodiment, a variegated library of FCTRX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of FCTRX variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a 5 degenerate set of potential FCTRX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of FCTRX sequences therein. There are a variety of methods which can be used to produce libraries of potential FCTRX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, 0 and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential FCTRX sequences. Methods for synthesizing degenerate oligonucleotides are well-known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; 62 WO 01/46231 PCT/USOO/34898 Itakura, et al., 1984. Annu. Rev. Biochem. 53: 323; Itakura, et al., 1984. Science 198: 1056; Ike, et al., 1983. Nucl. Acids Res. 11: 477. Polypeptide Libraries In addition, libraries of fragments of the FCTRX protein coding sequences can be used 5 to generate a variegated population of FCTRX fragments for screening and subsequent selection of variants of an FCTRX protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an FCTRX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double-stranded 0 DNA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Si nuclease, and ligating the resulting fragment library into an expression vector. By this method, expression libraries can be derived which encodes N-terminal and internal fragments of various sizes of the FCTRX proteins. 5 Various techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of FCTRX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large 0 gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the 5 libraries, can be used in combination with the screening assays to identify FCTRX variants. See, e.g., Arkin and Yourvan, 1992. Proc. Nati. Acad. Sci. USA 89: 7811-7815; Delgrave, et al., 1993. Protein Engineering 6:327-33 1. Anti-FCTRX Antibodies The invention encompasses antibodies and antibody fragments, such as Fab or (Fab)2, 0 that bind immunospecifically to any of the FCTRX polypeptides of said invention. An isolated FCTRX protein, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind to FCTRX polypeptides using standard techniques for polyclonal and monoclonal antibody preparation. The full-length FCTRX proteins can be 63 WO 01/46231 PCT/USOO/34898 used or, alternatively, the invention provides antigenic peptide fragments of FCTRX proteins for use as immunogens. The antigenic FCTRX peptides comprises at least 4 amino acid residues of the amino acid sequence shown in SEQ ID NO NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, and encompasses an epitope of FCTRX such that an antibody raised 5 against the peptide forms a specific immune complex with FCTRX. Preferably, the antigenic peptide comprises at least 6, 8, 10, 15, 20, or 30 amino acid residues. Longer antigenic peptides are sometimes preferable over shorter antigenic peptides, depending on use and according to methods well known to someone skilled in the art. In certain embodiments of the invention, at least one epitope encompassed by the 0 antigenic peptide is a region of FCTRX that is located on the surface of the protein (e.g., a hydrophilic region). As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation (see, e.g., Hopp and Woods, 1981. Proc. Nat. Acad. Sci. USA 5 78: 3824-3828; Kyte and Doolittle, 1982. J. Mol. Biol. 157: 105-142, each incorporated herein by reference in their entirety). As disclosed herein, FCTRX protein sequences of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, or derivatives, fragments, analogs or homologs thereof, may be utilized as immunogens in the generation of antibodies that immunospecifically-bind these 0 protein components. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically-active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically-binds (immunoreacts with) an antigen, such as FCTRX. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab and F(ab')2 fragments, and an Fab expression library. In a specific embodiment, :5 antibodies to human FCTRX proteins are disclosed. Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies to an FCTRX protein sequence of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, or a derivative, fragment, analog or homolog thereof. Some of these proteins are discussed below. For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, 0 goat, mouse or other mammal) may be immunized by injection with the native protein, or a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, recombinantly-expressed FCTRX protein or a chemically-synthesized FCTRX polypeptide. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, 64 WO 01/46231 PCT/USOO/34898 Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, etc.), human adjuvants such as Bacille Calmette-Guerin and Corynebacteriun parvum, or similar immunostimulatory agents. If desired, the antibody molecules directed 5 against FCTRX can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction. The term "monoclonal antibody" or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of FCTRX. A monoclonal 0 antibody composition thus typically displays a single binding affinity for a particular FCTRX protein with which it immunoreacts. For preparation of monoclonal antibodies directed towards a particular FCTRX protein, or derivatives, fragments, analogs or homologs thereof, any technique that provides for the production of antibody molecules by continuous cell line culture may be utilized. Such techniques include, but are not limited to, the hybridoma 5 technique (see, e.g., Kohler & Milstein, 1975. Nature 256: 495-497); the trioma technique; the human B-cell hybridoma technique (see, e.g., Kozbor, et al., 1983. Immunol. Today 4: 72) and the EBV hybridoma technique to produce human monoclonal antibodies (see, e.g., Cole, et al., 1985. In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the invention and may be 0 produced by using human hybridomas (see, e.g., Cote, et al., 1983. Proc Nati Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Barr Virus in vitro (see, e.g., Cole, et al., 1985. In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Each of the above citations is incorporated herein by reference in their entirety. According to the invention, techniques can be adapted for the production of 5 single-chain antibodies specific to an FCTRX protein (see, e.g., U.S. Patent No. 4,946,778). In addition, methods can be adapted for the construction of Fab expression libraries (see, e.g., Huse, et al., 1989. Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for an FCTRX protein or derivatives, fragments, analogs or homologs thereof. Non-human antibodies can be "humanized" by 0 techniques well known in the art. See, e.g., U.S. Patent No. 5,225,539. Antibody fragments that contain the idiotypes to an FCTRX protein may be produced by techniques known in the art including, but not limited to: (i) an F(ab')2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(ab')2 65 WO 01/46231 PCT/USOO/34898 fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent; and (iv) F, fragments. Additionally, recombinant anti-FCTRX antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made 5 using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in International Application No. PCT/US86/02269; European Patent Application No. 184,187; European Patent Application No. 171,496; European Patent Application No. 173,494; PCT International 0 Publication No. WO 86/01533; U.S. Patent No. 4,816,567; U.S. Pat. No. 5,225,539; European Patent Application No. 125,023; Better, et al., 1988. Science 240: 1041-1043; Liu, et al., 1987. Proc. Natl. Acad. Sci. USA 84: 3439-3443; Liu, et al., 1987. J Immunol. 139: 3521-3526; Sun, et al., 1987. Proc. Natl. Acad. Sci. USA 84: 214-218; Nishimura, et al., 1987. Cancer Res. 47: 999-1005; Wood, et al., 1985. Nature 314 :446-449; Shaw, et al., 1988. J. NatL. Cancer Inst. 5 80: 1553-1559); Morrison(1985) Science 229:1202-1207; Oi, et al. (1986) BioTechniques 4:214; Jones, et al., 1986. Nature 321: 552-525; Verhoeyan, et al., 1988. Science 239: 1534; and Beidler, et al., 1988. J. Immunol. 141: 4053-4060. Each of the above citations are incorporated herein by reference in their entirety. In one embodiment, methods for the screening of antibodies that possess the desired 0 specificity include, but are not limited to, enzyme-linked immunosorbent assay (ELISA) and other immunologically-mediated techniques known within the art. In a specific embodiment, selection of antibodies that are specific to a particular domain of an FCTRX protein is facilitated by generation of hybridomas that bind to the fragment of an FCTRX protein possessing such a domain. Thus, antibodies that are specific for a desired domain within an 5 FCTRX protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein. Anti-FCTRX antibodies may be used in methods known within the art relating to the localization and/or quantitation of an FCTRX protein (e.g., for use in measuring levels of the FCTRX protein within appropriate physiological samples, for use in diagnostic methods, for 0 use in imaging the protein, and the like). In a given embodiment, antibodies for FCTRX proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antibody derived binding domain, are utilized as pharmacologically-active compounds (hereinafter "Therapeutics"). 66 WO 01/46231 PCT/USOO/34898 An anti-FCTRX antibody (e.g., monoclonal antibody) can be used to isolate an FCTRX polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. An anti-FCTRX antibody can facilitate the purification of natural FCTRX polypeptide from cells and of recombinantly-produced FCTRX polypeptide expressed 5 in host cells. Moreover, an anti-FCTRX antibody can be used to detect FCTRX protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the FCTRX protein. Anti-FCTRX antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for. example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling 0 (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, p-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; 5 examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131, S or 3 H. !0 FCTRX Recombinant Expression Vectors and Host Cells Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding an FCTRX protein, or derivatives, fragments, analogs or homologs thereof. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a 5 "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., 0 non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are referred to herein as "expression vectors". In general, 67 WO 01/46231 PCT/USOO/34898 expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, plasmidd" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective 5 retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions. The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid 0 sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). 5 The term "regulatory sequence" is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell 0 and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including 5 fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., FCTRX proteins, mutant forms of FCTRX proteins, fusion proteins, etc.). The recombinant expression vectors of the invention can be designed for expression of FCTRX proteins in prokaryotic or eukaryotic cells. For example, FCTRX proteins can be expressed in bacterial cells such as Escherichia coli, insect cells (using baculovirus expression 0 vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase. 68 WO 01/46231 PCT/USOO/34898 Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors 5 typically serve three purposes: (i) to increase expression of recombinant protein; (ii) to increase the solubility of the recombinant protein; and (iii) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety 0 subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988. Gene 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) that fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the 5 target recombinant protein. Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al., (1988) Gene 69:301-315) and pET 1ld (Studier et al., GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89). 0o One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the 5 individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g., Wada, et al., 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques. In another embodiment, the FCTRX expression vector is a yeast expression vector. Examples of vectors for expression in yeast Saccharomyces cerivisae include pYepSecl 0 (Baldari, et al., 1987. EMBO J 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al., 1987. Gene 54: 113-123), pYES2 (Invitrogen Corporation, San Diego, Calif.), and picZ (InVitrogen Corp, San Diego, Calif.). Alternatively, FCTRX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., 69 WO 01/46231 PCT/USOO/34898 SF9 cells) include the pAc series (Smith, et al., 1983. Mol. Cell. Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39). In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors 5 include pCDM8 (Seed, 1987. Nature 329: 840) and pMT2PC (Kaufmlan, et al., 1987. EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of .0 Sambrook, et al., MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989. In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific .5 regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert, et al., 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988. Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J. 8: 729-733) and immunoglobulins (Banerji, et al., 1983. Cell 33: 729-740; Queen and Z0 Baltimore, 1983. Cell 33: 741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle, 1989. Proc. Natl. Acad. Sci. USA 86: 5473-5477), pancreas-specific promoters (Edlund, et al., 1985. Science 230: 912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also 5 encompassed, e.g., the murine hox promoters (Kessel and Gruss, 1990. Science 249: 374-379) and the c-fetoprotein promoter (Campes and Tilghman, 1989. Genes Dev. 3: 537-546). The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows 0 for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to FCTRX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression 70 WO 01/46231 PCT/USOO/34898 of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene 5 expression using antisense genes see, e.g., Weintraub, et al., "Antisense RNA as a molecular tool for genetic analysis," Reviews-Trends in Genetics, Vol. 1(1) 1986. Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer 0 not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein. A host cell can be any prokaryotic or eukaryotic cell. For example, FCTRX protein 5 can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art. Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and 0 "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring 5 Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals. For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that 0 encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding FCTRX or can be introduced on a separate vector. Cells stably transfected with the introduced 71 WO 01/46231 PCT/USOO/34898 nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die). A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) FCTRX protein. Accordingly, the invention further provides 5 methods for producing FCTRX protein using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding FCTRX protein has been introduced) in a suitable medium such that FCTRX protein is produced. In another embodiment, the method further comprises isolating FCTRX protein from the medium or the host cell. 0 Transgenic FCTRX Animals The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which FCTRX protein-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous 5 FCTRX sequences have been introduced into their genome or homologous recombinant animals in which endogenous FCTRX sequences have been altered. Such animals are useful for studying the function and/or activity of FCTRX protein and for identifying and/or evaluating modulators of FCTRX protein activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in 0 which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types 5 or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous FCTRX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal. 0 A transgenic animal of the invention can be created by introducing FCTRX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal. The human FCTRX cDNA sequences of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17,19, 21, 23, 25, 72 WO 01/46231 PCT/USOO/34898 27, and 29, can be introduced as a transgene into the genome of a non-human animal. Alternatively, a non-human homologue of the human FCTRX gene, such as a mouse FCTRX gene, can be isolated based on hybridization to the human FCTRX cDNA (described further supra) and used as a transgene. Intronic sequences and polyadenylation signals can also be 5 included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably-linked to the FCTRX transgene to direct expression of FCTRX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866; 0 4,870,009; and 4,873,191; and Hogan, 1986. In: MANIPULATING THE MOUSE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the FCTRX transgene in its genome and/or expression of FCTRX mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed 5 additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene encoding FCTRX protein can further be bred to other transgenic animals carrying other transgenes. To create a homologous recombinant animal, a vector is prepared which contains at least a portion of an FCTRX gene into which a deletion, addition or substitution has been !0 introduced to thereby alter, e.g., functionally disrupt, the FCTRX gene. The FCTRX gene can be a human gene (e.g., the cDNA of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29), but more preferably, is a non-human homologue of a human FCTRX gene. For example, a mouse homologue of human FCTRX gene of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, can be used to construct a homologous recombination vector 5 suitable for altering an endogenous FCTRX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous FCTRX gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector). Alternatively, the vector can be designed such that, upon homologous recombination, 0 the endogenous FCTRX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous FCTRX protein). In the homologous recombination vector, the altered portion of the FCTRX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the FCTRX gene to allow for homologous recombination to occur between the exogenous FCTRX gene 73 WO 01/46231 PCT/USOO/34898 carried by the vector and an endogenous FCTRX gene in an embryonic stem cell. The additional flanking FCTRX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector. See, e.g., Thomas, et al., 1987. Cell 51: 5 503 for a description of homologous recombination vectors. The vector is ten introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced FCTRX gene has homologously-recombined with the endogenous FCTRX gene are selected. See, e.g., Li, et al., 1992. Cell 69: 915. The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to l0 form aggregation chimeras. See, e.g., Bradley, 1987. In: TERATOCARCINOMAS AND EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the .5 homologously-recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley, 1991. Curr. Opin. Biotechnol. 2: 823-829; PCT International Publication Nos.: WO 90/11354; WO 91/01140; WO 92/0968; and WO 93/04169. In another embodiment, transgenic non-humans animals can be produced that contain !0 selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage Pl. For a description of the cre/loxP recombinase system, See, e.g., Lakso, et al., 1992. Proc. Natl. Acad. Sci. USA 89: 6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharonyces cerevisiae. See, O'Gorman, et al., 1991. Science 251:1351-1355. If a cre/loxP 5 recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase. 0 Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al., 1997. Nature 385: 810-813. In brief, a cell (e.g., a somatic cell) from the transgenic animal can be isolated and induced to exit the growth cycle and enter Go phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the 74 WO 01/46231 PCT/USOO/34898 quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated. 5 Pharmaceutical Compositions The FCTRX nucleic acid molecules, FCTRX proteins, and anti-FCTRX antibodies (also referred to herein as "active compounds") of the invention, and derivatives, fragments, analogs and homologs thereof, can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, t0 or antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, 15 which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active 2O compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), 5 transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such 30 as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral 75 WO 01/46231 PCT/USOO/34898 preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous 5 preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL * (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating 10 action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of .5 surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by 0 including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions can be prepared by incorporating the active compound (e.g., an FCTRX protein or anti-FCTRX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered 5 sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered 0 solution thereof. Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier 76 WO 01/46231 PCT/USOO/34898 for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar 5 nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. 0 For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be 5 permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art. 0 The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery. In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release 5 formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal 0 suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811. 77 WO 01/46231 PCT/USOO/34898 It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the 5 desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. 0 The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al., 1994. Proc. NatL. Acad. Sci. USA 91: 3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector 5 in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system. The pharmaceutical compositions can be included in'a container, pack, or dispenser 0 together with instructions for administration. Screening and Detection Methods The isolated nucleic acid molecules of the invention can be used to express FCTRX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect FCTRX mRNA (e.g., in a biological sample) or a genetic lesion in an FCTRX gene, 5 and to modulate FCTRX activity, as described further, below. In addition, the FCTRX proteins can be used to screen drugs or compounds that modulate the FCTRX protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of FCTRX protein or production of FCTRX protein forms that have decreased or aberrant activity compared to FCTRX wild-type protein (e.g.; diabetes (regulates insulin release); 0 obesity (binds and transport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious disease(possesses anti-microbial activity) and the various dyslipidemias. In addition, the anti-FCTRX antibodies of the invention can be used to 78 WO 01/46231 PCT/USOO/34898 detect and isolate FCTRX proteins and modulate FCTRX activity. In yet a further aspect, the invention can be used in methods to influence appetite, absorption of nutrients and the disposition of metabolic substrates in both a positive and negative fashion. The invention further pertains to novel agents identified by the screening assays 5 described herein and uses thereof for treatments as described, supra. Screening Assays The invention provides a method (also referred to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) that bind to FCTRX proteins or have a 10 stimulatory or inhibitory effect on, e.g., FCTRX protein expression or FCTRX protein activity. The invention also includes compounds identified in the screening assays described herein. In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of an FCTRX protein or polypeptide or biologically-active portion thereof. The test compounds of the .5 invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, !0 while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997. Anticancer Drug Design 12: 145. A "small molecule" as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, :5 lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention. Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt, et al., 1993. Proc. Natl. Acad. Sci. U.S.A. 90: 6909; Erb, et al., 1994. 0 Proc. Natl. Acad. Sci. U.S.A. 91: 11422; Zuckermann, et al., 1994. J Med. Chem. 37: 2678; Cho, et al., 1993. Science 261: 1303; Carrell, et al., 1994. Angew. Chem. Int. Ed. Engl. 33: 2059; Carell, et al., 1994. Angew. Chem. Int. Ed. Engl. 33: 2061; and Gallop, et al., 1994. J. Med. Chem. 37: 1233. 79 WO 01/46231 PCT/USOO/34898 Libraries of compounds may be presented in solution (e.g., Houghten, 1992. Biotechniques 13: 412-421), or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al., 1992. Proc. Natl. Acad. Sci. USA 89: 5 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390; Devlin, 1990. Science 249: 404-406; Cwirla, et al., 1990. Proc. Natl. Acad. Sci. U.S.A. 87: 6378-6382; Felici, 1991. J. Mol. Biol. 222: 301-3 10; Ladner, U.S. Patent No. 5,233,409.). In one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of FCTRX protein, or a biologically-active portion thereof, on the cell 0 surface is contacted with a test compound and the ability of the test compound to bind to an FCTRX protein determined. The cell, for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the FCTRX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the FCTRX protein or biologically-active 5 portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 125, 35S, 14 C, or 'H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by 0 determination of conversion of an appropriate substrate to product. In one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of FCTRX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds FCTRX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an FCTRX 5 protein, wherein determining the ability of the test compound to interact with an FCTRX protein comprises determining the ability of the test compound to preferentially bind to FCTRX protein or a biologically-active portion thereof as compared to the known compound. In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of FCTRX protein, or a biologically-active portion 0 thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the FCTRX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of FCTRX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the FCTRX protein to bind to or interact with an 80 WO 01/46231 PCT/USOO/34898 FCTRX target molecule. As used herein, a "target molecule" is a molecule with which an FCTRX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses an FCTRX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell 5 membrane or a cytoplasmic molecule. An FCTRX target molecule can be a non-FCTRX molecule or an FCTRX protein or polypeptide of the invention . In one embodiment, an FCTRX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound FCTRX molecule) through the cell membrane and into the cell. The target, 0 for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with FCTRX. Determining the ability of the FCTRX protein to bind to or interact with an FCTRX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the FCTRX protein to bind to or 5 interact with an FCTRX target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca 2 +, diacylglycerol, IP 3 , etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising an FCTRX-responsive 0 regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation. In yet another embodiment, an assay of the invention is a cell-free assay comprising contacting an FCTRX protein or biologically-active portion thereof with a test compound and 5 determining the ability of the test compound to bind to the FCTRX protein or biologically active portion thereof. Binding of the test compound to the FCTRX protein can be determined either directly or indirectly as described above. In one such embodiment, the assay comprises contacting the FCTRX protein or biologically-active portion thereof with a known compound which binds FCTRX to form an assay mixture, contacting the assay mixture with a test 0 compound, and determining the ability of the test compound to interact with an FCTRX protein, wherein determining the ability of the test compound to interact with an FCTRX protein comprises determining the ability of the test compound to preferentially bind to FCTRX or biologically-active portion thereof as compared to the known compound. 81 WO 01/46231 PCT/USOO/34898 In still another embodiment, an assay is a cell-free assay comprising contacting FCTRX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the FCTRX protein or biologically-active portion thereof. Determining the ability of the test 5 compound to modulate the activity of FCTRX can be accomplished, for example, by determining the ability of the FCTRX protein to bind to an FCTRX target molecule by one of the methods described above for determining direct binding. In an alternative embodiment, determining the ability of the test compound to modulate the activity of FCTRX protein can be accomplished by determining the ability of the FCTRX protein further modulate an FCTRX 0 target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as described, supra. In yet another embodiment, the cell-free assay comprises contacting the FCTRX protein or biologically-active portion thereof with a known compound which binds FCTRX protein to form an assay mixture, contacting the assay mixture with a test compound, and 5 determining the ability of the test compound to interact with an FCTRX protein, wherein determining the ability of the test compound to interact with an FCTRX protein comprises determining the ability of the FCTRX protein to preferentially bind to or modulate the activity of an FCTRX target molecule. The cell-free assays of the invention are amenable to use of both the soluble form or 0 the membrane-bound form of FCTRX protein. In the case of cell-free assays comprising the membrane-bound form of FCTRX protein, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of FCTRX protein is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, 5 decanoyl-N-methylglucamide, Triton* X-100, Triton* X- 114, Thesit*, Isotridecypoly(ethylene glycol ether)b, N-dodecyl--N,N-dimethyl-3-ammonio- 1-propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1-propane sulfonate (CHAPS), or 3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy-1-propane sulfonate (CHAPSO). In more than one embodiment of the above assay methods of the invention, it may be 0 desirable to immobilize either FCTRX protein or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to FCTRX protein, or interaction of FCTRX protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such 82 WO 01/46231 PCT/USOO/34898 vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix. For example, GST-FCTRX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or 5 glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or FCTRX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, 0 complex determined either directly or indirectly, for example, as described, supra. Alternatively, the complexes can be dissociated from the matrix, and the level of FCTRX protein binding or activity determined using standard techniques. Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention.~For example, either the FCTRX protein or its target 5 molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated FCTRX protein or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, Ill.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with FCTRX protein or target 0 molecules, but which do not interfere with binding of the FCTRX protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or FCTRX protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-innobilized complexes, include immunodetection of complexes using antibodies reactive with the FCTRX protein or target 5 molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the FCTRX protein or target molecule. In another embodiment, modulators of FCTRX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of FCTRX mRNA or protein in the cell is determined. The level of expression of FCTRX mRNA or 0 protein in the presence of the candidate compound is compared to the level of expression of FCTRX mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of FCTRX mRNA or protein expression based upon this comparison. For example, when expression of FCTRX mRNA or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than 83 WO 01/46231 PCT/USOO/34898 in its absence, the candidate compound is identified as a stimulator of FCTRX mRNA or protein expression. Alternatively, when expression of FCTRX mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of FCTRX mRNA or protein expression. 5 The level of FCTRX mRNA or protein expression in the cells can be determined by methods described herein for detecting FCTRX mRNA or protein. In yet another aspect of the invention, the FCTRX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos, et al., 1993. Cell 72: 223-232; Madura, et al., 1993. J. Biol. Chem. 268: 12046-12054; [0 Bartel, et al., 1993. Biotechniques 14: 920-924; Iwabuchi, et al., 1993. Oncogene 8: 1693-1696; and Brent WO 94/10300), to identify other proteins that bind to or interact with FCTRX ("FCTRX-binding proteins" or "FCTRX-bp") and modulate FCTRX activity. Such FCTRX-binding proteins are also likely to be involved in the propagation of signals by the FCTRX proteins as, for example, upstream or downstream elements of the FCTRX pathway. .5 The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for FCTRX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an 0 unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming an FCTRX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional 5 regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with FCTRX. The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein. 0 Detection Assays Portions or fragments of the cDNA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. By way of example, and not of limitation, these sequences can be used to:.(i) map their respective 84 WO 01/46231 PCT/USOO/34898 genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Some of these applications are described in the subsections, below. 5 Chromosome Mapping Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the FCTRX sequences, SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or fragments or derivatives 0 thereof, can be used to map the location of the FCTRX genes, respectively, on a chromosome. The mapping of the FCTRX sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease. Briefly, FCTRX genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the FCTRX sequences. Computer analysis of the 5 FCTRX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the FCTRX sequences will yield an amplified fragment. !0 Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the .5 needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. See, e.g., D'Eustachio, et al., 1983. Science 220: 919-924. Somatic cell hybrids containing only fragments of human 0 chromosomes can also be produced by using human chromosomes with translocations and deletions. PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using 85 WO 01/46231 PCT/USOO/34898 a single thermal cycler. Using the FCTRX sequences to design oligonucleotide primers, sub localization can be achieved with panels of fragments from specific chromosomes. Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one 5 step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. 0 However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, see, Verma, et al., HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988). 5 Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents corresponding to noncoding regions of the genes actually are preferred for mapping purposes. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations 0 during chromosomal mapping. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, e.g., in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes 5 and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland, et al., 1987. Nature, 325: 783-787. Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the FCTRX gene, can be determined. If a mutation 0 is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete 86 WO 01/46231 PCT/USOO/34898 sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymorphisms. Tissue Typing The FCTRX sequences of the invention can also be used to identify individuals from 5 minute biological samples. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. The sequences of the invention are useful as additional DNA markers for RFLP ("restriction fragment length polymorphisms," described in U.S. Patent No. 5,272,057). Furthermore, the sequences of the invention can be used to provide an alternative 0 technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the FCTRX sequences described herein can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it. Panels of corresponding DNA sequences from individuals, prepared in this manner, 5 can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals and from tissue. The FCTRX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding ,0 regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymorphisms (SNPs), which include restriction fragment length polymorphisms (RFLPs). Each of the sequences described herein can, to some degree, be used as a standard 5 against which DNA from an individual can be compared for identification purposes. Because greater numbers of polymorphisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in 0 SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, are used, a more appropriate number of primers for positive individual identification would be 500-2,000. Predictive Medicine The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for 87 WO 01/46231 PCT/USOO/34898 prognostic (predictive) purposes to thereby treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining FCTRX protein and/or nucleic acid expression as well as FCTRX activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a 5 disease or disorder, or is at risk of developing a disorder, associated with aberrant FCTRX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic 0 syndrome X and wasting disorders associated with chronic diseases and various cancers. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with FCTRX protein, nucleic acid expression or activity. For example, mutations in an FCTRX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive purpose to thereby 5 prophylactically treat an individual prior to the onset of a disorder characterized by or associated with FCTRX protein, nucleic acid expression, or biological activity. Another aspect of the invention provides methods for determining FCTRX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (referred to herein as "phannacogenomics"). o Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.) Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., 5 drugs, compounds) on the expression or activity of FCTRX in clinical trials. These and other agents are described in further detail in the following sections. Diagnostic Assays An exemplary method for detecting the presence or absence of FCTRX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological 0 sample with a compound or an agent capable of detecting FCTRX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes FCTRX protein such that the presence of FCTRX is detected in the biological sample. An agent for detecting FCTRX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to FCTRX mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length FCTRX nucleic acid, such as the nucleic 88 WO 01/46231 PCT/USOO/34898 acid of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to FCTRX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are 5 described herein. An agent for detecting FCTRX protein is an antibody capable of binding to FCTRX protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab') 2 ) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass 0 direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently 5 labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect FCTRX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of FCTRX mRNA include Northern hybridizations and in situ 0 hybridizations. In vitro techniques for detection of FCTRX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of FCTRX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of FCTRX protein include introducing into a subject a labeled anti-FCTRX antibody. For example, the antibody 5 can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques. In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A preferred biological sample is a 0 peripheral blood leukocyte sample isolated by conventional means from a subject. In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting FCTRX protein, mRNA, or genomic DNA, such that the presence of FCTRX protein, mRNA or genomic DNA is detected in the biological sample, and comparing 89 WO 01/46231 PCT/USOO/34898 the presence of FCTRX protein, mRNA or genomic DNA in the control sample with the presence of FCTRX protein, mRNA or genomic DNA in the test sample. The invention also encompasses kits for detecting the presence of FCTRX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of 5 detecting FCTRX protein or mRNA in a biological sample; means for determining the amount of FCTRX in the sample; and means for comparing the amount of FCTRX in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect FCTRX protein or nucleic acid. Prognostic Assays 0 The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant FCTRX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with FCTRX protein, nucleic acid expression or activity. 5 Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the invention provides a method for identifying a disease or disorder associated with aberrant FCTRX expression or activity in which a test sample is obtained from a subject and FCTRX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of FCTRX protein or nucleic acid is diagnostic for a O subject having or at risk of developing a disease or disorder associated with aberrant FCTRX expression or activity. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue. Furthermore, the prognostic assays described herein can be used to determine whether 5 a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with aberrant FCTRX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. Thus, the invention provides methods for determining whether a subject can be effectively 0 treated with an agent for a disorder associated with aberrant FCTRX expression or activity in which a test sample is obtained and FCTRX protein or nucleic acid is detected (e.g., wherein the presence of FCTRX protein or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant FCTRX expression or activity). 90 WO 01/46231 PCT/USOO/34898 The methods of the invention can also be used to detect genetic lesions in an FCTRX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by aberrant cell proliferation and/or differentiation. In various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a 5 genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding an FCTRX-protein, or the misexpression of the FCTRX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of: (i) a deletion of one or more nucleotides from an FCTRX gene; (ii) an addition of one or more nucleotides to an FCTRX gene; (iii) a substitution of one or more nucleotides of an FCTRX gene, (iv) a 0 chromosomal rearrangement of an FCTRX gene; (v) an alteration in the level of a messenger RNA transcript of an FCTRX gene, (vi) aberrant modification of an FCTRX gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non-wild-type splicing pattern of a messenger RNA transcript of an FCTRX gene, (viii) a non-wild-type level of an FCTRX protein, (ix) allelic loss of an FCTRX gene, and (x) inappropriate post-translational 5 modification of an FCTRX protein. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in an FCTRX gene. A preferred biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells. 0 In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran, et al., 1988. Science 241: 1077-1080; and Nakazawa, et al., 1994. Proc. Natl. Acad. Sci. USA 91: 360-364), the latter of which can be particularly useful for detecting point 5 mutations in the FCTRX-gene (see, Abravaya, et al., 1995. Nucl. Acids Res. 23: 675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to an FCTRX gene under conditions such that hybridization and amplification of the FCTRX gene (if present) 0 occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein. 91 WO 01/46231 PCT/USOO/34898 Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et al., 1990. Proc. Natl. Acad. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et al., 1989. Proc. Natl. Acad. Sci. USA 86: 1173-1177); Qp Replicase (see, Lizardi, et al, 1988. BioTechnology 6: 1197), or any other nucleic acid amplification 5 method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers. In an alternative embodiment, mutations in an FCTRX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and 0 control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,493,531) can be used to score for the presence of specific mutations by development or 5 loss of a ribozyme cleavage site. In other embodiments, genetic mutations in FCTRX can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high-density arrays containing hundreds or thousands of oligonucleotides probes. See, e.g., Cronin, et al., 1996. Human Mutation 7: 244-255; Kozal, et al., 1996. Nat. Med. 2: 753-759. For example, genetic !0 mutations in FCTRX can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, et al., supra. Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This is followed by a second 5 hybridization array tha' allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene. In yet another embodiment, any of a variety of sequencing reactions known in the art 0 can be used to directly sequence the FCTRX gene and detect mutations by comparing the sequence of the sample FCTRX with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc. Natl. Acad. Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures 92 WO 01/46231 PCT/USOO/34898 can be utilized when performing the diagnostic assays (see, e.g., Naeve, et al., 1995. Biotechniques 19: 448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen, et al., 1996. Adv. Chromatography 36: 127-162; and Griffin, et al., 1993. Apple. Biochem. Biotechnol. 38: 147-159). 5 Other methods for detecting mutations in the FCTRX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. See, e.g., Myers, et al., 1985. Science 230: 1242. In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type FCTRX sequence with 0 potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with Si nuclease to enzymatically digesting the mismatched regions. In other embodiments, either 5 DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton, et al., 1988. Proc. Natl. Acad. Sci. USA 85: 4397; Saleeba, et al., 1992. Methods Enzymol. 217: 286-295. In an embodiment, the control !0 DNA or RNA can be labeled for detection. In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in FCTRX cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli 5 cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al., 1994. Carcinogenesis 15: 1657-1662. According to an exemplary embodiment, a probe based on an FCTRX sequence, e.g., a wild-type FCTRX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be 0 detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039. In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in FCTRX genes. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al., 1989. Proc. Natl. Acad. Sci. USA: 86: 2766; Cotton, 93 WO 01/46231 PCT/USOO/34898 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet. Anal. Tech. Apple. 9: 73-79. Single-stranded DNA fragments of sample and control FCTRX nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection 5 of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al., 1991. l0 Trends Genet. 7: 5. In yet another embodiment, the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE). See, e.g., Myers, et al., 1985. Nature 313: 495. When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely 15 denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum and Reissner, 1987. Biophys. Chem. 265: 12753. Examples of other techniques for detecting point mutations include, but are not limited 20 to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al., 1986. Nature 324: 163; Saiki, et al., 1989. Proc. Natl. A cad. Sci. USA 86: 6230. Such allele specific oligonucleotides 25 are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA. Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as 30 primers for specific amplification may carry the mutation of interest in the center of the' molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al., 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. Tibtech. 11: 238). In addition it may be desirable to introduce a novel 94 WO 01/46231 PCT/USOO/34898 restriction site in the region of the mutation to create cleavage-based detection. See, e.g., Gasparini, et al., 1992. Mol. Cell Probes 6: 1. It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc. Nati. Acad. Sci. USA 88: 189. In such cases, ligation will occur only if there is a 5 perfect match at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification. The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose 10 patients exhibiting symptoms or family history of a disease or illness involving an FCTRX gene. Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which FCTRX is expressed may be utilized in the prognostic assays described herein. However, any biological sample containing nucleated cells may be used, including, for example, buccal 15 mucosal cells. Pharmacogenomics Agents, or modulators that have a stimulatory or inhibitory effect on FCTRX activity (e.g., FCTRX gene expression), as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) disorders (The 20 disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.) In conjunction with such 25 treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the 30 selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of FCTRX protein, expression of FCTRX nucleic acid, or mutation content of FCTRX genes in an 95 WO 01/46231 PCT/USOO/34898 individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See 5 e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol. Physiol., 23: 983-985; Linder, 1997. Clin. Chem., 43: 254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body (altered drug action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drug metabolism). These pharmacogenetic conditions can 0 occur either as rare defects or as polymorphisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans. As an illustrative embodiment, the activity of drug metabolizing enzymes is a major 5 determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are 0 expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymorphic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side Z5 effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-forned metabolite morphine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification. 0 Thus, the activity of FCTRX protein, expression of FCTRX nucleic acid, or mutation content of FCTRX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymorphic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness 96 WO 01/46231 PCT/USOO/34898 phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an FCTRX modulator, such as a modulator identified by one of the exemplary screening assays described herein. 5 Monitoring of Effects During Clinical Trials Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of FCTRX (e.g., the ability to modulate aberrant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to 0 increase FCTRX gene expression, protein levels, or upregulate FCTRX activity, can be monitored in clinical trails of subjects exhibiting decreased FCTRX gene expression, protein levels, or downregulated FCTRX activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease FCTRX gene expression, protein levels, or downregulate FCTRX activity, can be monitored in clinical trails of subjects exhibiting 5 increased FCTRX gene expression, protein levels, or upregulated FCTRX activity. In such clinical trials, the expression or activity of FCTRX and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell. By way of example, and not of limitation, genes, including FCTRX, that are modulated .0 in cells by treatment with an agent (e.g., compound, drug or small molecule) that modulates FCTRX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of FCTRX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene .5 expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of FCTRX or other genes. In this manner, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, 0 and at various points during, treatment of the individual with the agent. In one embodiment, the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration 97 WO 01/46231 PCT/USOO/34898 sample from a subject prior to administration of the agent; (ii) detecting the level of expression of an FCTRX protein, mRNA, or genomic DNA in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the FCTRX protein, mRNA, or genomic DNA in the 5 post-administration samples; (v) comparing the level of expression or activity of the FCTRX protein, mRNA, or genomic DNA in the pre-administration sample with the FCTRX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of FCTRX to higher levels 0 than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of FCTRX to lower levels than detected, i.e., to decrease the effectiveness of the agent. Methods of Treatment The invention provides for both prophylactic and therapeutic methods of treating a 5 subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant FCTRX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic 0 syndrome X and wasting disorders associated with chronic diseases and various cancers. These methods of treatment will be discussed more fully, below. Disease and Disorders Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with 5 Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) administration of antisense nucleic acid 0 and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endoggenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989. Science 244: 1288-1292); or (v) modulators ( i.e., 98 WO 01/46231 PCT/USOO/34898 inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner. Diseases and disorders that are characterized by decreased (relative to a subject not 5 suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability. 10 Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by t5 sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like). Prophylactic Methods In one aspect, the invention provides a method for preventing, in a subject, a disease or 20 condition associated with an aberrant FCTRX expression or activity, by administering to the subject an agent that modulates FCTRX expression or at least one FCTRX activity. Subjects at risk for a disease that is caused or contributed to by aberrant FCTRX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation 25 of symptoms characteristic of the FCTRX aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending upon the type of FCTRX aberrancy, for example, an FCTRX agonist or FCTRX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the 30 following subsections. Therapeutic Methods Another aspect of the invention pertains to methods of modulating FCTRX expression or activity for therapeutic purposes. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of FCTRX protein 99 WO 01/46231 PCT/USOO/34898 activity associated with the cell. An agent that modulates FCTRX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of an FCTRX protein, a peptide, an FCTRX peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more FCTRX protein activity. Examples of 5 such stimulatory agents include active FCTRX protein and a nucleic acid molecule encoding FCTRX that has been introduced into the cell. In another embodiment, the agent inhibits one or more FCTRX protein activity. Examples of such inhibitory agents include antisense FCTRX nucleic acid molecules and anti-FCTRX antibodies. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., 10 by administering the agent to a subject). As such, the invention provides methods of treating an individual afflicted with a disease or disorder characterized by aberrant expression or activity of an FCTRX protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) 15 FCTRX expression or activity. In another embodiment, the method involves administering an FCTRX protein or nucleic acid molecule as therapy to compensate for reduced or aberrant FCTRX expression or activity. Stimulation of FCTRX activity is desirable in situations in which FCTRX is abnormally downregulated and/or in which increased FCTRX activity is likely to have a Z0 beneficial effect. One example of such a situation is where a subject has a disorder characterized by aberrant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia). Determination of the Biological Effect of the Therapeutic 5 In various embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue. In various specific embodiments, in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts 30 the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo 100 WO 01/46231 PCT/USOO/34898 testing, any of the animal model system known in the art may be used prior to administration to human subjects. Prophylactic and Therapeutic Uses of the Compositions of the Invention The FCTRX nucleic acids and proteins of the invention are useful in potential 5 prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders 0 associated with chronic diseases and various cancers. As an example, a cDNA encoding the FCTRX protein of the invention may be useful in gene therapy, and the protein may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, 5 anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias. Both the novel nucleic acid encoding the FCTRX protein, and the FCTRX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the !0 presence or amount of the nucleic acid or the protein are to be assessed. A further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of antibodies which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods. 5 EXAMPLES The following examples illustrate by way of non-limiting example various aspects of the invention. Example 1: Method of Identifying the Nucleic Acids The novel nucleic acids of the invention were identified by TblastN using CuraGen 0 Corporation's sequence file, run against the Genomic Daily Files made available by GenBank. The nucleic acids were further predicted by the program GenScanTM, including selection of exons. These were further modified by means of similarities using BLAST searches. The 101 WO 01/46231 PCT/USOO/34898 sequences were then manually corrected for apparent inconsistencies, thereby obtaining the sequences encoding the full-length proteins. Example 2. Quantitative expression analysis of FCTR2 in various cells and tissues The quantitative expression of clone AL078594 A (FCTR2) was assessed in a large 5 number of normal and tumor sample cells and cell lines (Panel 1), as well as in surgical tissue samples (Panel 2), by real time quantitative PCR (TAQMAN*) performed on a Perkin-Elmer Biosystems ABI PRISM® 7700 Sequence Detection System. First, 96 RNA samples were normalized to p-actin and GAPDH. RNA (~50 ng total or ~1 ng polyA+) was converted to cDNA using the TAQMAN* Reverse Transcription Reagents 10 Kit (PE Biosystems, Foster City, CA; Catalog No. N808-0234) and random hexamers according to the manufacturer's protocol. Reactions were performed in 20 ul and incubated for 30 min. at 48 0 C. cDNA (5 ul) was then transferred to a separate plate for the TAQMAN@ reaction using -actin and GAPDH TAQMAN@ Assay Reagents (PE Biosystems; Catalog Nos. 4310881E and 4310884E, respectively) and TAQMAN@ universal PCR Master Mix (PE 15 Biosystems; Catalog No. 4304447) according to the manufacturer's protocol. Reactions were performed in 25 ul using the following parameters: 2 min. at 50 0 C; 10 min. at 95 0 C; 15 sec. at 95 0 C/1 min. at 60 0 C (40 cycles). Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being 20 represented as 2 to the power of delta CT. The percent relative expression is then obtained by taking the reciprocal of this RNA difference and multiplying by 100. The average CT values obtained for B-actin and GAPDH were used to normalize RNA samples. The RNA sample generating the highest CT value required no further diluting, while all other samples were diluted relative to this sample according to their p-actin /GAPDH average CT values. 5 Normalized RNA (5 ul) was converted to cDNA and analyzed via TAQMAN@ using One Step RT-PCR Master Mix Reagents (PE Biosystems; Catalog No. 4309169) and gene specific primers according to the manufacturer's instructions. Probes and primers were designed for each assay according to Perkin Elmer Biosystem's Primer Express Software package (version I for Apple Computer's Macintosh Power PC) or a similar algorithm using 30 the target sequence as input. Default settings were used for reaction conditions and the following parameters were set before selecting primers: primer concentration= 250 nM, primer melting temperature (Tm) range = 58*-60' C, primer optimal Tm = 590 C, maximum primer difference = 2* C, probe does not have 5' G, probe Tm must be 100 C greater than 102 WO 01/46231 PCT/USOO/34898 primer Tm, amplicon size 75 bp to 100 bp. The probes and primers selected (see below) were synthesized by Synthegen (Houston, TX, USA). Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5' and 3' ends of the probe, respectively. Their final concentrations 5 were: forward and reverse primers, 900 nM each, and probe, 200nM. The expression was probed with the primer-probe set Ag 259. The Forward primer sequence is 5'-GGAGAGGCTCTGAAGCTACACAA-3' (SEQ ID NO:3 1); the Probe primer sequence is TET-5'-TCAGCTGCACAAGCCCCCTGCT-3'-TAMRA (SEQ ID NO:32); and the Reverse primer sequence is 5'-GCAGTGGTTGGAGCTGGAA-3' (SEQ ID NO:33). Table 0 15 shows the primer locations within the FCTR2 nucleic acid sequence. Table 15. Primer-Probe Set Ag259 Primers Length Start Position Forward 23 124 Probe 22 158 Reverse 19 181 PCR conditions: Normalized RNA from each tissue and each cell line was spotted in each well of a 96 well PCR plate (Perkin Elmer Biosystems). PCR cocktails including two 5 probes (a probe specific for the target clone and another gene-specific probe multiplexed with the target probe) were set up using 1X TaqMan T M PCR Master Mix for the PE Biosystems 7700, with 5 mM MgCl2, dNTPs (dA, G, C, U at 1:1:1:2 ratios), 0.25 U/ml AmpliTaq Gold T M (PE Biosystems), and 0.4 U/pl RNase inhibitor, and 0.25 U/ 1 reverse transcriptase. Reverse transcription was performed at 480 C for 30 minutes followed by amplification/PCR cycles as 0 follows: 950 C 10 min, then 40 cycles of 950 C for 15 seconds, 600 C for 1 minute. The results for various cells and cell lines that constitute Panel 1 are shown in Table 16. In Table 16, the following abbreviations are used: ca. = carcinoma; * = established from metastasis; met = metastasis; s cell var= small cell variant; non-s = non-sm =non-small; squam = squamous; pl. eff= pl effusion pleural effusion; glio = glioma; astro = astrocytoma; and 5 neuro = neuroblastoma. Table 16. Tissue Name Rel. Tissue Name Rel. Expr., Expr., Adipose 100.0 Colon ca. HT29 0.2 Adrenal gland 0.0 Colon ca. CaCo-2 0.0 Bladder 0.2 Colon ca. HCT-15 3.0 Bone marrow 0.0 Colon ca. HCT-1 16 0.0 103 WO 01/46231 PCT/USOO/34898 Endothelial cells 0.0 Colon ca. HCC-2998 0.2 Endothelial cells (treated) 0.0 Colon ca. SW480 1.5 Liver 1.5 Colon ca.* (SW480 met)SW620 0.0 Liver (fetal) 0.0 Fetal Skeletal 0.3 Spleen 0.0 Skeletal muscle 2.6 Thymus 0.0 Heart 6.4 Thyroid 0.0 Stomach 0.0 Trachea 0.0 Gastric ca.* (liver met) NCI-N87 0.3 Testis 0.1 Kidney 4.0 Spinal cord 0.6 Kidney (fetal) 0.1 Salavary gland 0.0 Renal ca. 786-0 0.0 Brain (amygdala) 0.0 Renal ca. A498 0.1 Brain (cerebellum) 2.9 Renal ca. ACHN 0.0 Brain (hippocampus) 0.0 Renal ca. TK-10 0.1 Brain (substantia nigra) 4.8 Renal ca. UO-31 0.1 Brain (thalamus) 0.1 Renal ca. RXF 393 0.0 Cerebral Cortex 0.0 Pancreas 1.5 Brain (whole) 0.0 Pancreatic ca. CAPAN 2 0.2 Brain (fetal) 0.0 Ovary 0.2 CNS ca. (glio/astro) U-118-MG 0.2 Ovarian ca. IGROV-1 0.7 CNS ca. (astro) SF-539 0.0 Ovarian ca. OVCAR-3 51.1 CNS ca. (astro) SNB-75 0.0 Ovarian ca. OVCAR-4 52.9 CNS ca. (astro) SW1783 0.0 Ovarian ca. OVCAR-5 21.6 CNS ca. (glio) U251 0.2 Ovarian ca. OVCAR-8 0.2 CNS ca. (glio) SF-295 0.0 Ovarian ca.* (ascites) SK-OV-3 0.0 CNS ca. (glio) SNB-19 3.3 Prostate 0.0 CNS ca. (glio/astro) U87-MG 0.0 Prostate ca.* (bone met)PC-3 0.0 CNS ca.* (neuro; met) SK-N-AS 0.0 Plancenta 0.0 Small intestine 0.1 Pituitary gland 0.5 Colorectal 0.1 Uterus 0.0 It is seen from Table 16 that there is high expression of sequence AL078594_A found in several ovarian cancer cell lines, and very high expression in normal adipose tissue. 5 Panel2 Panel 2 consists of a 96 well plate (2 control wells, 94 test samples) composed of RNA/cDNA isolated from human tissue procured by surgeons working in close cooperation with the National Cancer Institute's Cooperative Human Tissue Network (CHTN) or the 0 National Disease Research Initiative (NDRI). The tissues procured are derived from human malignancies and in cases where indicated many malignant tissues have "matched margins". 104 WO 01/46231 PCT/USOO/34898 The tumor tissue and the "matched margins" are evaluated by two independent pathologists (the surgical pathologists and again by a pathologists at NDRI or CHTN). This analysis provides a gross histopathological assessment of tumor differentiation grade. Moreover, most samples include the original surgical pathology report that provides information regarding the 5 clinical stage of the patient. These matched margins are taken from the tissue surrounding (i.e. immediately proximal) to the zone of surgery (designated "NAT", for normal adjacent tissue, in Table 17). In addition, RNA/cDNA was obtained from various human tissues derived from human autopsies performed on deceased elderly people or sudden death victims (accidents, etc.). These tissue were ascertained to be free of disease and were purchased from various 0 high quality commercial sources such as Clontech, Research Genetics, and Invitrogen. RNA integrity from all samples is controlled for quality by visual assessment of agarose gel electrophoresis using 28s and 18s ribosomal RNA staining intensity ratio as a guide (2:1 to 2.5:1 28s:18s) and the presence of low molecular weight RNAs indicative of degradation products. Samples are quality controlled for genomic DNA contamination by 5 reactions run in the absence of reverse transcriptase using probe and primer sets designed to amplify across the span of a single exon. Table 17. Tissue Name Rel. Tissue Name Rel. Expr. Expr. Normal Colon GENPAK 061003 0.0 Kidney Cancer Clontech 8120607 0.0 83219 CC Well to Mod Diff 0.0 Kidney NAT Clontech 8120608 0.0 (OD03866) 83220 CC NAT (OD03866) 0.0 Kidney Cancer Clontech 8120613 0.0 83221 CC Gr.2 rectosigmoid 0.0 Kidney NAT Clontech 8120614 0.0 (OD03868) 83222 CC NAT (OD03868) 0.0 Kidney Cancer Clontech 9010320 0.0 83235 CC Mod Diff (OD03920) 0.0 Kidney NAT Clontech 9010321 0.0 83236 CC NAT (OD03920) 0.0 Normal Uterus GENPAK 061018 0.0 83237 CC Gr.2 ascend colon 0.0 Uterus Cancer GENPAK 064011 0.0 (OD03921) 83238 CC NAT (OD03921) 0.0 Normal Thyroid Clontech A+ 6570-1** 0.0 83241 CC from Partial 0.0 Thyroid Cancer GENPAK 064010 0.0 Hepatectomy (OD04309) 83242 Liver NAT (OD04309) 0.0 Thyroid Cancer INVITROGEN 0.0 A302152 87472 Colon mets to lung 0.0 Thyroid NAT INVITROGEN A302153 0.0 (OD04451-01) 87473 Lung NAT (OD04451-02) 0.0 Normal Breast GENPAK 061019 0.0 Normal Prostate Clontech A+ 0.0 84877 Breast Cancer (OD04566) 0.0 105 WO 01/46231 PCT/USOO/34898 6546-1 84140 Prostate Cancer (OD04410) 0.0 85975 Breast Cancer (OD04590-01) 0.0 84141 Prostate NAT (OD04410) 0.0 85976 Breast Cancer Mets (OD04590- 0.0 03) 87073 Prostate Cancer (OD04720- 0.0 87070 Breast Cancer Metastasis 0.0 01) (OD04655-05) 87074 Prostate NAT (OD04720-02) 0.0 GENPAK Breast Cancer 064006 0.0 Normal Lung GENPAK 061010 0.0 Breast Cancer Clontech 9100266 34.6 83239 Lung Met to Muscle 0.0 Breast NAT Clontech 9100265 100.0 (OD04286) 83240 Muscle NAT (OD04286) 0.0 Breast Cancer INVITROGEN A209073 0.0 84136 Lung Malignant Cancer 0.1 Breast NAT INVITROGEN A2090734 0.0 (OD03126) 84137 Lung NAT (OD03126) 0.0 Normal Liver GENPAK 061009 0.0 84871 Lung Cancer (OD04404) 0.0 Liver Cancer GENPAK 064003 0.0 84872 Lung NAT (OD04404) 0.0 Liver Cancer Research Genetics RNA 0.0 1025 84875 Lung Cancer (OD04565) 0.0 Liver Cancer Research Genetics RNA 0.0 1026 85950 Lung Cancer (OD04237-01) 0.2 Paired Liver Cancer Tissue Research 0.0 Genetics RNA 6004-T 85970 Lung NAT (OD04237-02) 0.0 Paired Liver Tissue Research Genetics 0.0 RNA 6004-N 83255 Ocular Mel Met to Liver 0.0 Paired Liver Cancer Tissue Research 0.0 (OD043 10) Genetics RNA 6005-T 83256 Liver NAT (ODO43 10) 0.0 Paired Liver Tissue Research Genetics 0.0 RNA 6005-N 84139 Melanoma Mets to Lung 0.0 Normal Bladder GENPAK 061001 0.0 (OD04321) 84138 Lung NAT (OD04321) 0.0 Bladder Cancer Research Genetics 0.3 RNA 1023 Normal Kidney GENPAK 061008 0.0 Bladder Cancer INVITROGEN 0.0 A302173 83786 Kidney Ca, Nuclear grade 2 0.0 87071 Bladder Cancer (OD04718-01) 0.0 (OD04338) 83787 Kidney NAT (OD04338) 0.0 87072 Bladder Normal Adjacent 0.0 (OD04718-03) 83788 Kidney Ca Nuclear grade 1/2 0.0 Normal Ovary Res. Gen. 0.0 (OD04339) 83789 Kidney NAT (OD04339) 0.0 Ovarian Cancer GENPAK 064008 0.0 83790 Kidney Ca, Clear cell type 0.0 87492 Ovary Cancer (OD04768-07) 0.0 (OD04340) 83791 Kidney NAT (OD04340) 0.0 87493 Ovary NAT (OD04768-08) 0.0 83792 Kidney Ca, Nuclear grade 3 0.0 Normal Stomach GENPAK 061017 0.0 (OD04348) 83793 Kidney NAT (OD04348) 0.0 NAT Stomach Clontech 9060359 0.0 106 WO 01/46231 PCT/USOO/34898 87474 Kidney Cancer (OD04622- 0.0 Gastric Cancer Clontech 9060395 0.0 01) 87475 Kidney NAT (OD04622-03) 0.0 NAT Stomach Clontech 9060394 0.0 85973 Kidney Cancer (OD04450- 0.0 Gastric Cancer Clontech 9060397 0.0 01) 85974 Kidney NAT (OD04450-03) 0.0 NAT Stomach Clontech 9060396 0.5 Gastric Cancer GENPAK 064005 0.2 There is high expression of sequence AL078594_A found in normal adjacent breast tissue and in breast cancer tissue. Panel 2 includes only two ovarian cancer samples, neither of which express this sequence. 5 Therefore, the FCTR2 protein of clone A1078594_A may serve as the target for a diagnostic assay in certain ovarian cancers, and as a potential therapeutic target for this subset of ovarian cancer and possibly for breast cancer. The citation of any reference herein should not be deemed as an admission that such reference is available as prior art to the instant invention. 0 EQUIVALENTS Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims which follow. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be 5 made to the invention without departing from the spirit and scope of the invention as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims. 107

Claims (49)

1. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; (b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form; (c) an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; and (d) a variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence.
2 The polypeptide of claim 1, wherein said polypeptide comprises the amino acid sequence of a naturally-occurring allelic variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30.
3. The polypeptide of claim 2, wherein said allelic variant comprises an amino acid sequence that is the translation of a nucleic acid sequence differing by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29.
4. The polypeptide of claim 1, wherein the amino acid sequence of said variant comprises a conservative amino acid substitution. 108 WO 01/46231 PCT/USOO/34898
5. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of: (a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; (b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form; (c) an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30; (d) a variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence; (e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising an amino acid sequence chosen from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, or a variant of said polypeptide, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence; and (f) a nucleic acid molecule comprising the complement of (a), (b), (c), (d) or (e).
6. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally-occurring allelic nucleic acid variant.
7. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule encodes a polypeptide comprising the amino acid sequence of a naturally-occurring polypeptide variant.
8. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29. 109 WO 01/46231 PCT/USOO/34898
9. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29; (b) a nucleotide sequence differing by one or more nucleotides from a nucleotide sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, provided that no more than 20% of the nucleotides differ from said nucleotide sequence; (c) a nucleic acid fragment of (a); and (d) a nucleic acid fragment of (b).
10. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule hybridizes under stringent conditions to a nucleotide sequence chosen from the group consisting of SEQ ID NOS:1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, and 29, or a complement of said nucleotide sequence.
11. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of: (a) a first nucleotide sequence comprising a coding sequence differing by one or more nucleotide sequences from a coding sequence encoding said amino acid sequence, provided that no more than 20% of the nucleotides in the coding sequence in said first nucleotide sequence differ from said coding sequence; (b) an isolated second polynucleotide that is a complement of the first polynucleotide; and (c) a nucleic acid fragment of (a) or (b).
12. A vector comprising the nucleic acid molecule of claim 11.
13. The vector of claim 12, further comprising a promoter operably-linked to said nucleic acid molecule.
14. A cell comprising the vector of claim 12.
15. An antibody that binds immunospecifically to the polypeptide of claim 1. 110 WO 01/46231 PCT/USOO/34898
16. The antibody of claim 15, wherein said antibody is a monoclonal antibody.
17. The antibody of claim 15, wherein the antibody is a humanized antibody.
18. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising: (a) providing the sample; (b) contacting the sample with an antibody that binds immunospecifically to the polypeptide; and (c) determining the presence or amount of antibody bound to said polypeptide, thereby determining the presence or amount of polypeptide in said sample.
19. A method for determining the presence or amount of the nucleic acid molecule of claim 5 in a sample, the method comprising: (a) providing the sample; (b) contacting the sample with a probe that binds to said nucleic acid molecule; and (c) determining the presence or amount of the probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
20. The method of claim 19 wherein presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type.
21. The method of claim 20 wherein the cell or tissue type is cancerous.
22. A method of identifying an agent that binds to a polypeptide of claim 1, the method comprising: (a) contacting said polypeptide with said agent; and (b) determining whether said agent binds to said polypeptide.
23. The method of claim 22 wherein the agent is a cellular receptor or a downstream effector. 111 WO 01/46231 PCT/USOO/34898
24. A method for identifying an agent that modulates the expression or activity of the polypeptide of claim 1, the method comprising: (a) providing a cell expressing said polypeptide; (b) contacting the cell with said agent, and (c) determining whether the agent modulates expression or activity of said polypeptide, whereby an alteration in expression or activity of said peptide indicates said agent modulates expression or activity of said polypeptide.
25. A method for modulating the activity of the polypeptide of claim 1, the method comprising contacting a cell sample expressing the polypeptide of said claim with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
26. A method of treating or preventing a FCTRX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the polypeptide of claim 1 in an amount sufficient to treat or prevent said FCTRX-associated disorder in said subject.
27. The method of claim 26 wherein the disorder is selected from the group consisting of diabetes, metabolic disturbances associated with obesity, the metabolic syndrome X, anorexia, wasting disorders associated with chronic diseases, metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias
28. The method of claim 26 wherein the disorder is related to organismal energy metabolism that effect adipose stores, muscle mass, insulin secretion, glucose utilization and serum lipid levels including triglycerides and cholesterol
29. The method of claim 26, wherein said subject is a human.
30. A method of treating or preventing a FCTRX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the 112 WO 01/46231 PCT/USOO/34898 nucleic acid of claim 5 in an amount sufficient to treat or prevent said FCTRX-associated disorder in said subject.
31. The method of claim 30 wherein the disorder is selected from the group consisting of diabetes, metabolic disturbances associated with obesity, the metabolic syndrome X, anorexia, wasting disorders associated with chronic diseases, metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.
32. The method of claim 30 wherein the disorder is related to organismal energy metabolism that effects adipose stores, muscle mass, insulin secretion, glucose utilization and serum lipid levels including, triglycerides and cholesterol
33. The method of claim 30, wherein said subject is a human.
34. A method of treating or preventing a FCTRX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the antibody of claim 15 in an amount sufficient to treat or prevent said FCTRX-associated disorder in said subject
35. The method of claim 34 wherein the disorder is selected from the group consisting of diabetes, metabolic disturbances associated with obesity, the metabolic syndrome X, anorexia, wasting disorders associated with chronic diseases, metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.
36. The method of claim 34 wherein the disorder is related to organismal energy metabolism that effects adipose stores, muscle mass, insulin secretion, glucose utilization and serum lipid levels including, triglycerides and cholesterol
37. The method of claim 34, wherein the subject is a human. 113 WO 01/46231 PCT/USOO/34898
38. A pharmaceutical composition comprising the polypeptide of claim 1 and a pharmaceutically-acceptable carrier.
39. A pharmaceutical composition comprising the nucleic acid molecule of claim 5 and a pharmaceutically-acceptable carrier.
40. A pharmaceutical composition comprising the antibody of claim 15 and a pharmaceutically-acceptable carrier.
41. A kit comprising in one or more containers, the pharmaceutical composition of claim 38.
42. A kit comprising in one or more containers, the pharmaceutical composition of claim 39.
43. A kit comprising in one or more containers, the pharmaceutical composition of claim 40.
44. A method for determining the presence of or predisposition to a disease associated with altered levels of the polypeptide of claim 1 in a first mammalian subject, the method comprising: (a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and (b) comparing the amount of said polypeptide in the sample of step (a) to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease; wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to said disease.
45. The method of claim 44 wherein the predisposition is to cancers. 114 WO 01/46231 PCT/USOO/34898
46. A method for determining the presence of or predisposition to a disease associated with altered levels of the nucleic acid molecule of claim 5 in a first mammalian subject, the method comprising: (a) measuring the amount of the nucleic acid in a sample from the first mammalian subject; and (b) comparing the amount of said nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
47. The method of claim 46 wherein the predisposition is to cancers.
48. A method of treating a pathological state in a mammal, the method comprising administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising an amino acid sequence of at least one of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 and 30, or a biologically active fragment thereof.
49. A method of treating a pathological state in a mammal, the method comprising administering to the mammal the antibody of claim 15 in an amount sufficient to alleviate the pathological state. 115
AU22871/01A 1999-12-21 2000-12-21 Novel proteins and nucleic acids encoding same Abandoned AU2287101A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
USNOTGIVEN 1999-09-23
US17132999P 1999-12-21 1999-12-21
US60171329 1999-12-21
PCT/US2000/034898 WO2001046231A2 (en) 1999-12-21 2000-12-21 Polynucleotides and polypeptides encoded thereby

Publications (1)

Publication Number Publication Date
AU2287101A true AU2287101A (en) 2001-07-03

Family

ID=22623353

Family Applications (1)

Application Number Title Priority Date Filing Date
AU22871/01A Abandoned AU2287101A (en) 1999-12-21 2000-12-21 Novel proteins and nucleic acids encoding same

Country Status (1)

Country Link
AU (1) AU2287101A (en)

Similar Documents

Publication Publication Date Title
US20050208040A1 (en) Polynucleotides and polypeptides encoded thereby
EP1222272A2 (en) Polypeptides and polynucleotides encoding same
AU779667B2 (en) Polynucleotides and polypeptides encoded thereby
US20070166756A1 (en) Polypeptides and polynucleotides encoding same
US20020137202A1 (en) Novel proteins and nucleic acids encoding same
AU1817201A (en) Proteins and nucleic acids encoding the same
US6992170B2 (en) Polypeptides and polynucleotides homologous to thymosin, ephrin a receptors, and fibromodulin
US20030082174A1 (en) Novel proteins and nucleic acids encoding same
US6855806B1 (en) Thymosin beta 10-like proteins and nucleic acids encoding same
US7001766B2 (en) Nucleic acid sequences encoding human angiopoietin-like polypeptides
WO2001074897A2 (en) Protein amf-1 to amf-10 and nucleic acids encoding the same
US20030082757A1 (en) Novel proteins and nucleic acids encoding same
AU2287101A (en) Novel proteins and nucleic acids encoding same
CA2384749A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
US20030198958A1 (en) Noval human proteins, polynucleotides encoding them and methods of using the same
US20030068671A1 (en) Novel proteins and nucleic acids encoding same
AU2001243444A1 (en) Novel proteins and nucleic acids encoding same
EP1259612A2 (en) G-protein coupled receptor related polypeptides
CA2425302A1 (en) Novel polypeptides homologous to thymosin, ephrin a, receptors, and fibromodulin, and polynucleotides encoding same
AU6899700A (en) Polynucleotides and polypeptides encoded thereby
AU2001249828A1 (en) Protein AMF-1 to AMF-10 nucleic acids encoding the same