Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TRAMDORINS AND METHODS OF USING TRAMDORINS
Document Type and Number:
WIPO Patent Application WO/2003/016502
Kind Code:
A2
Abstract:
A family of transmembrane domain proteins, the tramdorins, has been identified in human, mouse and rat. A variety of uses for these proteins is contemplated, including but not limited to peripheral myelination, the etiology of parathyroid adenomas, and the diagnosis and treatment of Central Nervous System (CNS) and non-CNS disorders.

Inventors:
BERMINGHAM JOHN R JR (US)
Application Number:
PCT/US2002/026637
Publication Date:
February 27, 2003
Filing Date:
August 21, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MCLAUGHLIN RES INST (US)
BERMINGHAM JOHN R JR (US)
International Classes:
C07K14/705; A61K38/00; (IPC1-7): C12N/
Attorney, Agent or Firm:
Brown, Thomas  et al W. (LLP 101 Howard Street, Suite 35, San Francisco CA, US)
Download PDF:
Claims:
CLAIMS
1. An isolated nucleic acid sequence selected from the group consisting of the cDNA sequence encoding mouse tramd 2 (SEQ ID NO : 1), the cDNA sequence encoding mouse tramd 3 (SEQ ID NO : 2), the cDNA sequence encoding human tramd 1 (SEQ ID NO : 3), the cDNA sequence encoding human tramd 2 (SEQ ID NO : 4), the cDNA sequence encoding human tramd 3 (SEQ ID NO : 5), the cDNA sequence encoding rat tramd 1 (SEQ ID NO : 6), the genomic sequence of mouse tramd 1 (SEQ ID NO : 7) and the genomic sequence of mouse tramd 3 (SEQ ID NO : 8).
2. The RNA transcribed from any of the sequences of Claim 1.
3. A vector comprising any of the sequences of Claim 1.
4. The vector of Claim 3, wherein the vector is an expression vector.
5. A host cell transfected by any of the vectors of Claim 3.
6. The transfected host cell of Claim 5, wherein said host cell is selected from the group consisting of COS7 cells, mouse embryonic stem cells and yeast cells.
7. A Xenopus oocyte injected with nucleic acid comprising any of the RNAs of Claim 2.
8. A method of detecting a tramdLSTK4/MrsI/Krs2 fusion in isolated human parathyroid adenomas comprising: (a) providing: (i) isolated tissue or cultured cells derived from human parathyroid adenomas, (ii) differentially labeled tramdL and STK4/Mrsl/Krs2 probes; (b) hybridizing said probes to said tissue, and; (c) detecting the hybridization pattern of said differentially labeled probes.
9. A method of identifying proteins which interact with mouse tramd 1, comprising (a) providing: (i) a first recombinant vector comprising a portion of mouse tramd 1 in operable combination with the DNA binding domain of a transcriptional activator, such that a chimeric protein will be expressed, (ii) a population of second recombinant vectors, wherein said population comprises a library of cDNA sequences in operable combination with the activation domain of a transcriptional activator, such that a population of chimeric proteins will be expressed, and (iii) a yeast host comprising a reporter gene in operable combination with the DNA binding sites for said transcriptional activator; (b) introducing said first vector and said population of second vectors into said yeast host to generate a population of transformed yeast; (c) subjecting said population of transformed to yeast to conditions such that said chimeric proteins are expressed; (d) screening said population for members which express said reporter gene, and; (e) isolating the coda fragment from said second vector from members of the population which express the reporter gene.
Description:
TRAMDORINS AND METHODS OF USING TRAMDORINS This invention was made with government support under grant R01-NS40751, from the National Institute of Neurological Disorders and Stroke. The government has certain rights in the invention.

FIELD OF THE INVENTION The present invention relates to a gene family, the tramdorins. The genes may be used in a variety of applications, including diagnostics and therapeutics.

BACKGROUND Identification of genes involved in nervous system function may provide markers for various steps in nervous system development and/or repair following disease or injury. Some of these steps may also be important in diseases affecting the nervous system, including but not limited to Charcot-Marie-Tooth syndrome, multiple sclerosis (MS) and Guillain-Barre syndrome, which are characterized by demyelination of axons in the peripheral or central nervous system. The frequencies of these diseases range from 1/1000 (U. S. ) for multiple sclerosis to 1. 5/100, 000 for Guillain-Barre syndrome. These diseases are devastating for those affected by them, often resulting in the loss of the use of limbs. For example, Guillain- Barre syndrome generally requires hospitalization to provide respiratory and cardiac support during an episode. In terms of economic costs, MS carries a substantial economic burden. A cost of illness (COI) study conducted by Bourdette et al. retrospectively examined costs to the US Veterans affairs for the treatment of 165 patients with MS over a 3-year period [Bourdette D. N. et al., Arch Phys Med Rehabil74 : 26-31 (1993) ]. Drug costs were not included in the study. The average cost to the VA associated with these patients was estimated at $35,000 per year.

Gene products of genes involved in nervous system function may also localize in tissues not associated with the nervous system and also may provide markers for various steps in the development of tissues not associated with the nervous system and/or repair of the same proximal to disease or injury.

What is needed are markers for various tissue processes (including but not limited to the nervous system) which can be used in the development of diagnostic and therapeutic agents for a variety of disorders including, but not limited to, pathologies of the nervous system.

SUMMARY OF THE INVENTION In several embodiments, isolated nucleic acid sequences encoding tramdorins are contemplated. In one embodiment, an isolated nucleic acid sequence selected from the group consisting of the cDNA sequence encoding mouse tramd 2 (SEQ ID NO : 1), the cDNA sequence encoding mouse tramd 3 (SEQ ID NO : 2), the cDNA sequence encoding human tramd 1 (SEQ ID NO : 3), the cDNA sequence encoding human tramd 2 (SEQ ID NO : 4), the cDNA sequence encoding human tramd 3 (SEQ ID NO : 5), the cDNA sequence encoding rat tramd 1 (SEQ ID NO : 6), the genomic sequence of mouse tramd 1 (SEQ ID NO : 7) and the genomic sequence of mouse tramd 3 (SEQ ID NO : 8) is contemplated.

In another embodiment, portions of nucleic acid sequences are contemplated, including, but not limited to, a portion of the cDNA sequence encoding mouse tramd2 (SEQ ID NO : 1), a portion of the cDNA sequence encoding mouse tramd3 (SEQ ID NO : 2), a portion of the cDNA sequence encoding human tramdl (SEQ ID NO : 3), a portion of the cDNA sequence encoding human tramd2 (SEQ ID NO : 4), a portion of the cDNA sequence encoding human tramd3 (SEQ ID NO : 5), a portion of the cDNA sequence encoding rat tramdl (SEQ ID NO : 6), a portion of genomic sequence encoding mouse tramdl (SEQ ID NO : 7) and a portion of the genomic sequence encoding mouse tramd3 (SEQ ID NO : 8). Polymorphisms within any of these sequences are also contemplated. Polymorphisms within the genomic sequences of human tramdl (SEQ ID NO : 9), human tramd2 (SEQ ID NO : 10), human tramd 3 (SEQ ID NO : 1 l) and human tramdL (SEQ ID NO : 12) are also contemplated.

In some embodiments, said nucleic acid sequences or portions of nucleic acid sequences may be fused to other nucleic acid sequences, such that gene fusions are provided.

In some embodiments, RNA transcribed from any of the sequences is contemplated. The RNA transcripts may be synthesized in vivo, or in vitro, by means known in the art. In some embodiments, RNA transcribed from the group consisting of mouse tramdorin 1 cDNA (SEQ

ID NO : 13), mouse tramdorin 2 cDNA (SEQ ID NO : 1), mouse tramdorin 3 cDNA (SEQ ID NO : 2), human tramdorin 1 cDNA (SEQ ID NO : 3), human tramdorin 2 cDNA (SEQ ID NO : 4) and human tramdorin 3 cDNA (SEQ ID NO : 5) is contemplated.

In some embodiments, a vector comprising said isolated nucleic acid sequences (for example, human tramd 1 cDNA (SEQ ID NO : 3), human tramd2 cDNA (SEQ ID NO : 4), human tramd3 cDNA (SEQ ID NO : 5), mouse tramd 1 cDNA (SEQ ID NO : 13), mouse tramd 2 cDNA (SEQ ID NO : 1), mouse tramd 3 cDNA (SEQ ID NO : 2), or portions thereof) is contemplated. In some embodiments, the vector is an expression vector. In some embodiments, the expression vector is suitable for expression of the nucleic acid sequences of interest in a eukaryotic host, while in other embodiments, the expression vector is suitable for expression in a prokaryotic host. Representative eukaryotic hosts and host cells for the expression vectors include, but are not limited to mouse, yeast, Xenopus and cultured mammalian cells.

In some embodiments, a host cell transfected by a vector comprising the nucleic acid sequences of interest is contemplated. In some embodiments, the host cell is a eukaryotic cell.

In some embodiments, the host cell is a Xenopus oocyte. In other embodiments, the host cell is a yeast cell. In yet other embodiments, the host cell is a mouse embryonic stem cell. Other host cells are contemplated, including but not limited to cultured mammalian cells (for example, COS-7 cells).

In some embodiments, a Xenopus oocyte is injected with nucleic acid comprising an RNA transcribed from a nucleic acid sequence of interest. In some embodiments, the RNA is a synthetic RNA and comprises additional flanking sequences and a cap.

In some embodiments, a method of identifying proteins which interact with tramdorins is contemplated. In one embodiment, the method comprises the yeast two-hybrid method. In one embodiment, the method comprises (a) providing: (i) a first recombinant vector comprising a portion of a tramdorin in operable combination with the DNA binding domain of a transcriptional activator, such that a chimeric protein will be expressed, (ii) a population of second recombinant vectors, wherein said population comprises a library of cDNA sequences in operable combination with the activation domain of a transcriptional activator, such that a population of chimeric proteins will be expressed, and (iii) a yeast host comprising a reporter gene in operable combination with the DNA binding sites for said transcriptional activator ; (b)

introducing said first vector and said population of second vectors into said yeast host to generate a population of transformed yeast; (c) subjecting said population of transformed to yeast to conditions such that said chimeric proteins are expressed; (d) screening said population for members which express said reporter gene, and; (e) isolating the cDNA fragment from said second vector from members of the population which express the reporter gene. In some embodiments, the portion of a tramdorin is a portion of mouse tramdorin 1 (SEQ ID NO : 14). In some embodiments, the portion of mouse tramdorin 1 may be the N-terminal 51 amino acids (SEQ ID NO : 15) or the first intracellular loop of tramdorin 1 (SEQ ID NO : 16). In other embodiments, the portion of the tramdorin is a portion of a tramdorin selected from the group consisting of mouse tramdorin 2 (SEQ ID NO : 17), mouse tramdorin 3 (SEQ ID NO : 18), human tramdorin 1 (SEQ ID NO : 19), human tramdorin 2 (SEQ ID NO : 20), human tramdorin 3 (SEQ ID NO : 21) and rat tramdorin 1 (SEQ ID NO : 22).

In other embodiments, a variation on the yeast two hybrid method is contemplated to detect proteins which interact with tramdorins. The method comprises providing (i) a first recombinant vector comprising a portion of a tramdorin in operable combination with Sos, such that a chimeric protein will be expressed, (ii) a population of second recombinant vectors, wherein said population comprises a library of cDNA sequences in operable combination with myristylation signal, such that a population of chimeric proteins will be expressed, and (iii) a yeast host comprising a temperature sensitive mutation in the cdc25 gene; (b) introducing said first vector and said population of second vectors into said yeast host to generate a population of transformed yeast; (c) subjecting said population of transformed to yeast to conditions such that said chimeric proteins are expressed and said myristylation fusions will be targeted to the membrane; (d) screening said population for members which are viable at elevated temperatures, and; (e) isolating the cDNA fragment from said second vector from members of the population which are viable at elevated temperatures. In some embodiments, said tramdorin is a portion of mouse tramdorin 1 (SEQ ID NO : 14). In other embodiments, said portion of tramdorin is selected from the group consisting of mouse tramdorin 2 (SEQ ID NO : 17), mouse tramdorin 3 (SEQ ID NO : 18), human tramdorin 1 (SEQ ID NO : 19), human tramdorin 2 (SEQ ID NO : 20), human tramdorin 3 (SEQ ID NO : 21) and rat tramdorin 1 (SEQ ID NO : 22).

In yet other embodiments, another variation on the yeast two-hybrid system is contemplated to identify proteins which interact with tramdorins. In some embodiments, the method comprises (a) providing (i) a first recombinant vector comprising a full length tramdorin, (ii) a population of second recombinant vectors comprising a library of cDNA sequences fused to a ras gene lacking a myristylation signal, (iii) a yeast host comprising a temperature sensitive cdc25 mutation; (b) introducing said first recombinant vector and said population of second recombinant vectors into said yeast host to generate a population of transformed yeast; (c) subjecting said population of transformed yeast to conditions such that said tramdorin protein is expressed and localizes to the membrane and said ras fusions are expressed; (d) screening said population for members which are viable at elevated temperatures, and; (e) isolating the cDNA fragment from said second vector from members of the population which are viable at elevated temperatures. In some embodiments, said expressed tramdorin protein is mouse tramdorin 1 (SEQ ID NO : 14). In other embodiments, said tramdorin is selected from the group consisting of mouse tramdorin 2 (SEQ ID NO : 17), mouse tramdorin 3 (SEQ ID NO : 18), human tramdorinl (SEQ ID NO : 19), human tramdorin 2 (SEQ ID NO : 20), human tramdorin 3 (SEQ ID NO : 21) and rat tramdorin 1 (SEQ ID NO : 22).

In some embodiments, methods to detect tramdorin ligands are contemplated. In one embodiment, the method comprises (a) providing (i) a recombinant expression vector comprising a tramdorin nucleic acid sequence, (ii) a host cell and (iii) labeled candidate ligand molecules; (b) introducing said recombinant expression vector into said host cell under conditions such that said tramdorin nucleic acid sequence is expressed; (c) contacting said tramdorin-expressing host cells with said labeled candidate ligand molecules, and; (d) measuring the relative uptake of said labeled candidate ligand molecules into said tramdorin- expressing host cells. In some embodiments, said host cells are COS-7 cells. In other embodiments, said host cells are Xenopus oocytes. In some embodiments, the tramdorin nucleic acid sequence is selected from the group consisting of mouse tramdorin 1 cDNA (SEQ ID NO : 13), mouse tramdorin 2 cDNA (SEQ ID NO : 1), mouse tramdorin 3 cDNA (SEQ ID NO : 2), human tramdorin 1 cDNA (SEQ ID NO : 3), human tramdorin 2 cDNA (SEQ ID NO : 4), human tramdorin 3 cDNA (SEQ ID NO : 5) and rat tramdorin 1 cDNA (SEQ ID NO : 6). In some embodiments, said labeled candidate ligand molecules are labeled with tritium [3H]. In some

embodiments, said labeled candidate ligand molecules include, but are not limited to, small neutral amino acids and y-aminobutyric acid.

In some embodiments, methods to detect myelination are contemplated. In such embodiments, the expression levels of tramdorin 1 are measured in tissue or cells following injury or a disease process. In some embodiments, the tissue is damaged nervous tissue. In other embodiments, the cells are Schwann cells co-cultured with neurons. In some embodiments, tramdorin expression levels are measured by detection of the levels of tramdorin 1 mRNA.

Tramdorin 1 mRNA levels are detected by Northern blot hybridization in some embodiments, while in other embodiments, amplification of reverse transcribed mRNA (i. e. amplification of cDNA) is contemplated.

In some embodiments, methods to detect tramdL fusions in parathyroid tumors, including but not limited to parathyroid adenomas are contemplated. In some embodiments, the tramdL fusion is a fusion (SEQ ID NO : 23) between human tramdL sequences and human STK4/Mst-l/Krs-2 sequences (located on human chromosome 5 and human chromosome 20 respectively). In some embodiments, the fusion is detected by amplification of fusion cDNA sequences. The method comprises (a) providing (i) human parathyroid tumor tissue and (ii) tramdL and STK4/Mst-l/Krs-2 primers; (b) isolating RNA from said parathyroid tumor tissue; (c) reverse-transcribing said RNA to generate cDNA sequences; (d) using said primers to amplify said cDNA, and; (e) detecting an amplification product. In other embodiments, the fusion is detected by fluorescence in situ hybridization (FISH). The method comprises; (a) providing: (i) isolated tissue derived from human parathyroid tumors and (ii) differentially labeled tramdL and STK4/Mst-l/Krs-2 probes; (b) hybridizing said probes to said tissue, and; (c) detecting the hybridization pattern of said differentially labeled probes. In some embodiments, the method provides cells cultured from parathyroid tumors in step (a). In some embodiments the tramdL probe comprises labeled sequences from a tramdL-containing bacterial artificial chromosome (BAC). In some embodiments, the label comprises biotin, while in other embodiments the label comprises a fluorescent moiety, including but not limited to fluorescein. In some embodiments, the STK4/Mst-l/Krs-2 probe comprises labeled sequences from a STK4/Mst-l/Krs-2 containing BAC. In some embodiments, the label comprises

digoxigenin, while in other embodiments the label comprises a fluorescent moiety, including but not limited to rhodamine.

In some embodiments, a transgenic animal model of parathyroid tumors, including parathyroid adenomas, is contemplated. In some embodiments, the transgenic animal is a transgenic mouse. The transgenic mouse comprises a recombinant construct wherein the STK4/Mst-l/Krs-2-tramdL fusion is expressed under the control of a sequence expressed primarily, if not exclusively, in the parathyroid. Thus, the transgenic animal comprises recombinant nucleic acid constructs comprising the STK4/Mst-l/Krs-2-tramdL fusion (SEQ ID NO : 38) operably linked to a regulatory sequence which enable parathyroid-specific expression of the fusion. In some embodiments, the regulatory sequence is the Gcm2 promoter (the Gcm2 cDNA sequence has the accession number AF081556 (SEQ ID NO : 24) ), while in other embodiments, the regulatory sequence is the regulatory region of the mouse parathyroid hormone gene (mouse parathyroid hormone cDNA has the accession number NM020623 (SEQ ID NO : 25) and mouse parathyroid hormone precursor, exons 2 and 3 cDNA has the accession number AF066075 (SEQ ID NO : 33). In some embodiments, the Gcm2 coding region is replaced with the STK4/Mst-l/Krs-2-tramdL fusion (SEQ ID NO : 38). In other embodiments, the parathyroid hormone coding region is replaced with the STK4/Mst-l/Krs-2- tramdL fusion (SEQ ID NO : 38). In some embodiments, the transgenic animals are contemplated for the testing of therapeutics. Methods of generating transgenic animal models for parathyroid adenomas are also contemplated.

In some embodiments, human tramdorin sequences are contemplated for use in detection of disease-associated mutations in an autosomal recessive Charcot-Marie-Tooth Syndrome demyelinating neuropathy which maps to 5q31-33. Human tramdorin exons can be isolated and sequenced in subjects with the disease and in subjects without the disease to identify mutations in the affected subjects, which are not present in the unaffected subjects.

In some embodiments, tramdorin is contemplated for use in assays of Schwann cell myelination. In some embodiments, tramdorin is overexpressed in isolated Schwann cells, which are co cultured with neurons. The rate of myelination is measured. In some embodiments, tramdorin is overexpressed from a recombinant expression vector comprising tramdorin cDNA. In some embodiments, said cDNA is selected from the group consisting of

SEQ ID NO : 13 and SEQ ID NO : 3. In some embodiments, the overexpressed tramdorin protein is mouse tramdorin 1 (SEQ ID NO : 14), while in other embodiments, the overexpressed tramdorin is human tramdorin 1 (SEQ ID NO : 19). In some embodiments, myelination is detected by Sudan black staining, while in other embodiments, myelination is detected by measuring Protein 0 (PO) expression.

In some embodiments, mice deficient in tramdorin 1 expression are contemplated. In some embodiments, tramdorin 1 sequences are deleted in the tramdorin 1-deficient mice, while in other embodiments, a portion of the tramdorin sequence is replaced by a marker gene, the expression of which is detectable. In some embodiments, said marker gene is the LacZ gene, the product of which, P-galactosidase, is detectable. In some embodiments, the deletion of tramdorin 1 sequences is mediated by a conditional knock-out of tramdorin 1 sequences, although a variety of means to accomplish tramdorin 1 deletion are also contemplated.

In some embodiments, portions of tramdorin nucleic acid sequences are contemplated for use in assays to detect tramdorin expression. In some embodiments, said nucleic acid sequences comprise portions of nucleic acid sequences selected from the group consisting of mouse tramdorin 1 cDNA (SEQ ID NO : 13), mouse tramdorin 2 cDNA (SEQ ID NO : 1), mouse tramdorin 3 cDNA (SEQ ID NO : 2), human tramdorin 1 cDNA (SEQ ID NO : 3), human tramdorin 2 cDNA (SEQ ID NO : 4), human tramdorin 3 cDNA (SEQ ID NO : 5) and rat tramdorin 1 cDNA (SEQ ID NO : 6). In yet other embodiments, said portions comprise 300 to 600 base pair fragments that contain the extreme C-terminal and 3'untranslated sequences. In some embodiments, tramdorin expression is detected in tissues. In some embodiments, the detection uses in situ hybridization to sections of fixed tissue. In some embodiments, the tissue comprises human tissue, while in other embodiments the tissue comprises mouse tissue, and in still other embodiments, the tissue comprises rat tissues. Any tissue is contemplated, including but not limited to brain tissue, peripheral nervous tissue and spinal cord tissue. Any method of tissue fixation is also contemplated, including formalin fixation.

In some embodiments, human tramdorin sequences are contemplated for use in the treatment of neuropathic pain.

In some embodiments, human tramdorin sequences are contemplated for use as a marker for bone growth, remodelling and/or osteoclast differentiation. In other embodiments, human tramdorin sequences are contemplated to regulate bone growth.

In some embodiments, human tramdorin sequences are contemplated for use as a marker for adipose tissue. In other embodiments, human tramdorin sequences are contemplated to regulate the proliferation and/or differentiation of adipocytes. In other embodiments human tramdorin sequences are contemplated to regulate the sequestration of glycine into adipocytes.

BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows RDA clone JBSN125; a clone that contains two distinct sequences, one of which is differentially expressed in wild-type and Oct-6 (-/-) siatic nerve and is homologous to multiple EST cDNA's. Figure 1A shows Southern blots of PCR-amplified cDNA from Oct- 6 wild-type (+/+) and mutant (-/-) sciatic nerve. Figure 1B diagrammatically shows cDNA fragments that correspond to RDA clone JBSN 125-10, which contains only differentially expressed sequences that are contained in JBSN125.

Figure 2 shows the sequences of mouse tramdorin 1 EST cDNA 1920302 (SEQ ID NO : 2), a putative rat tramdorin 1 cDNA (SEQ ID NO : 6), and the corresponding amino acid sequences (SEQ ID NO : 14 and SEQ ID NO : 22). Image Consortium numbers are used for EST cDNAs.

Figure 3 shows the structure predicted for the protein encoded by mouse tramdorin 1 EST cDNA 1920302. Figure 3A shows the putative amino acid sequence (SEQ ID NO : 14) encoded by the putative full length EST cDNA 1920302, with structure predictions. Figure 3B shows a diagram depicting the predicted topology of tramdorin 1.

Figure 4 shows a Northern blot depicting the tissue-specific expression of mouse tramdorin 1.

Figure 5 shows Northern blots depicting the expression of tramdorin 1 and other genes at various times following nerve injury. Figure 5A shows tramdorin 1 expression following transection injury. Figure 5B shows Oct-6 expression following transection injury. Figure 5C shows PO expression following transection injury. Figure 5D shows NGFR expression following transection injury. Figure 5E shows GADPH expression following transection injury.

Figure 5F shows tramdorin expression following crush injury. Figure 5G shows Oct-6 expression following crush injury. Figure 5H shows PO expression following crush injury.

Figure 51 shows NGFR expression following crush injury. Figure 5J shows GADPH expression following crush injury.

Figure 6 summarizes the mouse mapping results for tramdorin. Figure 6A summarizes the mapping data. Figure 6B shows a partial mouse chromosome 11 linkage map.

Figure 7 shows the cluster of four human tramdorin genes defined by human tramdorin EST cDNAs.

Figure 8 shows the organization of the human tramdorin loci. Figure 8A depicts the human tramdorin 1 locus. Figure 8B depicts the human tramdorin 2 locus. Figure 8C depicts the human tramdorin 3 locus. Figure 8D depicts EST cDNA 1388139, which contains sequences that correspond to both chromosome 5q and to the STK4/Mst-l/Krs-2 locus on chromosome 20.

Figure 9 depicts the human genomic sequence of the tramdorin gene region (SEQ ID NO : 26). Sequence between nucleotides 50001 and 300000 of Homo Sapiens chromosome 5 working draft sequence segment NT006951. 4 is shown.

Figure 10 depicts the putative cDNA sequence for human tramdorin 1 (SEQ ID NO : 3) and the corresponding amino acid sequence (SEQ ID NO : 19).

Figure 11 depicts the putative cDNA sequence for human tramdorin 2 (SEQ ID NO : 4) and the corresponding amino acid sequence (SEQ ID NO : 20).

Figure 12 depicts human STK4/Mst-l/Krs-2 and tramdL sequences. Figure 12A depicts genomic sequence from nucleotides 8189548 to 8196472 of human chromosome 20 contig NT011382 (SEQ ID NO : 27 (nucleotide sequence) and SEQ ID NO : 28 (corresponding amino acid sequence) ). Figure 12B depicts human chromosome 5 genomic sequence between nts 55336 and 56128 (SEQ ID NO : 29) on Celera scaffold sequence Gax2HTBL3TT27: 2500000- 3000000 and an open reading frame (SEQ ID NO : 30). Figure 12C depicts the fusion junction between the 5'splice site of STK4/Mst-l/Krs-2 exon 9 and a 3'splice site of tramdorin L (SEQ ID NO : 31 (nucleotide sequence) and SEQ ID NO : 32 (amino acid sequence) ). Figure 12D depictsdiagrams oftheproteins encodedbySTK4/Mst-l/Krs-2 andEST cDNA 1833139.

Figure 13 depicts Northern blots of the tissue-specific expression of human tramdorin genes. Figure 13A shows tramdorin 1 expression. Figure 13B shows tramd 2 expression. Figure 13C shows tramd3 expression. Figure 13D shows GADPH expression.

Figure 14 depicts the conservation between the putative translation products of human tramdorin 1 (SEQ ID NO : 19), human tramdorin 2 (SEQ ID NO : 20), and human tramdorin 3 (SEQ ID NO : 21).

Figure 15 depicts the conservation between putative tramdorin 1 proteins from human (SEQ ID NO : 19), rat (SEQ ID NO : 22), mouse (SEQ ID NO : 14), Drosophila melanogaster (SEQ ID NO : 34) and C. elegans (SEQ ID NO : 35).

Figure 16 depicts the conservation between human tramd 1 (SEQ ID NO : 19), human tramd 3 (SEQ ID NO : 21), and a rat (SEQ ID NO : 36), confirmed experimentally, and C. elegans (SEQ ID NO : 37), based on homology, VGAT.

Figure 17 depicts the organization of mouse tramdorin loci. Exons are denoted as boxes; filled boxes denote coding regions. Coordinates denote the location of the sequences on Celera mouse genome scaffold sequence GAxK02T2QP88 : 1-500000. A. Mouse tramdl. A partial restriction map is shown at the top of the figure. Shown below the map are three tramdl EST cDNAs that define tramdl exons. Genomic regions that were sequenced prior to the availability of the Celera mouse genome sequence are show as thick lines below the cDNAs, and below them, select subcloned fragments are shown. B. Mouse tramd2. Two mouse tramdorin2 RACE cDNA clones are shown. C. Mouse tramd3. A partial restriction map is shown at the top of the figure. Exons 2-8 were defined by homology to mouse tramdl or human tramd3. Three mouse tramdorin3 cDNA clones are shown; two are RACE clones, and one was amplified with tramd3-specific primers. Regions that were sequenced prior to the availability of the Celera mouse genome sequence are shown below the exons; these sequences were obtained from the ends of the cloned fragments shown at the bottom of the figure, or from the Ensembl mouse genome sequence database. Note that tramdorin3 is transcribed in the opposite direction from tramdorin and tramdorin2.

Figure 18 depicts the genomic sequence of mouse tramdorin 1 (SEQ ID NO : 7).

Figure 19 depicts the genomic sequence of mouse tramdorin 3 (SEQ ID NO : 8).

Figure 20 depicts the conservation between human (SEQ ID NO : 21) and mouse tramd 3 (SEQ ID NO : 18) proteins.

Figure 21 depicts the composite human tramd 3 cDNA sequence (SEQ ID NO : 5) and the corresponding amino acid sequence (SEQ ID NO : 21).

Figure 22 depicts the composite mouse tramdorin 2 cDNA (SEQ ID NO : 1) sequence and the corresponding amino acid sequence (SEQ ID NO : 17).

Figure 23 depicts mouse tramd3 cDNA sequences. Figure 23A depicts a composite mouse tramd 3 cDNA (SEQ ID NO : 2). Figure 23B depicts mouse tramd 3 3'RACE cDNA 58&num 37 (SEQ ID NO : 39). Figure 23C depicts mouse tramd 3 3'RACE cDNA 56#22 (SEQ ID NO : 40).

Figure 24 depicts the rat LYAAT-1 mRNA, and its translation. Figure 24A depicts the translation of rat LYAAT-1, Genebank Accesion No. AAK67316.1, (SEQ ID NO : 41). Figure 24B depicts the rat LYAAT-1, Genebank Accesion No. AF361239, mRNA (SEQ ID NO : 42).

Figure 25 depicts the tramdL EST cDNA 1388139 sequence (SEQ ID NO : 23) and the corresponding amino acid sequence (SEQ ID NO: 71).

Figure 26 depicts the predicted cDNA sequence of a STK4/Mst-l/Krs-2-tramdL fusion containing 5'STK4 sequences not included in EST cDNA 1388139 (SEQ ID NO: 38) and the corresponding amino acid sequence (SEQ ID NO: 44).

Figure 27 depicts mouse Gcm2 and Pth cDNA sequences. Figure 27A shows the mouse Gcm2 cDNA sequence, Accession AF081556 (SEQ ID NO : 24). Figure 27B shows mouse parathyroid hormone cDNA, Accession NM02063 (SEQ ID NO : 25). Figure 27C depicts mouse parathyroid hormone precursor, exons 2,3 and cDNA AF066075 (SEQ ID NO: 33).

Figure 28 depicts the phylogenetic relationship of tramdl to other cloned genes.

Specifically, this dendrogram illustrates that tramdl is closely related to, but distinct from tramd3/LYAAT-1, and that these proteins along with the hypothetical proteins CG13384 from Drosophila melanogaster, and T27A1. 5 from Caenorhabditis elegans, constitutes a new family of putative amino acid transport proteins. They are related to the yeast vacuolar amino acid transport proteins AVT3 and AVT4. Other AVT proteins, and vesicular GABA transport proteins are more distantly related. The glycine transporters Glytl and Glyt2 are shown also for comparison.

Figure 29 depicts immunoblot analysis of tramdorinl. Specifically this figure presents a Western blot analysis of anti-tramdorinl antiserum. 100 llg of lysates from (see, panel A) adult rat sciatic nerve and (see, panel B) 293 cells transfected with tramdorinl expression vector (+), and untransfected 293 cells (-) were used for western blot analysis of the tramdorinl antiserum (diluted 1: 1000) ; the blots were developed with chemiluminescence. See, panels C and D.

Micrographs showing transfected (panel C) or untransfected (panel D) 293 cells viewed by immunofluorescence using the anti-tramdorinl antiserum. Nuclei were visualized using DAPI.

Figure 30 depicts localization of tramdorinl in myelinating Schwann cells. These are images of unfixed teased fibers from adult rat sciatic nerve, double-labeled with a rabbit tramdorinl antiserum (see, panels A and D ; TRITC) and a mouse monoclonal antibody against MAG (see, panel B; FITC) or LAMP1 (see, panel E; FITC). Tramdorin-immunoreactivity is found at paranodes (arrows) and incisures (arrowheads), colocalizing with MAG (panels A-C), and in puncta along the outer surface. At paranodes (panels D-F), tramdorinl-immunostaining does not co-localize with that of LAMP 1, a lysosomal marker.

Figure 31 depicts darkfield micrographs of mouse bone and fat pad tissue hybridized with probes specific for tramdl, tramd2, and tramd3.

Figure 32 depicts the nucleic acid sequence of the mouse tramdl probe (SEQ ID NO : 71) used in the in situ hybridizations presented in Figure 31.

Figure 33 depicts the nucleic acid sequence of the mouse tramd2 probe (SEQ ID NO : 72) used in the in situ hybridizations presented in Figure 31.

Figure 34 depicts the nucleic acid sequence of the mouse tramd3 probe (SEQ ID NO : 73) used in the in situ hybridizations presented in Figure 31.

DEFINITIONS As used herein,"tramdorin","tramd"or"tramdorins"and"tramds"refers to a family of related genes and related proteins that have been identified in at least human, mouse and rat.

Tramdorin genes encode putative transmembrane proteins. At least four tramdorin family members exist: tramdorin 1 (tramdl), tramdorin 2 (tramd2), tramdorin 3 (tramd3) and tramdL.

Tramdorin 1 has been identified in human, mouse and rat. The cDNA sequences for each are presented in Figure 10 (putative cDNA for human tramdorinl (SEQ ID NO : 3) ), Figure 2 (mouse tramdorinl cDNA (SEQ ID NO : 13) ) and Figure 2 (putative rat tramdorin 1 cDNA (SEQ ID NO : 6) ). The mouse tramdorin 1 genomic sequence is presented in Figure 18 (SEQ ID NO : 7). The genomic sequence for human tramdorin 1 (SEQ ID NO : 9) is contained within the sequence presented in Figure 9 (SEQ ID NO : 26). Specifically, the human tramdorin 1 gene is predicted to be contained within nucleotides 214333-256951 of Figure 9, although it is also contemplated that additional regulatory sequences may lie outside of this span of nucleotides.

Tramd2 has been identified in human and mouse. The genomic sequence of human tramd 2 (SEQ ID NO : 10) is contained within the sequence presented in Figure 9 (SEQ ID NO : 26). Specifically, the human tramdorin 2 gene is predicted to be contained within nucleotides 258170-294569 of Figure 9, although it is also contemplated that additional regulatory regions may lie outside of this span of nucleotides. The cDNA sequence encoding tramd2 in human is presented in Figure 11 (putative human tramd2 cDNA sequence (SEQ ID NO : 4) ) and the cDNA sequence encoding tramd 2 in mouse is presented in Figure 22 (composite mouse tramdorin2 cDNA (SEQ ID NO : 1)).

Tramdorin3 has also been identified in human, rat and mouse. The genomic sequence encoding tramd3 in human (SEQ ID NO: 11) is contained in the sequence presented in Figure 9 (SEQ ID NO : 26) (note that tramd3 is shown in the antisense orientation in this figure).

Specifically, the human tramdorin 3 gene is predicted to be contained within nucleotides 58749-136740 of Figure 9, although it is also contemplated that additional regulatory regions may lie outside of this span of nucleotides. The genomic sequence encoding tramd 3 in mouse is shown in Figure 19 (genomic sequence of mouse tramdorin 3 (SEQ ID NO : 8) ). The cDNA sequence encoding human tramd 3 is presented in Figure 21 (composite human tramd 3 cDNA

(SEQ ID NO : 5)). The coding region of the cDNA sequence encoding mouse tramd 3 is shown in Figure 23A (composite mouse tramd 3 cDNA (SEQ ID NO : 2) ). The mRNA sequence (SEQ ID NO : 42), and its translation (SEQ ID NO : 41), of rat LYAAT-1 (isolated by Sagne et al., PNAS USA 98: 7206 (2001) ), which appears to be a rat homolog of tramd 3 is presented in Figure 24.

An additional tramdorin family member has been identified in human, tramdL, which exists as a splice junction fusion between a tramdorin 3'exon (SEQ ID NO : 29) (Figure 12B) and 5'STK4/Mst-l/Krs-2 sequences encoded on human chromosome 20 (SEQ ID NO : 27) (Figure 12A). The cDNA sequence of the fusion junction is shown in Figure 12 C (SEQ ID NO : 31), and the putative STK4/Mst-l/Krs-2-tramdL fusion protein is depicted in Figure 12 D.

The cDNA sequence of EST cDNA 1388139 (SEQ ID NO : 23) is depicted in Figure 25, as is the corresponding predicted amino acid sequence (SEQ ID NO : 71).

With respect to nucleic acid sequences, one of skill in the art will appreciate that poplymorphisms are often found in populations, and any such polymorphisms contained within any of the tramdorin genomic or cDNA sequences are also within the scope of the present invention.

The predicted proteins encoded by human tramdorins 1 (SEQ ID NO : 19), 2 (SEQ ID NO : 20) and 3 (SEQ ID NO : 21) are depicted in Figure 14. The putative amino acid sequence encoded by mouse tramdl cDNA is depicted in Figure 3 (SEQ ID NO : 14). The amino acid sequences for human (SEQ ID NO : 21) and mouse (SEQ ID NO : 18) tramd3 proteins are shown in Figure 20. In addition to the human, rat and mouse tramdorins described above, tramdorins appear to exist in other species. For example, homologous sequences have been identified in Drosophila melanogaster (Figure 15) and Caenorhabditis elegans (Figure 15). Thus one of skill in the art will understand that tramdorin family members may be identified based on homology to the sequences described above.

The putative amino acid sequence (corresponding to the sequence labeled"1920303"in Figure 3A) encoded by mouse tramdl cDNA is depicted in Figure 3 (SEQ ID NO : 14). This sequence has been assigned GenBank accession number AF12429. That is to say, the amino acid sequence referenced by GenBank accession number AF12429 is identical to the sequence labeled"1920303"in Figure 3A. This mouse cDNA encodes a 478AA protein.

The amino acid sequences for human (SEQ ID NO : 21) and mouse (SEQ ID NO : 18) tramd3 proteins are shown in Figure 20. In addition to the human, rat and mouse tramdorins described above, tramdorins appear to exist in other species. For example, homologous sequences have been identified in Drosophila melanogaster (; Figure 15) and Cae7lorhabditis elegans (Figure 15). Thus one of skill in the art will understand that tramdorin family members may be identified based on homology to the sequences described above.

As used herein, "derivatives"of tramdorin proteins can refer to a number of alterations in such proteins. In some embodiments, the derivatives comprise proteins with amino acid sequence changes. Such changes can be conservative amino acid substitutions, amino acid deletions or amino acid insertions, provided that the activity of the tramdorin is retained.

Preferably, the alterations are conservative amino acid changes. For example, it is contemplated that an isolated replacement of a leucine with an isoleucine or valine, an alanine with a glycine, a threonine with a serine or a similar replacement of an amino acid with a structurally related amino acid (i. e. conservative substitutions) will not have a major effect on the biological activity of the resulting molecule. Conservative substitutions are those that take place within a family of amino acids that are related by their side chains. Amino acids can be divided into four families : (1) acidic (aspartate, glutamate) ; (2) basic (lysine, arginine, histidine) ; (3) nonpolar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); and (4) uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine).

Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. In an alternative, yet similar fashion, the amino acid repertoire can be grouped as: (1) acidic (aspartate, glutamate) ; (2) basic (lysine, arginine, histidine); (3) aliphatic (glycine, alanine, valine, leucine, isoleucine, serine, threonine), with serine and threonine optionally grouped separately as aliphatic-hydroxyl; (4) aromatic (phenylalanine, tyrosine, tryptophan) ; (5) amide (asparagine, glutamine); and (6) sulfur-containing (cysteine and methionine) (See e. g.

Stryer ed. , Biochemistry, 2E, WH Freeman and Co. (1981) pp. 13-16). Thus, in certain embodiments, modifications of the tramdorin protein sequence (s) is contemplated by the present invention. Guidance in determining which and how many amino acid residues may be substituted, inserted, or deleted without abolishing biological activity may be found by using

computer programs well known in the art, for example, DNAStar software or GCG (Univ. of Wisconsin).

As used herein, "central nervous system", or"CNS"refers to the brain and spinal cord.

The tissues of the CNS comprise both neurons and accessory, or"glial"cells. The accessory cells of the CNS are referred to as"oligodendrocytes", which form insulating myelin sheaths around the axons of many neurons, and"astrocytes"which provide structural and metabolic support for neurons and also induce tight junctions between cells lining the capillaries in the brain.

As used herein, the"peripheral nervous system", or"PNS"refers to the nerves and supporting cells that communicate motor and sensory signals between the CNS and the rest of the body. The supporting cells of the PNS are referred to as"Schwann cells", which form the insulating myelin sheaths around the axons of many neurons.

As used herein,"myelin"refers to the lipoproteinaceous material, composed of regularly alternating membranes of lipid lamellae (consisting of cholesterol, phospholipids, sphingolipids, phosphatidates) and protein, of the myelin sheath.

As used herein, the term"demyelinating disease"refers to any pathological process that results in the degradation or loss of the myelin sheath surrounding an axon, including, but not limited to Multiple Sclerosis, Charcot-Marie Tooth (CMT) and Guillain-Barre syndrome.

As used herein, the term"gene"refers to a DNA sequence that comprises control and coding sequences necessary for the production of a polypeptide or protein precursor. The polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence, as long as the desired protein activity is retained.

"Nucleoside", as used herein, refers to a compound consisting of a purine [guanine (G) or adenine (A) ] or pyrimidine [thymine (T), uridine (U), or cytidine (C) ] base covalently linked to a pentose, whereas"nucleotide"refers to a nucleoside phosphorylated at one of its pentose hydroxyl groups.

A"nucleic acid", as used herein, is a covalently linked sequence of nucleotides in which the 3'position of the pentose of one nucleotide is joined by a phosphodiester group to the 5' position of the pentose of the next, and in which the nucleotide residues (bases) are linked in specific sequence; i. e., a linear order of nucleotides. A"polynucleotide", as used herein, is a

nucleic acid containing a sequence that is greater than about 100 nucleotides in length. An "oligonucleotide", as used herein, is a short polynucleotide or a portion of a polynucleotide. An oligonucleotide typically contains a sequence of about two to about one hundred bases. The word"oligo"is sometimes used in place of the word"oligonucleotide". Nucleic acid molecules are said to have a"5'-terminus" (5'end) and a"3'-terminus" (3'end) because nucleic acid phosphodiester linkages occur to the 5'carbon and 3'carbon of the pentose ring of the substituent mononucleotides. The end of a nucleic acid at which a new linkage would be to a 5' pentose carbon is its 5'terminal nucleotide (by convention sequences are written, from right to left, in the 5'to 3'direction). The end of a nucleic acid at which a new linkage would be to a 3' pentose carbon is its 3'terminal nucleotide. A terminal nucleotide, as used herein, is the nucleotide at the end position of the 3'-or 5'-terminus. DNA molecules are said to have"5' ends"and"3'ends"because mononucleotides are reacted to make oligonucleotides in a manner such that the 5'phosphate of one mononucleotide pentose ring is attached to the 3'oxygen of its neighbor in one direction via a phosphodiester linkage. Therefore, an end of an oligonucleotide is referred to as the"5'end"if its 5'phosphate is not linked to the 3'oxygen of a mononucleotide pentose ring and as the"3'end"if its 3'oxygen is not linked to a 5'phosphate of a subsequent mononucleotide pentose ring.

As used herein, a nucleic acid sequence, even if internal to a larger oligonucleotide or polynucleotide, also may be said to have 5'and 3'ends. In either a linear or circular DNA molecule, discrete elements are referred to as being"upstream"or 5'of the"downstream"or 3' elements. This terminology reflects the fact that transcription proceeds in a 5'to 3'fashion along the DNA strand. Typically, promoter and enhancer elements that direct transcription of a linked gene are generally located 5'or upstream of the coding region. However, enhancer elements can exert their effect even when located 3'of the promoter element and the coding region. Transcription termination and polyadenylation signals are located 3'or downstream of the coding region.

The term"wild-type"when made in reference to a gene refers to a gene which has the characteristics of a gene isolated from a naturally occurring source. The term"wild-type"when made in reference to a gene product refers to a gene product which has the characteristics of a gene product isolated from a naturally occurring source. A wild-type gene is that which is most

frequently observed in a population and is thus arbitrarily designated the"normal"or"wild- type"form of the gene. In contrast, the term"modified"or"mutant"when made in reference to a gene or to a gene product refers, respectively, to a gene or to a gene product which displays modifications in sequence and/or functional properties (i. e. , altered characteristics) when compared to the wild-type gene or gene product. It is noted that naturally-occurring mutants can be isolated; these are identified by the fact that they have altered characteristics when compared to the wild-type gene or gene product. For some variants (i. e. modified or mutant forms of a gene), there is no selection against individuals carrying the variant form, and so both forms (wild-type and modified) may be relatively common in the general population. This is referred to as"genetic polymorphism", which is defined herein as a genetic variation in which the frequency of two or more forms, or alleles, is at least 1% in the population. Polymorphisms within the tramdorin sequences are specifically contemplated.

As used herein in reference to a nucleic acid sequence (for example a cDNA or genomic sequence), the term"portion" (as in"a portion of a genomic sequence") refers to fragments of that nucleic acid sequence. The fragments may range in size from five nucleotides to the entire nucleic acid sequence minus one nucleotide.

The term"antisense"as used herein refers to a deoxyribonucleotide sequence whose sequence of deoxyribonucleotide residues is in reverse 5'to 3'orientation in relation to the sequence of deoxyribonucleotide residues in a sense strand of a DNA duplex. Antisense also refers to the"reverse complement orientation". By way of example, the reverse complement of the nucleotide sequence 5'-GATCC-3'is 5'-CTAGG-3'. A"sense strand"of a DNA duplex refers to a strand in a DNA duplex which is transcribed by a cell in its natural state into a"sense mRNA."Thus an"antisense"sequence is a sequence having the same sequence as the non- coding strand in a DNA duplex. The term"antisense RNA"refers to a RNA transcript that is complementary to all or part of a target primary transcript or mRNA and that blocks the expression of a target gene by interfering with the processing, transport and/or translation of its primary transcript or mRNA. The complementarity of an antisense RNA may be with any part of the specific gene transcript, i. e. , at the 5'non-coding sequence, 3'non-coding sequence, introns, or the coding sequence. In addition, as used herein, antisense RNA may contain regions of ribozyme sequences that increase the efficacy of antisense RNA to block gene

expression."Ribozyme"refers to a catalytic RNA and includes sequence-specific endoribonucleases. "Antisense inhibition"refers to the production of antisense RNA transcripts capable of preventing the expression of the target protein.

As used herein, the term"overexpression"refers to the production of a gene product in transgenic organisms that exceeds levels of production in normal or non-transformed organisms. As used herein, the term"altered levels"refers to the production of gene product (s) in transgenic organisms in amounts or proportions that differ from that of normal or non- transformed organisms.

The term"recombinant"when made in reference to a DNA molecule refers to a DNA molecule which is comprised of segments of DNA joined together by means of molecular biological techniques. The term"recombinant"when made in reference to a protein or a polypeptide refers to a protein molecule which is expressed using a recombinant DNA molecule.

The term"nucleotide sequence of interest"refers to any nucleotide sequence, the manipulation of which may be deemed desirable for any reason (e. g., confer improved qualities), by one of ordinary skill in the art. Such nucleotide sequences include, but are not limited to, coding sequences of structural genes (e. g. , reporter genes, selection marker genes, oncogenes, drug resistance genes, growth factors, etc.), and non-coding regulatory sequences which do not encode an mRNA or protein product, (e. g., promoter sequence, polyadenylation sequence, termination sequence, enhancer sequence, etc.).

As used herein the term"coding region"when used in reference to structural gene refers to the nucleotide sequences which encode the amino acids found in the nascent polypeptide as a result of translation of a mRNA molecule. Typically, the coding region is bounded on the 5' side by the nucleotide triplet"ATG"which encodes the initiator methionine and on the 3'side by a stop codon (e. g., TAA, TAG, TGA). In some cases the coding region is also known to initiate by a nucleotide triplet"TTG".

As used herein, the terms"complementary"or"complementarity"when used in reference to polynucleotides refer to polynucleotides which are related by the base-pairing rules.

For example, for the sequence 5'-AGT-3'is complementary to the sequence 5'-ACT-3'.

Complementarity may be"partial, "in which only some of the nucleic acids'bases are matched

according to the base pairing rules. Or, there may be"complete"or"total"complementarity between the nucleic acids. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands.

This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.

A"complement"of a nucleic acid sequence as used herein refers to a nucleotide sequence whose nucleic acids show total complementarity to the nucleic acids of the nucleic acid sequence.

The term"homology"when used in relation to nucleic acids refers to a degree of complementarity. There may be partial homology or complete homology (i. e. , identity).

"Sequence identity"refers to a measure of relatedness between two or more nucleic acids or proteins, and is given as a percentage with reference to the total comparison length. The identity calculation takes into account those nucleotide or amino acid residues that are identical and in the same relative positions in their respective larger sequences. Calculations of identity may be performed by algorithms contained within computer programs such as"GAP" (Genetics Computer Group, Madison, Wis. ) and"ALIGN" (DNAStar, Madison, Wis. ). A partially complementary sequence is one that at least partially inhibits (or competes with) a completely complementary sequence from hybridizing to a target nucleic acid is referred to using the functional term"substantially homologous. "The inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or Northern blot, solution hybridization and the like) under conditions of low stringency. A substantially homologous sequence or probe will compete for and inhibit the binding (i. e. , the hybridization) of a sequence which is completely homologous to a target under conditions of low stringency. This is not to say that conditions of low stringency are such that non-specific binding is permitted ; low stringency conditions require that the binding of two sequences to one another be a specific (i. e., selective) interaction. The absence of non-specific binding may be tested by the use of a second target which lacks even a partial degree of complementarity (e. g., less than about 30% identity); in the absence of non-specific binding the probe will not hybridize to the second non-complementary target.

When used in reference to a double-stranded nucleic acid sequence such as a cDNA or genomic clone, the term"substantially homologous"refers to any probe which can hybridize to either or both strands of the double-stranded nucleic acid sequence under conditions of low stringency as described infra.

Low stringency conditions when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42°C in a solution consisting of 5X SSPE (43.8 g/1 NaCI, 6.9 g/1 NaH2PO4H2O and 1.85 g/1 EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5X Denhardt's reagent [50X Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; Sigma) ] and 100 pg/ml denatured salmon sperm DNA followed by washing in a solution comprising 5X SSPE, 0.1% SDS at 42°C when a probe of about 500 nucleotides in length is employed.

High stringency conditions when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42°C in a solution consisting of 5X SSPE (43.8 g/1 NaCI, 6.9 g/1 NaH2PO4tH2O and 1. 85 g/1 EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5X Denhardt's reagent and 100 g/ml denatured salmon sperm DNA followed by washing in a solution comprising 0. 1X SSPE, 1.0% SDS at 42°C when a probe of about 500 nucleotides in length is employed.

When used in reference to nucleic acid hybridization the art knows well that numerous equivalent conditions may be employed to comprise either low or high stringency conditions; factors such as the length and nature (DNA, RNA, base composition) of the probe and nature of the target (DNA, RNA, base composition, present in solution or immobilized, etc. ) and the concentration of the salts and other components (e. g., the presence or absence of formamide, dextran sulfate, polyethylene glycol) are considered and the hybridization solution may be varied to generate conditions of either low or high stringency hybridization different from, but equivalent to, the above listed conditions.

"Stringency"when used in reference to nucleic acid hybridization typically occurs in a range from about Tm-5°C (5°C below the Tm of the probe) to about 20°C to 25°C below Tm. As will be understood by those of skill in the art, a stringent hybridization can be used to identify or detect identical polynucleotide sequences or to identify or detect similar or related

polynucleotide sequences. Under"stringent conditions"a nucleic acid sequence of interest will hybridize to its exact complement and closely related sequences.

Polypeptide molecules are said to have an"amino terminus" (N-terminus) and a "carboxy terminus" (C-terminus) because peptide linkages occur between the backbone amino group of a first amino acid residue and the backbone carboxyl group of a second amino acid residue. Typically, the terminus of a polypeptide at which a new linkage would be to the carboxy-terminus of the growing polypeptide chain, and polypeptide sequences are written from left to right beginning at the amino terminus.

As used herein in reference to an amino acid sequence or a protein, the term"portion" (as in"a portion of an amino acid sequence") refers to fragments of that protein. The fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.

As used herein, the term"fusion protein"refers to a chimeric protein containing the protein of interest (e. g., tramdorins and fragments thereof) joined to an exogenous protein fragment (e. g. , the fusion partner which consists of a non-tramdorin protein). The fusion partner may enhance the solubility of tramdorin protein as expressed in a host cell, may provide an affinity tag to allow purification of the recombinant fusion protein from the host cell or culture supernatant, or both. The fusion partner may also enable screening assays, such as the yeast two-hybrid assay. If desired, the fusion protein may be removed from the protein of interest (e. g. , tramdorins or fragments thereof) by a variety of enzymatic or chemical means known to the art.

The term"isolated"when used in relation to a nucleic acid, as in"an isolated nucleic acid sequence"refers to a nucleic acid sequence that is identified and separated from at least one contaminant nucleic acid with which it is ordinarily associated in its natural source.

Isolated nucleic acid is nucleic acid present in a form or setting that is different from that in which it is found in nature. In contrast, non-isolated nucleic acids are nucleic acids such as DNA and RNA which are found in the state they exist in nature. For example, a given DNA sequence (e. g., a gene) is found on the host cell chromosome in proximity to neighboring genes; RNA sequences, such as a specific mRNA sequence encoding a specific protein, are found in the cell as a mixture with numerous other mRNAs which encode a multitude of proteins.

However, an isolated nucleic acid sequence encoding human tramdorin 1 (for example) includes, by way of example, such nucleic acid sequences in cells which ordinarily contain the sequence encoding human tramdorin 1, where the nucleic acid sequence is in a chromosomal or extrachromosomal location different from that of natural cells, or is otherwise flanked by a different nucleic acid sequence than that found in nature. The isolated nucleic acid sequence may be present in single-stranded or double-stranded form. When an isolated nucleic acid sequence is to be utilized to express a protein, the nucleic acid sequence will contain at a minimum at least a portion of the sense or coding strand (i. e., the nucleic acid sequence may be single-stranded). Alternatively, it may contain both the sense and anti-sense strands (i. e., the nucleic acid sequence may be double-stranded).

As used herein, the term"purified"refers to molecules, either nucleic or amino acid sequences, that are removed from their natural environment, isolated or separated. An"isolated nucleic acid sequence"is therefore a purified nucleic acid sequence. "Substantially purified" molecules are at least 60% free, preferably at least 75% free, and more preferably at least 90% free from other components with which they are naturally associated.

As used herein, the terms"vector"and"vehicle"are used interchangeably in reference to nucleic acid molecules that transfer DNA segment (s) from one cell to another. Vectors may include plasmids, bacteriophages, viruses, cosmids, and the like.

The term"expression vector"or"expression cassette"as used herein refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.

The terms"targeting vector"and"targeting construct"refer to nucleic acid sequences comprising a sequence of interest flanked on either side by recognition sequences that are capable of homologous recombination with cognate sequences in the genome in such a way that the sequence of interest replaces any DNA sequences that are located between the cognate sequences in the genome. The sequence of interest can consist of recognition sites for

restriction enzymes or site specific recombinases such as Flp or Cre, exongenous genes including but not limited to those that encode thymidinekinase or confer resistance to antibiotics such as neomycin and hygromycin, marker genes such as LacZ and eGFP, as well as portions of the targeted gene itself or sequences from other genes of interest.

The terms"in operable combination", "in operable order"and"operably linked"as used herein refer to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.

The term"selectable marker"as used herein, refer to a gene which encodes an enzyme having an activity that confers resistance to an antibiotic or drug upon the cell in which the selectable marker is expressed. Selectable markers may be"positive"or"negative."Examples of positive selectable markers include the neomycin phosphotrasferase (NPTIT) gene which confers resistance to G418 and to kanamycin, and the bacterial hygromycin phosphotransferase gene (hyg), which confers resistance to the antibiotic hygromycin. Negative selectable markers encode an enzymatic activity whose expression is cytotoxic to the cell when grown in an appropriate selective medium. For example, the HSV-tk gene is commonly used as a negative selectable marker. Expression of the HSV-tk gene in cells grown in the presence of gancyclovir or acyclovir is cytotoxic; thus, growth of cells in selective medium containing gancyclovir or acyclovir selects against cells capable of expressing a functional HSV TK enzyme.

Transcriptional control signals in eukaryotes comprise"promoter"and"enhancer" elements. Promoters and enhancers consist of short arrays of DNA sequences that interact specifically with cellular proteins involved in transcription (Maniatis, et al., Science 236: 1237, 1987). Promoter and enhancer elements have been isolated from a variety of eukaryotic sources including genes in yeast, insect, mammalian and plant cells. Promoter and enhancer elements have also been isolated from viruses and analogous control elements, such as promoters, are also found in prokaryotes. The selection of a particular promoter and enhancer depends on the cell type used to express the protein of interest. Some eukaryotic promoters and enhancers have a broad host range while others are functional in a limited subset of cell types (for review, see Voss, et al., Trends Biochem. Sci. , 11: 287, 1986; and Maniatis, et al., supra 1987).

The terms"promoter element, ""promoter,"or"promoter sequence"as used herein, refer to a DNA sequence that is located at the 5'end (i. e. precedes) the protein coding region of a DNA polymer. The location of most promoters known in nature precedes the transcribed region. The promoter functions as a switch, activating the expression of a gene. If the gene is activated, it is said to be transcribed, or participating in transcription. Transcription involves the synthesis of mRNA from the gene. The promoter, therefore, serves as a transcriptional regulatory element and also provides a site for initiation of transcription of the gene into mRNA.

Promoters may be tissue specific or cell specific. The term"tissue specific"as it applies to a promoter refers to a promoter that is capable of directing selective expression of a nucleotide sequence of interest to a specific type of tissue in the relative absence of expression of the same nucleotide sequence of interest in a different type of tissue. Tissue specificity of a promoter may be evaluated by, for example, operably linking a reporter gene to the promoter sequence to generate a reporter construct, introducing the reporter construct into the genome of an organism such that the reporter construct is integrated into every tissue of the resulting transgenic organism, and detecting the expression of the reporter gene (e. g., detecting mRNA, protein, or the activity of a protein encoded by the reporter gene) in different tissues of the transgenic. The detection of a greater level of expression of the reporter gene in one or more tissues relative to the level of expression of the reporter gene in other tissues shows that the promoter is specific for the tissues in which greater levels of expression are detected. The term "cell type specific"as applied to a promoter refers to a promoter which is capable of directing selective expression of a nucleotide sequence of interest in a specific type of cell in the relative absence of expression of the same nucleotide sequence of interest in a different type of cell within the same tissue. The term"cell type specific"when applied to a promoter also means a promoter capable of promoting selective expression of a nucleotide sequence of interest in a region within a single tissue. Cell type specificity of a promoter may be assessed using methods well known in the art, e. g., immunohistochemical staining. Briefly, tissue sections are embedded in paraffin, and paraffin sections are reacted with a primary antibody which is specific for the polypeptide product encoded by the nucleotide sequence of interest whose expression is controlled by the promoter. A labeled (e. g., peroxidase conjugated) secondary

antibody which is specific for the primary antibody is allowed to bind to the sectioned tissue and specific binding detected (e. g., with avidin/biotin) by microscopy. Alternatively, mRNA localization can be determined by in situ hybridization.

Promoters may be constitutive or regulatable. The term"constitutive"when made in reference to a promoter means that the promoter is capable of directing transcription of an operably linked nucleic acid sequence in the absence of a stimulus (e. g., heat shock, chemicals, light, etc.). Typically, constitutive promoters are capable of directing expression of a transgene in substantially any cell and any tissue.

In contrast, a"regulatable"promoter is one which is capable of directing a level of transcription of an operably linked nuclei acid sequence in the presence of a stimulus (e. g., heat shock, chemicals, light, etc.) which is different from the level of transcription of the operably linked nucleic acid sequence in the absence of the stimulus.

As used herein, the term"regulatory element"refers to a genetic element that controls some aspect of the expression of nucleic acid sequence (s). For example, a promoter is a regulatory element that facilitates the initiation of transcription of an operably linked coding region. Other regulatory elements are splicing signals, polyadenylation signals, termination signals, etc.

The enhancer and/or promoter may be"endogenous"or"exogenous"or"heterologous." An"endogenous"enhancer or promoter is one that is naturally linked with a given gene in the genome. An"exogenous"or"heterologous"enhancer or promoter is one that is placed in juxtaposition to a gene by means of genetic manipulation (i. e. , molecular biological techniques) such that transcription of the gene is directed by the linked enhancer or promoter. For example, an endogenous promoter in operable combination with a first gene can be isolated, removed and placed in operable combination with a second gene, thereby making it a"heterologous" promoter in operable combination with said second gene. A variety of such combinations are contemplated (e. g. the first and second genes can be from the same species, or from different species).

The presence of"splicing signals"on an expression vector often results in higher levels of expression of the recombinant transcript in eukaryotic host cells. Splicing signals mediate the removal of introns from the primary RNA transcript and consist of a splice donor and

acceptor site (Sambrook, et al., Molecular Cloning : A Laboratory Manual, 2nd ed. , Cold Spring Harbor Laboratory Press, New York [1989] pp. 16.7-16. 8). A commonly used splice donor and acceptor site is the splice junction from the 16S RNA of SV40.

Efficient expression of recombinant DNA sequences in eukaryotic cells requires expression of signals directing the efficient termination and polyadenylation of the resulting transcript. Transcription termination signals are generally found downstream of the polyadenylation signal and are a few hundred nucleotides in length. The term"poly (A) site"or "poly (A) sequence"as used herein denotes a DNA sequence which directs both the termination and polyadenylation of the nascent RNA transcript. Efficient polyadenylation of the recombinant transcript is desirable, as transcripts lacking a poly (A) tail are unstable and are rapidly degraded. The poly (A) signal utilized in an expression vector may be"heterologous"or "endogenous. "An endogenous poly (A) signal is one that is found naturally at the 3'end of the coding region of a given gene in the genome. A heterologous poly (A) signal is one which has been isolated from one gene and positioned 3'to another gene. A commonly used heterologous poly (A) signal is the SV40 poly (A) signal. The SV40 poly (A) signal is contained on a 237 bp BamHIlBclI restriction fragment and directs both termination and polyadenylation (Sambrook, supra, at 16.6-16. 7).

The term"transfection"as used herein refers to the introduction of foreign DNA into eukaryotic cells. Transfection may be accomplished by a variety of means known to the art including calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, retroviral infection, and biolistics. When introducing foreign DNA into yeast cells, the term"transformation"is commonly used.

The term"transgenic"when used in reference to a cell refers to a cell which contains a transgene, or whose genome has been altered by the introduction of a transgene. The term "transgenic"when used in reference to a tissue or to an organism, such as a mouse, refers to a tissue or organism, respectively, which comprises one or more cells that contain a transgene, or whose genome has been altered by the introduction of a transgene. Transgenic cells, tissues and organisms may be produced by several methods including the introduction of a"transgene" comprising nucleic acid (usually DNA) into a target cell or integration of the transgene into a

chromosome of a target cell by way of human intervention, such as by the methods described herein.

The term"transgene"as used herein refers to any nucleic acid sequence which is introduced into the genome of a cell by experimental manipulations. A transgene may be an "endogenous DNA sequence, "or a"heterologous DNA sequence" (i. e. , "foreign DNA"). The term"endogenous DNA sequence"refers to a nucleotide sequence which is naturally found in the cell into which it is introduced so long as it does not contain some modification (e. g., a point mutation, the presence of a selectable marker gene, etc.) relative to the naturally-occurring sequence. The term"heterologous DNA sequence"refers to a nucleotide sequence which is ligated to, or is manipulated to become ligated to, a nucleic acid sequence to which it is not ligated in nature, or to which it is ligated at a different location in nature. Heterologous DNA is not endogenous to the cell into which it is introduced, but has been obtained from another cell.

Heterologous DNA also includes an endogenous DNA sequence which contains some modification. Generally, although not necessarily, heterologous DNA encodes RNA and proteins that are not normally produced by the cell into which it is expressed. Examples of heterologous DNA include reporter genes, transcriptional and translational regulatory sequences, selectable marker proteins (e. g., proteins which confer drug resistance), etc.

The term"foreign gene"refers to any nucleic acid (e. g., gene sequence) which is introduced into the genome of a cell by experimental manipulations and may include gene sequences found in that cell so long as the introduced gene contains some modification (e. g., a point mutation, the presence of a selectable marker gene, etc.) relative to the naturally-occurring gene.

Transformation of a cell may be stable or transient. The term"transient transformation" or"transiently transformed !' refers to the introduction of one or more transgenes into a cell in the absence of integration of the transgene into the host cell's genome. Transient transformation may be detected by, for example, enzyme-linked immunosorbent assay (ELISA) which detects the presence of a polypeptide encoded by one or more of the transgenes. Alternatively, transient transformation may be detected by detecting the activity of the protein (e. g"B-glucuronidase) encoded by the transgene. The term"transient transformant"refers to a cell which has transiently incorporated one or more transgenes. In contrast, the term"stable transformation"or

"stably transformed"refers to the introduction and integration of one or more transgenes into the genome of a cell. Stable transformation of a cell may be detected by Southern blot hybridization of genomic DNA of the cell with nucleic acid sequences which are capable of binding to one or more of the transgenes. Alternatively, stable transformation of a cell may also be detected by the polymerase chain reaction of genomic DNA of the cell to amplify transgene sequences. The term"stable transformant"refers to a cell which has stably integrated one or more transgenes into the genomic DNA. Thus, a stable transformant is distinguished from a transient transformant in that, whereas genomic DNA from the stable transformant contains one or more transgenes, genomic DNA from the transient transformant does not contain a transgene.

The term"amplification"is defined as the production of additional copies of a nucleic acid sequence and is generally carried out using polymerase chain reaction technologies well known in the art (Dieffenbach and GS Dvekler, PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview NY [1995]). As used herein, the term"polymerase chain reaction" ("PCR") refers to the methods disclosed in U. S. Patent Nos. 4,683, 195,4, 683, 202 and 4,965, 188, all of which are incorporated herein by reference, which describe a method for increasing the concentration of a segment of a target sequence in a mixture of genomic DNA without cloning or purification. This process for amplifying the target sequence consists of introducing a large excess of two oligonucleotide primers to the DNA mixture containing the desired target sequence, followed by a precise sequence of thermal cycling in the presence of a DNA polymerase. The two primers are complementary to their respective strands of the double stranded target sequence. To effect amplification, the mixture is denatured and the primers then annealed to their complementary sequences within the target molecule. Following annealing, the primers are extended with a polymerase so as to form a new pair of complementary strands.

The steps of denaturation, primer annealing and polymerase extension can be repeated many times (i. e., denaturation, annealing and extension constitute one"cycle" ; there can be numerous "cycles") to obtain a high concentration of an amplified segment of the desired target sequence.

The length of the amplified segment of the desired target sequence is determined by the relative positions of the primers with respect to each other, and therefore, this length is a controllable parameter. By virtue of the repeating aspect of the process, the method is referred to as the

"polymerase chain reaction" (hereinafter"PCR"). Because the desired amplified segments of the target sequence become the predominant sequences (in terms of concentration) in the mixture, they are said to be"PCR amplified." With PCR, it is possible to amplify a single copy of a specific target sequence in genomic DNA to a level detectable by several different methodologies (e. g., hybridization with a labeled probe; incorporation of biotinylated primers followed by avidin-enzyme conjugate detection; and/or incorporation of 32P-labeled deoxyribonucleotide triphosphates, such as dCTP or dATP, into the amplified segment). In addition to genomic DNA, any oligonucleotide sequence can be amplified with the appropriate set of primer molecules. In particular, the amplified segments created by the PCR process itself are, themselves, efficient templates for subsequent PCR amplifications. Amplified target sequences may be used to obtain segments of DNA (e. g. , genes) for the construction of targeting vectors, transgenes, etc.

As used herein, the term"sample template"refers to a nucleic acid originating from a sample which is analyzed for the presence of"target". In contrast,"background template"is used in reference to nucleic acid other than sample template, which may or may not be present in a sample. Background template is most often inadvertent. It may be the result of carryover, or it may be due to the presence of nucleic acid contaminants sought to be purified away from the sample. For example, nucleic acids other than those to be detected may be present as background in a test sample.

As used herein, the term"primer"refers to an oligonucleotide, whether occurring naturally (e. g., as in a purified restriction digest) or produced synthetically, which is capable of acting as a point of initiation of nucleic acid synthesis when placed under conditions in which synthesis of a primer extension product which is complementary to a nucleic acid strand is induced (i. e. , in the presence of nucleotides, an inducing agent such as DNA polymerase, and under suitable conditions of temperature and pH). The primer is preferably single-stranded for maximum efficiency in amplification, but may alternatively be double-stranded. If double- stranded, the primer is first treated to separate its strands before being used to prepare extension products. Preferably, the primer is an oligodeoxyribonucleotide. The primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing

agent. The exact lengths of the primers will depend on many factors, including temperature, source of primer and use of the method.

As used herein, the term"probe"refers to a polynucleotide sequence (for example an oligonucleotide), whether occurring naturally (e. g., as in a purified restriction digest) or produced synthetically, recombinantly or by PCR amplification, which is capable of hybridizing to another nucleic acid sequence of interest. A probe may be single-stranded or double- stranded. Probes are useful in the detection, identification and isolation of particular gene sequences. It is contemplated that the probe used in the present invention is labeled with any "reporter molecule, "so that it is detectable in a detection system, including, but not limited to enzyme (i. e. , ELISA, as well as enzyme-based histochemical assays), fluorescent, radioactive, and luminescent systems. It is not intended that the present invention be limited to any particular detection system or label. The terms"reporter molecule"and"label"are used herein interchangeably. In addition to probes, primers and deoxynucleoside triphosphates may contain labels ; these labels may comprise, but are not limited to, 32p, 33p, 35S, enzymes, or fluorescent molecules (e. g. , fluorescent dyes).

As used herein, "FISH"refers to fluorescence in situ hybridization, a technique in which detectably labeled DNA probes (which can be prepared, for example, from cDNA sequences or genomic sequences contained in cosmids or bacterial artificial chromosomes (BACs) ) are hybridized to cytogenetic or histological specimens. Such specimens include, but are not limited to, metaphase chromosome spreads and interphase nuclei prepared from tissue or blood specimens, and formaldehyde-fixed, paraffin-embedded tissue sections. Fluorescent labels can be directly incorporated into the probe, or can be applied as antibody-label conjugates which bind to affinity labels (for example, biotin or digoxigenin) incorporated into the probe, either directly, or as an antibody"sandwich" (i. e. a primary and a secondary antibody). The fluorescent dyes include, but are not limited to rhodamine, texas red, FITC (fluorescein isothiocyanate) and TRITC (tetramethyl rhodamine isothiocyanate). The fluorescent labels are detected using a fluorescence microscope equipped with a mercury or xenon lamp (as an illumination source) and appropriate filters for excitation and emission. The pattern of fluorescence can be used to assess copy number of the locus recognized by the probe, or, in cases where two or more (differentially) labeled probes are used, to assess the relative positions

of the probes (for example to detect chromosomal rearrangements, such as translocations and inversions).

As used herein, "parathyroid tumors"refers to tumors (benign or malignant) of the parathyroid gland. By way of example, a parathyroid adenoma is a benign tumor of the parathyroid gland. Other parathyroid tumors include parathyroid carcinomas.

As used herein,"parathyroid adenoma"refers to a benign tumor of a parathyroid gland.

This enlargment and overactivity of the affected gland results in continued production of parathyroid hormone (hyperparathyroidism). In turn, elevated levels of calcium are found in the blood.

As used herein, the term"regulatory region of a gene expressed in the parathyroid" refers to the upstream regulatory sequences (including but not limited to promoter sequences) of a gene that is expressed primarily (if not exclusively) in the parathyroid gland. Such genes (and their regulatory regions) include, but are not limited to the parathyroid hormone gene and the Gcm2 gene. Such regulatory regions, when placed in operable combination with a sequence of interest will permit expression of the sequence of interest in a tissue-specific fashion in the parathyroid.

As used herein,"POU domain transcription factor"refers to a member of a family of transcription factors, characterized by possession of a bipartite DNA binding domain (the POU domain) that also provides binding sites for many proteins that interact with POU domains.

POU domain transcription factors include, but are not limited to the transcription factor Oct-6 (also known as Tst-1 or SCIP).

As used herein, a"STK4/Mst-l/Krs-2-tramd L fusion"or"STK4/Mst-l/Krs-2- tramdorin L fusion"refers to a fusion between tramdorin L sequences on human chromosome 5 and STK4/Mst-l/Krs-2 sequences on human chromosome 20. The fusion may be at the level of genomic sequences, or at the level of cDNA sequences, or both. The fusion involves at least the known exon of tramdL, as shown in Figure 12B, and at least the genomic DNA encoding exons 6-9 of STK4/Mst-l/Krs-2, as shown in Figure 12A. STK4/Mst-l/Krs-2 is a member of the STE20 family of kinases. The STK4/Mst-l/Krs-2-tramd L fusion, as represented in EST cDNA 1388139 (SEQ ID NO : 23), comprises the STK4/Mst-l/Krs-2 kinase domain sequences fused to 3'sequences of tramd L. A sequence of a representative STK4/Mst-l/Krs-2-tramdL fusion

junction (in a cDNA) is shown in Figure 12C (SEQ ID NO : 31). The genomic sequences from the STK4/Mst-l/Krs-2 and tramdL partners of a representative fusion are shown in Figures 12A (SEQ ID NO : 27) and 12B (SEQ ID NO : 29), respectively. A predicted cDNA (SEQ ID NO : 38) and protein sequence (SEQ ID NO : 44) of a fusion containing additional 5'STK4 sequences not contained in EST cDNA 1388139 is provided in Figure 26.

As used herein,"NGFR"refers to a nerve growth factor receptor gene.

As used herein, "GADPH"refers to a glyceraldehydes-3-phosphate dehydrogenase gene.

As used herein, "Sos" (an acronym for"son-of-sevenless") refers to a ras guanine nucleotide exchange factor.

As used herein,"GABA"refers to y-aminobutyric acid.

As used herein, "Gcm2"refers to the glial cells missing protein homolog (Gcm2) gene.

As used herein, the term"syntenic"refers to the preservation of"blocks"of genes in some fashion across species. That is, syntenic refers to a region of conserved synteny with another species, wherein"synteny"refers to the physical presence together on the same chromosome of two or more gene loci, whether or not they are close enough together for linkage to be demonstrated.

As used herein, the term"incisures" (also referred to as"Schmidt-Lanterman incisures"), are regions of non-compact myelin (i. e. regions of the myelin sheath that contain cytoplasm).

As used herein, the term"paranode"refers to a region adjacent to the node of Ranvier (i. e. the junction between adjacent myelin sheathes). The paranode contains a set of proteins that are not found in the node itself, or in the internode, the myelinated region between nodes.

A single Schwann cell will contain half a node of Ranvier, a paranode, an internode, another paranode, and another half a node of Ranvier.

As used herein, "KLH"refers to the protein called Keyhole Limpet Hemocyanin.

DETAILED DESCRIPTION OF THE INVENTION Mouse tramdorin 1 was identified in a screen for target genes of the POU domain transcription factor, Oct-6. Tramdl encodes a putative transmembrane domain protein, and the

expression pattern of tramdl suggests that it is related to myelination in the peripheral nervous system. Homology searching identified tramdorin 1 homologues in human and rat, as well as identified a family of closely-related tramdorin genes, with at least four members. The location of the tramdorin gene cluster in humans is suggestive of a potential role in a peripheral neuropathy. One of the family members is present as a gene fragment, and may play a role in the etiology of the parathyroid tumors which is, in one embodiment, an adenoma.

Compositions comprising the tramdorins (nucleic acid and amino acid sequences) and host cells transfected with tramdorins are contemplated. Additionally, the use of tramdorins in a variety of diagnostic assays is contemplated, as is the development of a transgenic mouse model for parathyroid tumors. The description of the invention includes I. The initial identification and characterization of mouse tramdorin 1, II. Identification of tramdorin homologs in mouse and other species, III. Organization of the tramdorin family locus in mouse and human, IV.

Functional characterization of tramdorins V. Tramdorins and parathyroid tumors and VI.

Tramdorins and peripheral neuropathies. VII. Remyelination by Schwann cells in the CNS following disease or injury.

I. THE INITIAL CHARACTERIZATION OF MOUSE TRAMDORIN 1 Tramdorin 1 was identified in experiments directed to the study of myelination in the peripheral nervous system. Myelin sheathes are membranous structures that surround large caliber axons. By limiting axonal depolarization to the nodes of Ranvier that separate individual sheaths, myelin permits rapid axonal conductance over long distances with small axonal volumes, a property that is essential for normal functioning of vertebrate nervous systems. The debilitating effects of diseases that affect myelin, such as Charcot Marie Tooth Syndrome and multiple sclerosis, demonstrate its importance. Myelin is produced by Schwann cells in the PNS and by oligodendrocytes in the CNS. Both cell types ensheathe axons, then wrap them with multiple layers of membrane that, following exclusion of cytoplasm and compaction, constitute the myelin sheathes. However, the molecular mechanisms that control the synthesis of myelin are poorly understood.

Schwann cells are the primary glial cells of the peripheral nervous system. Myelinating Schwann cells form a single myelin sheath around a single axon, whereas non-myelinating

Schwann cells typically ensheathe several unmyelinated axons. Axons regulate the proliferation of Schwann cells, and in addition, they are thought to regulate their differentiation: presumptive myelinated axons signal Schwann cells into the myelinating lineage, whereas the axons lacking this signal remain associated with non-myelinating Schwann cells. Intracellular cAMP signaling is thought to play a central role in axonal control of both Schwann cell proliferation and differentiation, although the nature of the signal (s) that induce elevation of the levels of cyclic AMP (cAMP) in Schwann cells remain unknown. In cultured Schwann cells, many aspects of axon-induced Schwann cell proliferation and differentiation can be mimicked by activators of adenylyl cyclase, such as forskolin. Forskolin treatment decreases expression of genes that are associated with non-myelinating Schwann cells and immature Schwann cells, and increases expression of genes associated with myelinating Schwann cells, including the POU domain transcription factor Oct-6 and the zinc-finger transcription factor Krox-20. These data indicate that cAMP may be a second messenger of the axonal signal that promotes myelination, and that the change to the myelinating phenotype is mediated by changes in the expression of several transcription factors. Both Oct-6 and Krox-20 play essential roles in the development of myelinating Schwann cells [Bermingham et al., Gene Develop 10: 1751 (1996) ]. However, the precise role played by cAMP signaling in Schwann cell myelination is not yet fully understood.

Oct-6 (also known as Tst-1 or SCIP) is a member of the POU domain family of transcription factors. These proteins are characterized by their possession of a bipartite DNA binding domain (the POU domain) that also provides binding sites for many proteins that interact with POU proteins. Oct-6 is expressed transiently during Schwann cell development [Monuki et al., Neuron 3: 783 (1989)], specifically in promyelinating Schwann cells, which have formed a 1: 1 relation with axons but have not yet formed a myelin sheath. Oct-6 represses expression of several myelin-related promoters in transient cotransfection assays, and on this basis, has been hypothesized to repress myelination. It is also expressed transiently in oligodendrocytes [Collarini et al., Development 116: 193 (1992) ]. The phenotypes of mutations in Oct-6 indicate that it can control the timing of myelination. In the absence of Oct-6, Schwann cell differentiation is delayed, transiently arrested at the promyelinating stage [Bermingham et al., Gene Develop 10: 1751 (1996) and Jaego, et al., Science 273: 507 (1996)].

CNS myelination appears normal, perhaps due to the presence in oligodendrocytes, but not

Schwann cells, of two other members of the class m POU domain family, Bm-1 and Brn-2 [Schreiber et al., JBiol Chem 272: 32286 (1997) ]. Transgenic mice that express a truncated form of Oct-6 in Schwann cells display precocious peripheral myelination [Weinstein et al., Mol Cell Neurosci 6 : 212 (1995) ], and Oct-6 misexpression in oligodendrocytes causes precocious CNS myelination and hypomyelination [Jensen et al., J Clin Invest 101 : 1292 (1998) ]. Oct-6 has been hypothesized to activate genes required for Schwann cell differentiation, then to repress terminal differentiation.

The identification of target genes that are regulated by Oct-6 would enhance our understanding of its role in Schwann cell development. Recently, representational difference analysis (RDA; [Lisitsyn, Trens Genet 11: 303 (1995) ], a PCR-based technique that permits the isolation of DNA fragments that are present in one DNA sample but absent in another, was used to identify genes that are differentially expressed in sciatic nerve from Oct-6 homozygous mutant (-/-) and wild-type (+/+) mice. Six cDNA fragments were identified that correspond to genes that are activated by Oct-6; no repressed genes were identified, suggesting that Oct-6 functions as an activator in Schwann cells [Bermingham et al., manuscript in preparation]. Of these genes, only one has been described previously in Schwann cells, and three are novel. The identities of two known putative target genes suggest cytoskeletal rearrangement or fatty acid transport as rate-limiting steps in myelination that are activated by Oct-6. One of the previously unknown genes named Tramdorin 1 (transmembrane domain rich protein). Tramdorin 1 has been assigned the human and mouse gene symbol tramdl.

RDA clone JBSN125 contained tramdorinl sequences that were differentially expressed between Oct-6 (-/-) and wild-type (+/+) sciatic nerve, as well as a sequence whose expression was unaffected by the absence of Oct-6. A subclone of this, 125-10, contained only differentially expressed sequences and was used to isolate additional sequence for the tramdorinl gene.

Mouse cDNAs that correspond to clone 125-10 were isolated by obtaining and sequencing three homologous EST clones. Mouse EST cDNA 1920302 defines a 2.5 kb cDNA (SEQ ID NO : 13/GenBank Acession No.: AI 1780664). One of the EST clones appears to correspond to a partially spliced transcript. The full-length mouse cDNA contains a 1.4 kb open reading frame (Figure 2) that encodes a protein of 52kd. The mouse cDNA and the

putative protein that it encodes do not match any genes in the GenBank non-redundant database, or the SwissProt database. Therefore, these cDNAs are derived from a novel gene.

That is to say, there are no exact matches to the protein sequence.

The protein encoded by mouse cDNA 1920302 (SEQ ID NO : 13) was predicted by several protein structure prediction programs to encode a transmembrane protein (see examples below). Regardless of the differing predictions about the number of transmembrane domains, or their topology, this protein appears to contain multiple transmembrane helices.

To determine the size of tramdorin mRNA (s) and to determine if tramdorin expression is restricted to sciatic nerve, a northern blot with RNAs from adult mouse sciatic nerve and other tissues was hybridized to a radiolabelled probe synthesized from tramdorin cDNA 1920302 (SEQ ID NO : 13). Tramdorinl expression is neither ubiquitous, nor restricted to sciatic nerve (see examples below).

It was of interest to determine whether the expression of tramdl in sciatic nerve is modulated by nerve injury. A northern blot of RNA from sciatic nerves collected at various times following crush or transection injury was hybridized with a mouse tramdl probe (see examples below). Tramdl expression is downregulated by transection injury, and remains low.

Following a crush injury, tramdl is downregulated, but subsequently it is reexpressed as axons regenerate and are remyelinated. Therefore, the tramdorin gene encodes a myelin-related protein. Several genes that are required for normal myelination show this pattern of expression; these include Oct-6, PO, connexin32 and PMP-22 [Podsulo, Regulation of myelin gene expression in the peripheral nervous system. In Dyck, P. J. et al. (eds.), Peripheral Neuropathy, W. B. Saunders Co. , Philadelphia, [1993] pp. 282-289 ; Scherer et al., JNeurosci 14: 1930 (1994); Sohl et al., Eur J Cell biol 69: 267 (1996) ]. This result suggests that tramdl may play an important role in the timely formation of peripheral myelin. Thus, an important function of Oct-6 in myelination may be activating the expression of tramdorin 1, a gene that encodes a novel transmembrane domain protein. Additionally, the expression of tramdl following nerve injury indicates that it will be useful as a marker of myelinating Schwann cells. Following crush injuries, Schwann cells distal to the injury downregulate expression of myelin-related genes such as PO, and express markers of immature Schwann cells, such as p75 nerve growth factor [Poduslo, "Regulation of myelin gene expression in the peripheral nervous system. "In Dyck,

P. J. et al. (eds), Peripheral Neuropathy, W. B. Saunders Co. , Philadelphia, pp. 282-289 (1993)].

As axons regenerate, they are remyelinated, with concomitant re-expression of myelin-related genes. Tramdorinl shows this pattern of expression. Therefore the present invention contemplates using tramdorin 1 to augment current markers of myelinating Schwann cells.

Remyelination of axons in the CNS by Schwann cells has been explored extensively as a possible treatment for demyelinating diseases such as multiple sclerosis, and as a treatment for nerve injury. MS affects 1/1000 Americans, and in people of northern European descent, the frequency is 1/800 [Lynch et al., Dis Mon 42 : 1 (1996) ] ; it represents a significant public health problem in the United States and Europe. An understanding of the genes that regulate myelination by Schwann cells is needed for optimizing their use in remyelination in the CNS.

While it is not necessary to understand the underlying mechanisms to practice the invention (and the invention is not limited to any particular mechanism), it is believed that the tramdorin 1 gene may play a crucial role in regulating myelination for the following reasons: First, it is a putative target gene for Oct-6, a transcription factor that is essential for timely peripheral myelination [Bermingham et al., Gene Develop 10: 1751 (1996); Jaegle et al., Science 273: 507 (1996) ]. Second, its expression following nerve injury indicates that it encodes a myelin-related protein. Screens using tramdorin, and/or its putative ligands, will provide avenues for the development of therapeutic agents for the treatment of the effects of demyelination or nerve injury, both in the peripheral and central nervous systems. The ability to identify molecules that promote myelination will significantly enhance the prospects for successfully using Schwann cells, and perhaps oligodendrocytes and olfactory ensheathing cells as well, to mitigate the damage caused by MS and other demyelinating diseases. A significant use of tramdorin 1 will be in identifying such molecules (see section IV, "Functional Characterization of Tramdorins").

The mouse chromosomal location of Tramdl was determined by interspecific backcross analysis using progeny derived from matings of [ (C57BL/6J x Mus spretus) Fl X C57BL/6J] mice (see examples below). The mapping results indicated that Tramdl is located in the proximal region of mouse chromosome 11 linked to 7773 and Handl. The most likely gene order is: cenkomere-m3-1/142-Tramdl-6/179-Handl. The recombination frequencies (expressed as genetic distances in centiMorgans (cM) the standard error) are-Ill3-0. 7 +/- 0. 7-Tramdl-3. 4 +/-1. 4-Handl. The proximal region of mouse chromosome 11 is syntenic

with human chromosome 5q31-33. An autosomal recessive Charcot-Marie-Tooth Syndrome demyelinating neuropathy has been mapped to this region [Guilbot et al., Ann NYAcad Sci 883: 56 (1999a); Guilbot et al., Eur JHum Genet 7: 849 (1999b); LeGuern et al., Hum Mol Genet 5: 1685 (1996) ], suggesting that tramdorinl could be a candidate peripheral neuropathy disease gene.

II. IDENTIFICATION OF TRAMDORIN HOMOLOGS IN RAT AND OTHER SPECIES A. Rat A Rat cDNA that corresponds to mouse clone 125-10 was isolated by 5'and 3'RACE (rapid isolation of cDNA ends; [Frohman, Methods Enzymol 218 : 340 (1993); Frohman, PCR Methods Appl 4 : S40 (1994) ] ). The 5'and 3'rat RACE fragments combined define a 2.5 kb (SEQ ID NO : 55) cDNA. The full-length rat cDNA (SEQ ID NO : 6) contains a 1.4 kb open reading frame (Figure 2) that encodes a protein of 52 kd (SEQ ID NO : 22). The rat cDNA and the putative protein that it encodes (Figures 2 and 15) does not match any genes in the GenBank non-redundant database, or the SwissProt database. Therefore, this cDNA is derived from a novel gene.

A Northern blot using rat tissue did not show hybridization to lung and thymus (data not shown), suggesting that there may be variation in tramdorin 1 expression among species, or cross-hybridization to related sequences in the mouse Northern blots.

B. Human As noted above, the proximal region of mouse chromosome 11 is syntenic with human chromosome 5q31-33, suggesting that the human homolog of tramdl will map to 5q31-33 as well. An autosomal recessive Charcot-Marie-Tooth Syndrome demyelinating neuropathy has been mapped to this region, suggesting that tramdorin 1 could be a candidate peripheral neuropathy disease gene.

As a first step toward determining if tramdorinl mutations are associated with human peripheral neuropathy, the human tramdorin gene was studied. Six human EST cDNAs that possess homology to mouse tramdorinl, I. M. A. G. E numbers 3184556 (BE501426), 1738130

(AI140615), 1837427 (AI208756), 1388139 (AA843982), and 2549054 (AI953890), as well as EST cDNA DKFZp434G1123 from the Deutsches Ressourcezentrum für Genomforschung GmbH (RZPD) were obtained and sequenced in their entirety. The sequences were aligned on human genomic DNA from the International Human Genome Sequencing Consortium (IHGSC) and Celera databases. These cDNAs showed greatest homology to four discrete locations within 250 kb of chromosome 5q sequence (Figure 7), confirming the mapping of tramdorin in mouse, but in addition, suggesting the existence of multiple tramdorin genes (see examples below).

These genes have been assigned the names tramdl (SEQ ID NO : 9), tramd2 (SEQ ID NO : 10), tramd3 (SEQ ID NO : 11), and tramdL (SEQ ID NO : 12). The predicted cDNA and encoded amino acid sequences of human tramdl (cDNA : SEQ ID NO : 3; corresponding amino acid sequence: SEQ ID NO: 19), tramd2 (cDNA : SEQ ID NO : 4; corresponding amino acid sequence: SEQ ID NO: 20) and tramd3 (cDNA : SEQ ID NO : 5 ; corresponding amino acid sequence: SEQ ID NO: 21) are shown in Figures 10,11, and 21, respectively. The amino acid sequence of human tramd3 (SEQ ID NO : 21) is also depicted in Figure 20 (aligned with mouse tramd3), and the amino acid sequences of human tramd 1 (SEQ ID NO : 19), tramd 2 (SEQ ID NO : 20) and tramd 3 (SEQ ID NO : 21) are also depicted in Figures 14,15, 16 (tramdl), Figure 14 (tramd 2) and Figures 14,16, 20 (tramd 3).

A fourth tramdorin-related sequence, tramdl, is defined by the EST cDNA 1388139 (SEQ ID NO : 23) (Figures 8D and 12). tramdL contains only sequences that are homologous to exon 10 of tramdorinl. This cDNA is derived from a parathyroid adenoma, and is unusual in that its 5'end consists of sequences derived from a member of the STE20 family of kinases, STK4/Mst-l/Krs-2 (Figure 12A; [Creasy et al., JBiol Chem 270: 21695 (1995); Taylor et al., PNAS USA 93 : 10099 (1996) ]), located on human chromosome 20. The STK4/Mst-l/Krs-2 cDNA fragment is fused to 3'sequences of tramdL (Figure 12B), located on chromosome 5.

This observation raises the possibility that tramdL resides near a parathyroid tumor translocation breakpoint, between chromosomes 20 and 5, that results in expression of a STK4/Mst-l/Krs-2-tramdorinL fusion protein. Such a fusion protein may be relevant in the etiology of parathyroid tumors, such as parathyroid adenomas, and several applications involving the use of this fusion in the evaluation of parathyroid tumors, including but not limited to parathyroid adenomas, and in the development of an animal model of parathyroid

tumors, including but not limited to parathyroid adenomas, are contemplated (see Section V, Tramdorins and Parathyroid Tumors and Examples below).

To determine the tissues in which the various tramdorin genes are expressed in humans, a northern blot of RNA derived from 12 human tissues was sequentially hybridized to radiolabelled cDNA probes that correspond to human tramdl, 2,3 and tramdL that were constructed as follows: htramdl probe: Probe consists of 355bp EcoRI-PvuII fragment of EST cDNA 3184556. The Eco RI site resides in the vector that contains the cDNA ; the probe contains human tramdl exons 7 and 8, and part of the intron between exons 8 and 9, ending at the PvuII site at position 248018 in figure 9. htramd2 probe: Probe consists of 440bp EcoRI-NcoI fragment of EST cDNA 1837427. The Eco RI site resides in the vector that contains the cDNA ; the probe contains sequences from position 1209 to the Nco site at position 1636 in Figure 11, and contains part of human tramd2 exon 9, exon 10, and part of exon 11. htramd3 probe: Probe consists of 850bp StuI-KpnI fragment of EST cDNA 2549054, consisting of the sequences between the StuI site at position 232 and the Kpn site at position 1080 on Figure 21. htramdL probe: Probe consists of 340bp MscI-DraI fragment of EST cDNA 1388139, consisting of the sequences between the MscI site 7bp 3'to the 3'splice site of tramdL and the DraI site at the 5'end of the polyA tail of this cDNA, which is marked by an asterisk in Figure 12B.

Each probe produced a distinct hybridization result (see example below). Translation of the known putative coding exons for human tramdorins 1, 2, and 3 indicate that these proteins are highly conserved (see example below).

C. Tramdorins in Other Organisms The most closely related sequences to tramdorins are putative proteins that have been derived from the Drosophila, Caenorhabditis elegans and Saccaromyces cerevisiae genome sequences (see example below). Mouse tramdl protein is 39% identical to Drosophila protein CG13384, and 32% identical to the T27A1. 5 protein from C. elegans. The functions of these

proteins in invertebrates may be related to tramdorin function in vertebrates. If so, tramdorins would be required for a function that is conserved by evolution.

Seven novel yeast amino acid transporter genes, Avtl-7, were identified by virtue of their homology to vesicular GABA transporter (VGAT) sequences (Russnak et al., JBiol Chem 276 : 23849 (2001)). VGAT/VIAAT/Unc-47 proteins are the known proteins that are most related to tramdorin/LYAAT proteins (Figure 16). A CLUSTAL W comparison of human and mouse tramdl, human tramd2, human tramd3 and AVT1-4 and AVT7 proteins demonstrates that the tramdorins are most closely related to AVT3 and AVT4 (data not shown).

Most of the AVT proteins, including AVT3 and AVT4, were localized to vacuoles, a yeast organelle that closely resembles mammalian lysosomes, consistent with the localization of tramd3/LYAAT1 to lysosomes (Sagne et al, PNAS USA 98 : 7206 (2001) ). AVT3,4, and 6 appear to be involved in amino acid export from vacuoles, suggesting that tramdorins could perform a similar function in lysosomes. However, the observation that not all AVT proteins appear to be localized exclusively in vacuoles suggests that other mammalian homologs of these proteins may exist in different subcellular locations.

D. Structure of Tramd Proteins While an understanding of the precise structure of tramdorins is not necessary to the practice of the present invention, a number of considerations suggest that tramd proteins consist of 11 transmembrane domains, with the N-terminus facing the cytoplasm (see example below).

The vertebrate proteins to which the tramdorins are most closely related are vesicular y- aminobutyric acid (GABA) transporters (VGATs ; see example below; [McIntire et al., Nature 389 : 870 (1997); Sagne et al., FEBSLett 417: 177 (1997)]).

III. ORGANIZATION OF THE TRAMDORIN LOCUS IN HUMAN AND MOUSE A. Human As noted above, human EST cDNAs that possess homology to mouse tramdorin 1 were aligned on human genomic DNA from the International Human Genome Sequencing Consortium (IHGSC) Homo sapiens chromosome 5 working draft sequence segment

NT006951. 4 and Celera databases [Lander et al., Nature 409: 860 (2001); Venter et al., Science 291: 1304 (2001)]. These cDNAs showed greatest homology to four discrete locations within 250 kb of chromosome 5q sequence (Figure 7), confirming the mapping of tramdorin in mouse, but in addition, suggesting the existence of multiple tramdorin genes. These genes have been assigned the names tramdl, tramd2, tramd3, and tramdL. The human tramdl (SEQ ID NO : 9) gene has greatest homology to the original mouse tramdl (SEQ ID NO : 7) gene, and is flanked on its 3'end by tramd2 (SEQ ID NO : 10), and on its 5'end by tramd3 (SEQ ID NO : 11), which is transcribed in the opposite orientation to tramdl and tramd2. Nested between tramdl and tramd3 resides a tramdorin gene fragment, tramdL (SEQ ID NO : 12), in the same orientation as tramdl and tramd2. It consists only of 3'tramdorin sequences, and is defined by a single EST cDNA, 1388139 (SEQ ID NO : 23). All four putative genes are found on both the Celera and IHGSC sequences. Figure 8 presents the organization of the human tramdorin loci (see Example below for details of the organization). The cDNAs in Figure 8 are aligned with Celera human chromosome 5 scaffold sequence GA x2HTBL3TT27: 2500000-3000000, except where noted in the figure. The genomic sequence of 250 kb of sequence from the entire tramdorin region (nucleotides 50001-300000 of the IHGSC Homo Sapiens chromosome 5 working draft sequence segment NT-006951. 4) is presented in Figure 9. Known tramdorin exons are shown in bold type, and indicated to the right of the sequence. Initiation ATG and stop codons, and putative polyadenylation signal sites are shown in bold italic. Note that tramd3 is in the opposite orientation relative to the other tramd genes, and is shown in the antisense orientation.

Details of the structure of the individual tramdorin genes are presented in the example below. Briefly, the structure of human tramdorin 1 (SEQ ID NO : 9) was determined by identifying regions of homology between mouse and human tramdorins, and by cDNA sequences. Figure 10 depicts the putative human tramd 1 cDNA (SEQ ID NO : 3) and encoded protein (SEQ ID NO : 19). Human tramdorin 2 (SEQ ID NO : 4) exons are defined by a single EST cDNA and by homology with tramdorins 1 (SEQ ID NO : 3) and 3 (SEQ ID NO : 5), and mouse tramd2 (SEQ ID NO : 1) cDNA. Figure 11 depicts the putative cDNA (SEQ ID NO : 3) and encoded protein (SEQ ID NO : 20) for human tramd 2. The sequences of four alternatively spliced tramdorin 3 EST cDNAs were used to infer the structure of the tramdorin 3 gene. Figure

21 depicts a composite human tramd3 cDNA (SEQ ID NO : 5) and corresponding amino acid sequence (SEQ ID NO : 21). A fourth tramdorin-related sequence, tramdL (SEQ ID NO : 12), is defined by an EST cDNA which is derived from a parathyroid adenoma. TramdL contains sequences which are homologous to exon 10 of tramdorin 1. The 5'end of this EST cDNA consists of sequences derived from a member of the STE20 family of kinases, STK4/Mst- l/Krs-2 [Creasy et al., JBiol Chem 270: 21695 (1995); Taylor et al., PNAS USA 93: 10099 (1996) ], located on chromosome 20. The STK4/Mst-l/Krs-2 cDNA fragment is fused to 3' sequences of tramdL, located on chromosome 5. This observation raises the possibility that tramdL resides near a parathyroid tumor translocation breakpoint, between chromosomes 20 and 5, that results in expression ofaSTK4/Mst-l/Krs-2-tramdorinL fusion protein (see Section V, Tramdorins and Parathyroid Tumors below).

B. Mouse The organization of the mouse tramdl gene (SEQ ID NO : 7) was studied to understand better the origin of the alternatively spliced tramdorin cDNAs, and as a preliminary step toward generating mice that lack tramdl function. A 129 mouse genomic library in bacteriophage , was screened with a probe consisting of a 720bp XhoI-NcoI fragment from cDNA 1920302 (SEQ ID NO : 13) that contains sequence from exons 1-6. Three phage were isolated (Figure 17A). In addition, a bacterial artificial chromosome (BAC) library (Research Genetics) was screened using the polymerase chain reaction (PCR) using primers that amplify 169bp of exon 6 (see example below). A single tramdorin-containing BAC was isolated. Subclones from the BAC and from the lambda phage were sequenced to determine the structure of the 5'end of the tramdorin gene. The genomic sequence 3'to exon 7 corresponds to sequence in EST cDNA 1363993 (SEQ ID NO : 43), indicating that this cDNA is partially spliced. Exons 8,9, and 10 are defined by sequences from the Ensembl database. Mouse tramdorin 1 genomic sequences (SEQ ID NO : 7) are presented in Figure 18.

To determine if the BAC contained genomic sequences for additional mouse tramdorin genes, plasmids that contained shotgun-cloned BAC DNA were arrayed on dot blots. Sequence analysis of clones that hybridized to radiolabelled tramdl cDNA probes revealed that in addition to tramdl sequences, the BAC also contained tramdorin 3 sequences (Figure 17B).

Mouse tramdorin 3 genomic sequences (SEQ ID NO : 8) are presented in Figure 19, and a comparison between mouse (SEQ ID NO: 18) and human tramdorin 3 (SEQ ID NO : 21) proteins is presented in Figure 20. Because BACs typically contain approximately 100kb of DNA, the presence of tramdl and tramd3 sequences in the same BAC suggests that these genes may reside more closely to one another in mice than they do in humans.

Composite mouse tramd3 cDNA (SEQ ID NO : 3) is presented in Figure 23. As described in the example below, mouse tramdorin 3 cDNAs were obtained by 5'and 3'RACE amplification from Swiss Webster mouse brain RACE-ready cDNA (Ambion), and by amplification of the same mouse brain cDNA using primers, as detailed in the example below.

The cDNA depicted in Figure 23A contains the entire tramd3 protein coding region, but does not contain the entire 3'untranslated region.

Composite mouse tramdorin 2 cDNA (SEQ ID NO : 1) is presented in Figure 22. As described in the example below, 5'and 3'RLM-RACE (Maruyama et al., Gene 138 : 171 (1994); Schaefer, Anal Biochem 227: 255 (1995) ) were used to isolate mouse tramdorin 2 cDNAs from a mouse testicle cDNA library (Ambion). The absence of a consensus polyadenylation site and the observation that the poly (A) sequences at the 3'end are found in genomic DNA indicate that the 3'RACE cDNA is not full-length.

IV. FUNCTIONAL CHARACTERIZATION OF TRAMDORINS A. Identification of Tramdorin Ligands The signal (s) from axons that initiate myelination are unknown. Several molecules that stimulate Schwann cell myelination have been identified. Insulin-like growth factor promotes Schwann cell proliferation in conjunction with other growth factors, but in their absence, it promotes Schwann cell expression of PO and myelination (Russell et al., JNeuropathol Exp Neurol 59 : 575 (2000) ; Stewart et al., Eur JNeurosci 8: 553 (1996) ). Progesterone and glucocorticoids have been shown to enhance the rate of myelination. However, none of these molecules has been shown to be sufficient for triggering myelination by Schwann cells. Studies on regenerating nerves following injury indicate that axonal contact is required for Schwann cells to myelinate (reviewed in Scherer et al., Axon-Schwann cell interactions in peripheral nerve regeneration. In Jessen, K. R. and Richardson, W. D. (eds. ), Glial Cell Development, Bios

Scientific Publishers, Oxford, [1996] pp. 165-196). However, Schwann cells repressed expression of p75NGFR, and expressed Oct-6 and PO in response to neurons that were separated by a permeable membrane, suggesting that at least one important axonal signal for the differentiation of Schwann cells to a myelinating phenotype is a diffusible molecule (Bolin et al., J Cell Biol 123 : 237 (1993) ). In theory, axons can control the thickness of myelin sheathes that surround them by emanating a single myelinogenic signal which both triggers myelination, and instructs it to continue as long as the signal is present; cessation of the signal terminates myelination (Fraher et al., JAnat 193: 195 (1998) ). Based on its homology to the vesicular GABA transporter, tramdorin protein may function as a transporter of small molecules. If tramdorin is required for proper myelination, it may transduce a signal from axons that promotes myelination, and therefore may be a crucial reagent in the identification of the myelinogenic signal itself. Alternatively, tramdl could interact directly with molecules that are expressed on the surface of axons, or it could act as a cofactor for other Schwann cell signaling molecule (s). Tramd2 and tramd3 genes may function similarly in the differentiation of other cell types as well.

One of skill in the art will understand that there are many methods that can be used to screen for tramdorin ligands. The present invention contemplates that any such assay may be used. By way of non-limiting example, one such assay involves identification of tramdorin ligands by the differential import of radiolabelled candidate ligands following transient expression of tramdorins in cultured cells. COS-7 cells will be transiently transfected with pCDNA3.1 plasmids (Invitrogen) with or without tramdorin sequences. Following exposure to [3H] amino acids, the differential uptake of the radiolabelled amino acids will be measured by scintillation counting of cell extracts, as performed previously for rat LYAAT-1 [Sagne et al., PNAS USA 98 : 7206 (2001) ]. Briefly, tramdorin cDNA sequences will be cloned into pcDNA3. 1 plasmids, which will be introduced into COS-7 cells with Lipofectin (Life Technologies, Grand Island, NY). Transport assays will be carried out 36-48 hours after the beginning of transfection. Following exposure to [3H] amino acids, cells are lysed in 0.1 N NaOH, and the radioactivity will be measured by scintillation counting in Aquasol (Packard) (see Example below).

As rat LYAAT-1 is a tramdorin 3 homolog, and Sagne et al. (supra) used the COS-7 transfection and transport assay to successfully identify the ligand for LYAAT-1, there is compelling evidence that such an assay will be successful at identifying the ligands of human and mouse tramdorins, as well as rat tramdorin 1.

Another assay contemplated for the identification of tramdorin ligands is the expression of tramdorin in Xenopus oocytes. Xenopus oocytes have been used extensively to characterize transport proteins (reviewed in (Miller et al., Biochim BiopZlys Acta 1465 : 343 (2000); Theodoulou et al., Methods Mol Biol 49 : 317 (1995); Wagner et al., Cell Physiol Biochem 10: 1 (2000) ). In the Xenopus oocyte expression system, oocytes removed from ovaries of adult Xenopus are arrested at the first meiotic prophase. Because each oocyte is fairly large and has a large nucleus, it is technically easy to inject foreign DNA into the oocyte nucleus or mRNA injected into the cytoplasm. In this case, a synthetic tramdorin message is made that possesses a 5'cap and a poly (A) + tail, and in which the tramdorin coding sequences are flanked by Xenopus P-globin 5'and 3'untranslated regions. The synthetic message is microinjected into the cytoplasm, followed several days later by characterization of the expressed protein.

Alternatively, tramdorin genes can be microinjected directly into the nuclei of Xenopus oocytes, following cloning into the expression plasmid pMT3 (Swick et al., PNAS USA SA MM 1812 (1992) ). Tramdorin proteins will be characterized by measurement of the rate and extent of uptake of radiolabelled candidate ligands relative to mock injected oocytes, as described above in the COS-7 assay. The large size of the oocytes makes it relatively easy to insert electrodes into the oocytes in order to measure the electrophysiological responses (i. e. patch clamp electrophysiology). The two-electrode voltage clamp can be used to measure transport activity in the presence of ligand (s).

B. Effect of Overexpression of Tramdorin Proteins on Myelination by Schwann Cells Primary Schwann cells will form myelin when co cultured with neurons; such co cultures can form the basis for an assay of tramdorin function during myelination. Achieving myelination by primary Schwann cells cultured with dorsal root ganglion takes two months ; two recently published detailed protocols for this procedure will be relied upon (Kleitman et al.,

Tissue Culture Methods for the Study of Myelination. In Culturing Nerve Cells, Bank et al.

(eds. ) Cambridge, MA : MIT Press [1998] pp. 545-658 ; Li, Methods Cell Biol 57 : 167 (1998)).

Proliferation will be measured by counting Schwann cells. Any effect of tramdorin expression on the rate of myelination will be measured by any of the following four methods: 1) Sudan black staining; 2) myelin basic protein (MBP) or PO immunohistochemistry; 3) thin-layer chromatography to measure the amounts of myelin-specific lipids (Vitiello et al., J Chromatogr 166: 637 (1978) ) ; or 4) fluorescent ceramide incorporation (Bilderback et al., J Neurosci Res 49: 497 (1997); Chan et al., PNAS USA 95: 10459 (1998)).

Briefly, the uterus is removed from female rats that are 14-15 days pregnant (plug day is designated as day 0 of pregnancy) in order to obtain embryonic Schwann cells. Thereafter, the embryos are removed from the uterus. Using a dissecting microscope, the entire length of spinal cord is exposed. Dorsal root ganglia (DRGs) are removed from both sides of the spinal cord using forceps. The DRGS are put into culture with collagenase at 37C for 30-45 minutes, washed and resuspended in HamsF12/DMEM medium (1: 1 mixture) supplemented with growth factors. The cell suspension is then placed in 24-well plates that have been coated with natural mouse laminin.

Postnatal day 0-1 rats are used for the preparation of neonatal rat Schwann cells and the dissociation of DRGs is performed with collagenase as described above. Adult rat Schwann cells are usually prepared from young adult rats 60-90 days old. The dissociation is performed with a 10% collagenase/dispase solution at 37C for 45 minutes. Human Schwann cells can be prepared from donated organs. The sural nerve is particularly useful. Again, dissociation is achieved with mild enzyme treatment and culturing is done on laminin-coated plates.

After three days, the cultures on laminin will form a confluent monolayer of rapidly dividing Schwann cells. The cells can be subcultured every 4 days at a 1: 4 split ratio.

To obtain neurons for coculture with Schwann cells for remyelination, a portion of the dissociated DRGs (after washing but before culturing) are placed in a tube with a 5% BSA layer and centrifuged such that the cloudy cell suspension stays on top of the layer (and is discarded) and the pellet is recovered, treated with a 0. 125% trypsin solution, washed and resuspended.

The recovered material can be further purified on another 5% BSA layer at lg. The neurons are

grown in the laminin-coated wells in F12/DMEM supplemented with 5 ng/ml neurotropin 3 and 15% fetal bovine serum.

To test the effects of tramdorin expression on Schwann cell proliferation, differentiation, and survival, tramdorin proteins will be expressed in cultured primary Schwann cells using retroviral vectors. Retroviral infection of Schwann cells has several advantages.

First, the ability to transfect primary Schwann cells with retroviral vectors is well established, and infected cells can myelinate (Howe et al., JNeurosci Methods 83: 133 1998; Owens, Ann NYAcad Sci 633: 543 (1991); Owens et al., Development 112: 639 (1991); Owens et al., J Cell Biol 111 : 1171 (1990); Owens et al., Neuron 7: 565 (1991); Zoidl et al., Enibo J 14 : 1122 (1995) ). Second, the introduced DNA integrates into the genome, generating a stably transfected cell. To constitutively express exogenous genes in primary Schwann cells, a dicistronic retroviral system, LIRES-GFP, that permits myelination of infected primary Schwann cells (Howe et al., JNeurosci Methods 83: 133 (1998) ) will be used. The LIRES-GFP vector has been kindly provided by K. McCarthy. The reading frames will be inserted into the LIRES-GFP vector, after addition of Kozak start sites, and C-terminal HA tags. The pBKS plasmid (Stratagene) has been modified to generate a convenient shuttle vector for adding these sequences. Retroviruses will be made using the Ecopak ecotropic cell line (Clontech) that permits infection of only rat or mouse cells. After infection of primary Schwann cells with tramdorin retroviruses, the effects of tramdorin expression on Schwann cell proliferation, and on the expression of Oct-6 and myelin P0, will be measured, using antisera specific for these proteins (Bermingham et al., Gene Develop 10: 1751 (1996); Scherer et al., JNeurosci 15: 8281 (1995) ). Any effects of its expression in the presence and absence of the putative tramd ligand GABA will be measured.

Myelinating Schwann cell lines may provide an alternative to the use of primary Schwann cell lines. Two myelinating Schwann cell lines have been reported. The SCL4. 1/F7 cell line was generated by multiple passaging of primary Schwann cells with intermittent exposure to cholera toxin (Haynes et al., JNeurosci Methods 52: 119 (1994) ). CRlb4 is a Schwann cell line that was immortalized using a temperature sensitive oncogene (Thi et al., J Exp Biol 201 : 851 (1998)).

C. Generation of Mice That Lack Tramdorin Genes To determine the extent to which tramdorin expression is required for peripheral myelination, mice that lack the tramdl gene (SEQ ID NO : 7) will be generated. Two distinct mutations will be made. The first will generate a conditional knockout of the tramdorini gene (SEQ ID NO : 7), in which tramdorin 1 is deleted only in Schwann cells. This approach is based on use of the bacteriophage site-specific recombinase Cre, which recognizes 34bp loxP sites (reviewed in (Sauer, Methods 14: 381 (1998) ). To generate a mouse in which a gene is deleted in a specific tissue (reviewed in (Marth, J Clin Invest 97: 1999 (1996) ), the region to be deleted is flanked by loxP sites. For tramdorinl, exon 2 will be flanked with loxP sites, thereby permitting its specific deletion in the presence of Cre recombinase. Such a deletion will remove conserved (and therefore presumably important) sequences, and will cause the remainder of the tramdorin message to be shifted out of frame. Because the two loxP sites are located in the introns surrounding exon2, the mutation will have no effect in the absence of Cre recombinase. Such"floxed"mice will be crossed to transgenic mice in which Cre recombinase is expressed under control of the PO promoter (Feltri et al., Ann NYAcad Sci 883: 116 (1999); Giovannini etal., GenesDev 14: 1617 (2000); Yamauchi et al., Dev Biol 212 : 191 (1999)).

Mice that carry both the transgene, and are homozygous for the floxed tramdorin allele will have the tramdorin gene specifically mutated in Schwann cells. That is to say, it will take another backcross to the floxed mouse to get homozygous floxed and the transgene.

The second mutation will fuse the E. coli LacZ gene in frame to tramdorinl exon 2, thereby truncating the protein and permitting its expression to be monitored by P-galactosidase activity.

D. Screening for Tramd Interacting Proteins Although a precise understanding of the mechanism of tramdorin action is not required for the practice of the present invention, it is hypothesized that tramdorin is required for proper myelination. Tramdorin may transduce a signal from axons that promotes myelination, and therefore may be a crucial reagent in the identification of the myelinogenic signal itself (see Identification of Tramdorin Ligands, above). Alternatively, tramdl may interact directly with molecules that are expressed on the surface of axons, or it could act as a cofactor for other

Schwann cell signaling molecule (s). Thus, identification of proteins which interact with tramdorins (especially tramdorin 1) is likely to contribute to an understanding of the myelination process. A number of methods are available to detect protein-protein interactions, including but not limited to co-immuoprecipitation, affinity chromatography with immobilized tramdorin 1 protein or fragments or portions of the tramdorin 1 protein, the yeast two-hybrid system and cross-linking. Any suitable method for detection of tramdorin-interacting proteins is contemplated, including the yeast-two hybrid screen and its variations.

Yeast two-hybrid screening (Fields et al., Nature 340: 245 (1989); U. S. Patent 5,667, 973, herein incorporated by reference) is an important approach for identifying protein- protein interactions (reviewed in (Fields et al., Trends Genet 10: 286 (1994) ). Typically, a protein of interest, the bait, is fused to the DNA binding domain of yeast Gal4, or E. coli LexA.

A library of targets, or"prey"are fused to a transcriptional activator. The bait and prey fusion proteins are coexpressed, and if they interact, marker genes, which contain a binding site for the DNA-binding domain, are expressed that permit isolation of plasmids that encode the interacting protein. The marker gene may encode an enzyme or other product that can be readily measured. Such measurable activity may include the ability of a cell to grow only when the marker gene is transcribed, the presence of detectable enzymatic activity (e. g. (3-galactosidase encoded by the LacZ gene) only when the marker gene is transcribed, or light release, depending on the type of reporter or marker gene activated. Commercially available kits for yeast two-hybrid screening are available from Clontech and Stratagene. However, because the bait and prey proteins are imported to the nucleus, the standard yeast two-hybrid methodology precludes posttranslational protein modifications that may be critical for some protein-protein interactions. Furthermore, membrane proteins are underrepresented among interacting protein pairs that have been found by two-hybrid screening (Uetz et al., Nature 403: 623 (2000)).

Recently, a new yeast two-hybrid system, the Ras recruitment system (RRS), has been developed that utilizes a cytoplasmic interaction to activate a Ras signaling pathway (reviewed in (Broder et al., Curr Biol 8 : 1121 (1998) ). U. S. Patent 5,776, 689, herein incorporated by reference, also describes the protein recruitment system to detect protein-protein interaction at or in a specific cell compartment. A yeast two hybrid system, CytoTrap, that is based on RRS methodology, is available from Stratagene. Briefly, this system involves the complementation

of a temperature-sensitive mutation in the yeast cdc25 gene, the yeast homolog for hSos, by proper localization of human Sos to the plasma membrane by protein-protein interaction. A library of target proteins are expressed as fusion proteins with src myristylation sites that anchor the proteins to the membrane. The bait is fused to human Sos, and interaction with target proteins localizes Sos to the membrane, thereby permitting cell survival and growth at 37°C by activation of the Ras signaling pathway. Thus, when the cDNA library and the tramdl-hSos fusion construct are transformed into the cdc25H yeast strain (Stratagene), the only cells capable of growing at 37°C on galactose medium are those that have been rescued by a protein- protein interaction recruiting hSos to the cell membrane.

A reverse RRS technique permits full length transmembrane domain proteins to be used as bait (Hubsman et al., Nucleic Acids Res 29: E18 (2001) ), has recently been described. This reverse RRS system is contemplated herein for the isolation of proteins that interact with tramdorins. Full-length tramdorin will be expressed in yeast whereas the prey will be fused to a mutant Ras protein that lacks a membrane localization signal and stop codon. Those fusion proteins that interact with tramdorin will be localized to the membrane, thereby permitting growth at restrictive temperature of yeast that carry the temperature-sensitive Cdc25-2 mutation.

Although a precise understanding of the localization of tramdorin is not required for the practice of the invention, it is possible that tramdorin may be localized to lysosomes. If this is the case, then the recruitment of Ras activity to lysosomes may not properly activate ras signaling pathways using the reverse RRS technique. Therefore, conventional yeast two-hybrid screening for tramdorin-interacting proteins is also contemplated. The following two baits will be used in the conventional screening: (i) the N-terminal 51 amino acids of mouse tramdorinl : MSVTKSARSPQVATPLNLDLPESAKKLQSQDPSPANGTSSESSKKTKGITG (SEQ ID NO: 15) This is encoded by the DNA sequence: ATG TCT GTG ACC AAG AGT GCC AGG AGT CCG CAG GTA GCC ACC CCT CTC AAT CTG GAC CTT CCT GAG AGT GCC AAG AAG CTG CAG AGC CAG GAT CCC AGT CCA GCG AAT GGG ACC TCT TCA GAG TCA TCA AAG AAG ACC AAG GGC ATA ACC GGG (SEQ ID NO : 45)

(ii) the first putative intracellular loop of mouse tramdorin 1, which consists of the following 39 amino acid sequence: RCAQRFCHRLNKPFMDYGDTVMHGLAFSPNAWLQNHAHW (SEQ ID NO : 16) This is encoded by the DNA sequence: AGA TGT GCC CAG CGC TTC TGT CAC AGA CTG AAC AAG CCT TTC ATG GAC TAT GGG GAC ACA GTG ATG CAC GGA CTG GCT TTC AGT CCC AAT GCC TGG CTG CAA AAC CAC GCC CAC TGG (SEQ ID NO : 46) These are the largest amino acid sequences within the tramdorin 1 protein that do not contain predicted transmembrane domains. The N-terminal 51 amino acids have proven to be toxic when expressed in E. coli. If the same holds true in yeast, the intracellular loop bait will be used. DNA sequences that encode the baits will be cloned into pSos (Stratagene) for RRS two hybrid screening, or pBD-Gal4 (Stratagene) for conventional Gal4-based yeast two hybrid screening, or pLexA (Clontech) for conventional yeast two-hybrid screening based on LexA.

For both the RRS and conventional yeast two-hybrid screening the library of prey interacting proteins will be derived from an adult mouse sciatic nerve cDNA library that will be constructed as follows: Sciatic nerves have been collected from adult male FVB/NJ mice (these are available free of charge). Total RNA has been isolated from the sciatic nerves by CsCl2 gradient centrifugation (Chirgwin et al., Biochem 18: 5294 (1979)). cDNA will be synthesized from poly A+ purified RNA using Superscript cDNA synthesis kit (Gibco-BRL), and cloned into the pYes2 (Invitrogen) for reverse RRS screening, the yeast two hybrid library plasmid pMyr (Stratagene) for RRS screening, or pAD-Gal4-2.1 (Stratagene) for conventional Gal4-based yeast two hybrid screening, or pB42AD (Clontech) for conventional yeast two hybrid screening based on LexA.

By way of example of selection and screening for protein-protein interactions, in the Stratagene system, as described by the manufacturer, yeast (strain YRG-2; Mata, ura3-52, his3- 200, ade2-101, lys2-801, trpl-901, leu2-3 11, gal4-542, gal80-538, LYS2: : UASGALl- TATAGALo-HIS3 URA3 :: UASoAL4 i7mers (x3)-TATAcYci-lacZ) can be transformed with the pBD- GAL4/protein of interest fragment (i. e. tramd 1 in one embodiment contemplated herein) vector and the mouse sciatic nerve cDNA library contained in the pAD-GAL4-2.1 vector and selected on plates that lack leucine (selection for the pAD-GAL4-2. 1 vector) and tryptophan (selection

for the pBD-GAL4 vector), and screening can be done by virtue of the histidine and LacZ reporter genes in YRG-2.

E. Analysis of Tramdorin Gene Expression Patterns 1. Tissue Preparation The spatial and temporal expression patterns of the tramdorin genes were identified by in situ hybridization using radiolabelled RNA probes that specifically recognize individual tramdorin genes. For in situ hybridization, tissue sections were obtained as follows. Mice were anesthetized and perfused with 4% paraformaldehyde or formalin. Hindquarters were dissected to expose the nerve, then stored at 4°C in 4% paraformaldehyde or formalin, then processed as follows. Tissues were frozen in a 1: 1 mixture of OCT and Aquamount, and sectioned at 20 microns.

2. In Situ Hybridization In situ hybridization was performed as described by Simmons et al., J. Histotechnology 12: 169 (1989). Darkfield micrographs of adjacent sections of PO mouse leg (prepared as described in the previous section) containing femur and fat pad were hybridized with 35S labeled probes specific for tramdl, tramd2 or tramd3. As seen in Figure 31, the tramdl probe hybridized to fat pads, and to bone marrow, but not to growth plate. No hybridization to either tissue was detected for tramd2 or tramd3. While it is not intended that the present invention be limited to any specific mechanism, these data suggest a role for glycine signaling in the differentiation of bone and fatty tissue. The probes consist of 343nt between nucleotides 1549 and 1892 of mouse tramdorinl cDNA cDNA AF512429; 347nt between nucleotides 2 and 349 of mouse tramdorin 2 cDNA, and 323nt between nucleotides 1506 and 1829 of mouse tramdorin 3 cDNA. Scale bar: 500 microns.

It has been shown that that bone has NMDA (N-methyl-D-aspartate) receptors. See, Gu et al.,"Expression Of Functional Metabotropic Glutamate Receptors In Primary Cultured Rat Osteoblasts", J. Biol. Chem. , Vol. 275, pp. 34252-34259 (2000). In one embodiment of the present invention, therefore, it is contemplated that tramdorins may be used as a marker in bone. In another embodiment, it is contemplated that tramdorins may be administered to a

subject to modulate the proliferation and differentiation of bone cells (including, but not limited to, osteoblasts and osteoclasts).

In one embodiment of the present invention, it is contemplated that tramdorins may be used as a marker in adipose tissue. In another embodiment, it is contemplated that tramdorins may be administered to a subject to modulate the proliferation of fat cells (including, but not limited to, adipocytes) and the incorporation of glycine into fat cells.

3. Preparation Of Probes Specific For Tramdl, Tramd2 and Tramd3 The probes used in the in situ hybridization reactions, as depicted in Figure 31, were prepared as follows.

Mouse tramdorinl probe (SEQ ID NO: 71). Mouse tramdl sequences, shown in bold, were amplified by PCR, and cloned into pCRII (Invitrogen). The insert was excised using EcoRI and cloned into the EcoRI site of pBKSII+ (Stratagene). EcoRI sites are boxed. The probe is transcribed from the T7 promoter, shown in italics, using T7 polymerase, using plasmid DNA that has been linearized at the Hindi site, which is underlined. The mouse tramdorin sequence consists of 343bp between nucleotides 1549 and 1892 of AF512429, shown in antisense orientation.

Mouse tramdorin2 probe (SEQ ID NO: 72). 347 bp of mouse tramdorin 2 sequences between nucleotides 2 and 349, shown in bold, were amplified by PCR, and cloned into pCRII (Invitrogen) They are shown in antisense orientation. EcoRI sites are boxed. The probe was transcribed from the T7 promoter, shown in italics, using T7 polymerase, using plasmid DNA that has been linearized at the BamHI site, which is underlined.

Mouse tramdorin3 probe (SEQ ID NO; 73). 323 bp of mouse tramdorin3 sequences between nucleotides 1506 and 1829 of mouse tramdorin3 composite cDNA 50+61-45+76, shown in bold, were amplified by PCR, and cloned into pCRII (Invitrogen). They are shown in antisense orientation. EcoRI sites are boxed. The probe was transcribed from the T7 promoter, shown in italics, using T7 polymerase, using plasmid DNA that has been linearized at the BamHI site, which is underlined.

4. Tramdorin Antiserum i. Preparation Antisera against tramdorin proteins permit their subcellular localization to be determined by immunohistochemistry and confocal microscopy. Antisera were raised in two rabbits against the mouse tramdorin 1 C-terminal peptide CGTYQALDELIKSGNSPA (SEQ ID NO : 47). The antisera were affinity purified using a Sulfolink column (Pierce) that contained covalently bound peptide. However, the antisera showed extremely weak signals on tissue sections under a variety of conditions. To obtain antisera that provide better signal on tissue sections, a synthetic peptide: ESAKKLQSQDPSPANGTSC (SEQ ID NO: 74), containing amino acids 22-39 near the N-terminus of tramdorinl, was coupled to KLH and injected into two rabbits, one of which generated useful antiserum. It should be noted that the C-terminal cystine is used for linkage to KLH.

In other embodiments alternative approaches, such as expression the N-terminus as a GST fusion protein or to generate antisera against synthetic peptide (s) that correspond to this region, may be used.

While the cloning of this tramdorin 1 N-terminus expression plasmid has proven to be non-trivial, there are viable alternative strategies to obtain antisera against the N-terminus of mouse tramdorin 1. By way of non-limiting example, the N-terminus can be expressed as a GST-fusion protein, which permits purification of the fusion on an affinity column. The GST portion of the fusion can be removed before injection into animals for antisera generation, or the entire fusion can be used to generate antiserum, as long as the antiserum is affinity cleared of any GST-reactive antibodies before use in localization studies. Another strategy is to generate antisera against synthetic peptide (s) that correspond to this region. Such synthetic peptides can be chemically prepared by methods well known in the art, and do not require expression in a host organism.

The present invention provides isolated antibodies. An antibody against a protein of the present invention (i. e. a tramdorin protein or fragment or portion of a tramdorin protein) may be any monoclonal or polyclonal antibody, as long as it can recognize the protein of interest.

Antibodies can be produced by using a protein, or fragment thereof, or synthetic peptide, of the

present invention as the antigen according to a conventional antibody or antiserum preparation process.

The present invention contemplates the use of both monoclonal and polyclonal antibodies. Any suitable method may be used to generate the antibodies used in the methods and compositions of the present invention, including but not limited to, those disclosed herein.

For example, for preparation of a monoclonal antibody, protein, as such, or together with a suitable carrier or diluent is administered to an animal (e. g., a mammal) under conditions that permit the production of antibodies. For enhancing the antibody production capability, complete or incomplete Freund's adjuvant may be administered. Normally, the protein is administered once every 2 weeks to 6 weeks, in total, about 2 times to about 10 times. Animals suitable for use in such methods include, but are not limited to, primates, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, etc.

For preparing monoclonal antibody-producing cells, an individual animal whose antibody titer has been confirmed (e. g., a mouse) is selected, and 2 days to 5 days after the final immunization, its spleen or lymph node is harvested and antibody-producing cells contained therein are fused with myeloma cells to prepare the desired monoclonal antibody producer hybridoma. Measurement of the antibody titer in antiserum can be carried out, for example, by reacting the labeled protein, as described hereinafter and antiserum and then measuring the activity of the labeling agent bound to the antibody. The cell fusion can be carried out according to known methods, for example, the method described by Koehler and Milstein [Nature 256: 495 (1975) ]. As a fusion promoter, for example, polyethylene glycol (PEG) or Sendai virus (HVJ), preferably PEG, is used.

Examples of myeloma cells include NS-1, P3U1, SP2/0, AP-1 and the like. The proportion of the number of antibody producer cells (spleen cells) and the number of myeloma cells to be used is preferably about 1: 1 to about 20: 1. PEG (preferably PEG 1000-PEG 6000) is preferably added in concentration of about 10% to about 80%. Cell fusion can be carried out efficiently by incubating a mixture of both cells at about 20°C to about 40°C, preferably about 30°C to about 37°C for about 1 minute to 10 minutes.

Various methods may be used for screening for a hybridoma producing the antibody (e. g., against a tramdorin protein or portion thereof). For example, where a supernatant of the

hybridoma is added to a solid phase (e. g., microplate) to which protein is adsorbed directly or together with a carrier, and then an anti-immunoglobulin antibody (if mouse cells are used in cell fusion, anti-mouse immunoglobulin antibody is used) or Protein A labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase. Alternately, a supernatant of the hybridoma is added to a solid phase to which an anti-immunoglobulin antibody or Protein A is adsorbed and then the protein labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.

Selection of the monoclonal antibody can be carried out according to any known method or its modification. Normally, a medium for animal cells to which HAT (hypoxanthine, aminopterin, thymidine) are added is employed. Any selection and growth medium can be employed as long as the hybridoma can grow. For example, RPMI 1640 medium containing 1% to 20%, preferably 10% to 20% fetal bovine serum, GIT medium containing 1% to 10% fetal bovine serum, a serum free medium for cultivation of a hybridoma (SFM-101, Nissui Seiyaku) and the like can be used. Normally, the cultivation is carried out at 20°C to 40°C, preferably 37°C for about 5 days to 3 weeks, preferably 1 week to 2 weeks under about 5% C°2 gas. The antibody titer of the supernatant of a hybridoma culture can be measured according to the same manner as described above with respect to the antibody titer of the anti-protein in the antiserum.

Separation and purification of a monoclonal antibody (e. g., against tramdorin or a fragment of tramdorin) can be carried out according to the same manner as those of conventional polyclonal antibodies such as separation and purification of immunoglobulins, for example, salting-out, alcoholic precipitation, isoelectric point precipitation, electrophoresis, adsorption and desorption with ion exchangers (e. g., DEAE), ultracentrifugation, gel filtration, or a specific purification method wherein only an antibody is collected with an active adsorbent such as an antigen-binding solid phase, Protein A or Protein G and dissociating the binding to obtain the antibody.

Polyclonal antibodies may be prepared by any known method or modifications of these methods including obtaining antibodies from patients. For example, a complex of an immunogen (an antigen against the protein) and a carrier protein is prepared and an animal is

immunized by the complex according to the same manner as that described with respect to the above monoclonal antibody preparation. A material containing the antibody against is recovered from the immunized animal and the antibody is separated and purified.

As to the complex of the immunogen and the carrier protein to be used for immunization of an animal, any carrier protein and any mixing proportion of the carrier and a hapten can be employed as long as an antibody against the hapten, which is crosslinked on the carrier and used for immunization, is produced efficiently. For example, bovine serum albumin, bovine cycloglobulin, keyhole limpet hemocyanin, etc. may be coupled to an hapten in a weight ratio of about 0.1 part to about 20 parts, preferably, about 1 part to about 5 parts per 1 part of the hapten.

In addition, various condensing agents can be used for coupling of a hapten and a carrier. For example, glutaraldehyde, carbodiimide, maleimide activated ester, activated ester reagents containing thiol group or dithiopyridyl group, and the like find use with the present invention. The condensation product as such or together with a suitable carrier or diluent is administered to a site of an animal that permits the antibody production. For enhancing the antibody production capability, complete or incomplete Freund's adjuvant may be administered.

Normally, the protein is administered once every 2 weeks to 6 weeks, in total, about 3 times to about 10 times.

The polyclonal antibody is recovered from blood, ascites and the like, of an animal immunized by the above method. The antibody titer in the antiserum can be measured according to the same manner as that described above with respect to the supernatant of the hybridoma culture. Separation and purification of the antibody can be carried out according to the same separation and purification method of immunoglobulin as that described with respect to the above monoclonal antibody.

The protein used herein as the immunogen is not limited to any particular type of immunogen. For example, tramdorin can be used as the immunogen. Further, fragments of the protein may be used, including but not limited to the amino terminus of tramdorin. Fragments may be obtained by any methods including, but not limited to expressing a fragment of the gene, enzymatic processing of the protein, chemical synthesis, and the like.

V. Tramdorins and Parathyroid Tumors As noted above, a fourth human tramdorin-related sequence, tramdL, is defined by the EST cDNA 1388139 (SEQ ID NO : 23) (Figures 8D and 12). tramdL contains only sequences that are homologous to exon 10 of tramdorinl. This cDNA is derived from a parathyroid adenoma, and is unusual in that its 5'end consists of sequences derived from a member of the STE20 family of kinases, STK4/Mst-l/Krs-2 (Figure 12A; Creasy et al., JBiol Chem 270: 21695 (1995); Taylor et al., PNAS USA 93: 10099 (1996) ), located on human chromosome 20. The STK4/Mst-l/Krs-2 cDNA fragment is fused to 3'sequences of tramdL (Figure 12B), located on chromosome 5. Comparison of the genomic sequences for STK4/Mst-l/Krs-2 and tramdL reveal that the fusion site corresponds to a 5'splice site in the STK4/Mst-1/Krs-2 gene, and a 3'splice site in tramdL (Figure 12C). Such a fusion is unlikely to be an artifact of cDNA synthesis. While an understanding of the precise mechanism is not necessary to the practice of the invention, and noting that the invention is not to be limited to any particular mechanism, this observation raises the possibility that tramdL resides near a parathyroid tumor translocation breakpoint, between chromosomes 20 and 5, that results in expression of a STK4/Mst-l/Krs-2- tramdorinL fusion protein. STK4/Mst-l/Krs-2 contains a C-terminal inhibitory domain (Creasy et al., JBiol Chem 271: 21049 (1996) ) that is cleaved by Caspase-3 to generate a cleavage product, p36, with kinase activity (Graves et al., Embo J 17: 2224 (1998) ; Lee et al., Oncogene 16: 3029 (1998); Watabe et al., JBiol Chem 275: 8766 (2000) ). The STK4/Mst-l/Krs-2- tramdorinL fusion protein would replace STK4/Mst-l/Krs-2 C-terminal sequences with a C- terminal transmembrane domain from tramdL (Figure 12D). Based on the activity of C- terminal deletions of STK4/Mst-l/Krs-2 (Creasy et al., JBiol Chem 271 : 21049 (1996) ), the fusion protein may retain kinase activity, and may represent an important aspect of the etiology of parathyroid tumors.

In one embodiment of the present invention, it is contemplated that tramdorins may be used as a marker in bone. In another embodiment of the present invention, it is contemplated that tramdorins may be administered to a subject to modulate the proliferation and differentiation of bone cells (including, but not limited to, osteoblasts and osteoclasts).

A. Parathyroid Adenoma Background Primary hyperparathyroidism is a disorder of the parathyroid glands. Most people with this disorder have one or more enlarged, overactive parathyroid glands that secrete too much parathyroid hormone. The parathyroid glands are four pea-sized glands located on the thyroid gland in the neck. The parathyroid glands secrete parathyroid hormone (PTH), a hormone that helps maintain the correct balance of calcium and phosphorus in the body. PTH regulates the release of calcium from bone, absorption of calcium in the intestine, and excretion of calcium in the urine. When the amount of calcium in the blood falls too low, the parathyroid glands secrete just enough PTH to restore the balance.

If the glands secrete too much hormone, as in hyperparathyroidism, the balance is disrupted: blood calcium rises. This condition of excessive calcium in the blood, called hypercalcemia, is what usually signals the doctor that something may be wrong with the parathyroid glands. In 85 percent of people with this disorder, a benign tumor (an adenoma) has formed on one of the parathyroid glands, causing it to become overactive. The excess PTH triggers the release of too much calcium into the bloodstream. The bones may lose calcium, and too much calcium may be absorbed from food. The levels of calcium may increase in the urine, causing kidney stones. PTH also acts to lower the blood phosphorus levels by increasing excretion of phosphorus in the urine.

In the United States, about 100,000 people develop hyperparathyroidism each year.

Women outnumber men by 2 to 1, and risk increases with age. In women 60 years of age and older, 2 out of 1,000 will get hyperparathyroidism. Primary hyperparathyroidism is found in up to 2. 1% of postmenopausal women (Lundgren et al., Surgery 121: 287 (1997) ). A person with hyperparathyroidism may have severe symptoms, subtle symptoms, or no symptoms at all.

Routine blood tests that screen for high calcium levels are alerting doctors to people who, though symptom-free, have mild forms of the disorder. When symptoms do appear, they are often mild and nonspecific, such as feelings of weakness and fatigue, depression, or aches and pains. With more severe disease, a person may have a loss of appetite, nausea, vomiting, constipation, confusion or impaired thinking and memory, and increased thirst and urination.

Patients may have thinning of the bones without symptoms, but with risk of fractures. Increased calcium and phosphorus excretion in the urine may cause kidney stones. Subjects with

hyperparathyroidism may be more likely to develop peptic ulcers, high blood pressure, and pancreatitis.

Hyperparathyroidism is diagnosed when tests show that blood levels of calcium as well as parathyroid hormone are too high. Once the diagnosis is established, other tests may be done to assess complications. Because high PTH levels can cause bones to weaken from calcium loss, a measurement of bone density may be done to assess bone loss and the risk of fractures.

Abdominal radiographs may reveal the presence of kidney stones and a 24-hour urine collection may provide information on kidney damage and the risk of stone formation.

At the present time, surgery to remove the enlarged gland is the only treatment for the disorder. While surgery is an effective cure in 95% of cases, by nature, surgery is invasive and carries risks of complications, including but not limited to post-surgical infection. Some patients who have mild disease may not need immediate treatment. Such patients are subject to long-term monitoring which includes regular testing of calcium levels, bone mass and kidney function. Such subjects must avoid certain medications (such as diuretics such as the thiazides) and consult a physician in cases of immobilization or gastrointestinal disease with vomiting and diarrhea, which can cause calcium levels to rise. Thus, a non-invasive therapeutic, an animal model to test such therapeutics, and a diagnostic test to determine the presence of certain genomic rearrangements in parathyroid adenomas to direct the therapeutic regimen would all be useful.

At the present time, the underlying molecular genetic pathology of parathyroid adenomas is not fully understood. In a subset of parathyroid adenomas, a tumor-specific DNA rearrangement (a pericentromeric inversion, inv (l l) (pl 5 ; ql3)) which results in the fusion of the cyclin D 1 gene (CCND1) with the parathyroid hormone regulatory region is found.

Immunohistochemistry has shown that cyclin D 1 is overexpressed, in 20-40% of parathyroid adenomas. However, other (non cyclin dependent) mechanisms may account for parathyroid cell proliferation in the remaining cases (see for example, Mallya et al., Frontiers in Bioscience 5 : d367-371 (2000) ). Mallya et al. (supra) also note that mice which harbor a transgene in which the cyclin D 1 gene is placed under the control of the parathyroid hormone (PTH) regulatory region (to mimic the chromosome 11 rearrangement and resultant cyclin D1 overexpression in the human tumors) develop hyperparathyroidism by the age of six months, as

evidenced by parathyroid enlargement and increased serum calcium and parathyroid hormone levels. Thus, tissue specific expression of cyclin D1 does induce parathyroid cell proliferation resulting in hyperparathyroidism. However, this etiology is only likely to account for 20-40% of parathyroid adenomas.

B. Tramdorins in Parathyroid Adenomas While not limited to any particular mechanism, and with the note that an understanding of the underlying mechanism is not necessary to the practice of the invention, the identification of a putative STK4/Mst-l/Krs-2-tramdorinL fusion gene that is derived from parathyroid adenomas suggests that a membrane-bound, constitutively active kinase may function in the etiology of these tumors. Parathyroid adenoma DNA has been examined for allelic loss (Tahara et al., Cancer Res 56: 599 (1996) ), or chromosome gain or loss (Palanisamy et al., J Clin Endocrinol Metab 83: 1766 (1998) ), as an approach to identifying parathyroid tumor suppressor genes, but these studies would not necessarily detect translocations. The putative STK4/Mst- 1/Krs-2-tramdorinL fusion gene is suggestive of a t (20; 5) translocation in at least one parathyroid adenoma. STK4/Mst-l/Krs-2 has been shown to be activated by apoptotic signals (Graves et al., Enabo J 17: 2224 (1998); Lee et al., Oncogene 16: 3029 (1998) ; Watabe et al., J Biol Chem 275: 8766 (2000) ), and its active proteolytic fragment has been found in several tumor cell lines (Watabe et al., JBiol Chem 275: 8766 (2000), but its physiological role is not understood. If the putative truncated, membrane-bound form of the kinase contributes to the etiology of parathyroid adenomas, it may provide avenues toward new therapies for a disorder that will increase in frequency in the United States as the average age of Americans increases.

A number of means are contemplated for determining the role of the putative STK4/Mst-l/Krs-2-tramdorinL fusion gene in parathyroid adenomas, and the invention is not intended to be limited to any particular means of evaluating the putative STK4/Mst-l/Krs-2- tramdorinL fusion gene in parathyroid adenomas. For example, DNA-based assays are contemplated, wherein the fusion is detected by PCR amplification of cDNA from parathyroid adenomas using primers corresponding to the tramdL portion of the fusion and the STK4/Mst- l/Krs-2 portion of the fusion, such that amplification will occur across the fusion junction if the fusion is present, but no amplification will occur in the absence of the fusion. RNA will be

isolated from several independent parathyroid adenomas, as well as other parathyroid tumors, and RT-PCR will be carried out using primers that correspond to tramdL sequences. The cDNAs that will be obtained from these tissues will determine if the fusion transcript is present in parathyroid tumor tissue. Limited parathyroid adenoma tissue is available from Research Genetics. Additional samples will be obtained from tissue banks, or other sources, such as local physicians. Primers that can be used to detect the fusion transcript are: 5'TGCCAAAGGAGTGTCAATACTGCG 3' (STK4/Mst-l/Krs-l) (SEQ ID NO : 48) 5'TCCCCACCAGAAATCCCACAAAGC 3' (tramdL) (SEQ ID NO : 49) These primers have an optimal annealing temperature of 59°C, and will amplify a 494bp product from STK4/Mst-l/Krs-l-tramdL fusion cDNAs. Therefore they may also be used for diagnosis of translocations that produce STK4/Mst-l/Krs-l-tramdL fusion transcripts. If fusion cDNAs are detected, primers that correspond to the initiation ATG codon of STK4/Mst-l/Krs- 1, and the termination TAG codon of tramdL, or sequences 3'to it, will be used for RT-PCR to amplify the entire STK4/Mst-l/Krs-l-tramdL coding sequence for expression studies.

Molecular cytogenetic analysis, such as FISH, is also contemplated for detection of the presence of the STK4/Mst-l/Krs-2-tramdL fusion in cells harvested from parathyroid tumors.

Bacterial artificial chromosomes (BACs) with sequences corresponding to tramdL genomic sequences will be obtained, as will BACs with inserts corresponding to the genomic sequence of STK4/Mst-l/Krs-2. The tramdL and STK4/Mst-l/Krs-2 BACS will be differentially labeled.

For example, the tramdL BAC can be labeled with a marker which permits detection of a green fluorescent signal, such as by direct incorporation of fluorescein or FITC, or by incorporation of biotin, which can be detected by FITC-conjugated avidin. The STK4/Mst-l/Krs-2 BAC can be labeled with a marker which permits detection of a red fluorescent signal, such as by direct incorporation of TRITC or by incorporation of digoxigenin, which can be detected by rhodamine-conjugated anti-digoxigenin antibodies. The labeled probes are hybridized to a cytogenetic preparation of an isolated parathyroid tumor, or cultured cells derived from isolated parathyroid tumors. As a normal control, normal parathyroid tissue (or cultured cells) or peripheral blood cells can be used. The probes can be prepared by a number of methods,

including PCR amplification and nick translation. Suitable nick translation protocols are provided in Human Chromosomes, Principles and Techniques (2E), Verma and Babu (Ed.) Chapter 6 (pp. 185-231) (1995) and Lichter and Ried"Molecular Analysis of Chromosome Aberrations. In situ Hybridization"pp. 449-478 in Methods in Molecular Biology, Vol. 29: Chromosome Analysis Protocols. Edited by J. R. Godsen. (1994). Following hybridization, washing and detection steps, (as described in Verma and Babu and Lichter and Ried, supra) the specimens are counter-stained with 4', 6-diamidino-2-phenyl indole dihydrochloride (DAPI) (to permit visualization of the nuclei and metaphase chromosomes in blue), and observed and imaged on a fluorescence microscope equipped with a mercury or xenon lamp and the appropriate filter sets. In the normal controls, the expected result on an interphase nucleus is two green spots (or two closely juxtaposed"doublets"in nuclei that have replicated their DNA), corresponding to the two tramdL alleles on chromosome 4, and two red spots (or two closely juxtaposed"doublets"in nuclei that have replicated their DNA), corresponding to the two STK4/Mst-l/Krs-2 alleles on chromosome 20. In parathyroid adenomas with a STK4/Mst- 1/Krs-2-tramdL fusion, the expected result (in a diploid or near-diploid cell) is a single green spot (or doublet), corresponding to the tramdL allele that is not involved in the fusion, a single red spot (or doublet), corresponding to the STK4/Mst-l/Krs-2 allele that is not involved in the fusion, and a yellowish-orange"fusion spot"resulting from the near super-imposed hybridization of the green tramdL probe and the red STK4/Mst-l/Krs-2 probe hybridizing to the translocation-derived STK4/Mst-l/Krs-2-tramdL fusion.

The detection of"fusion"FISH signals is well known in the art as a means to detect translocations, for example, in the detection of the BCR/ABL rearrangement that fuses the ABL proto-oncogene on chromosome 9 to the BCR (breakpoint cluster region) gene on chromosome 22 and which is frequently present in chronic myelogenous leukemia (CML) (see, for example, "Cellular Genomic Products for Hematopoietic Disorders", available from Vysis, Inc. Downers Grove, IL, http://www. vysis. com). Using dual-color FISH, the proportion of parathyroid tumors harboring a STK4/Mst-l/Krs-2-tramdL fusion can be determined.

It is also possible, and known in the art to use"chromosome painting"probes to detect translocations in metaphase spreads (see for example, Speicher et al., Nat Genet 12: 368-375, (1996) ). Thus, a whole chromosome paint probe to chromosome 5, and a whole chromosome

paint probe to chromosome 20 can be hybridized to metaphase chromosome spreads from parathyroid tumors. Differentially labeled painting probes are commercially available, for example WCPO 20 Spectrum Green from Vysis (Downers Grove, IL) is a probe which hybridizes to both arms and the centromere of chromosome 20. WCP 50 Spectrum OrangeT"" from Vysis (Downers Grove, IL) hybridizes to both arms and the centromere of chromosome 5.

Hybridization of these probes to parathyroid adenomas metaphase spreads (according to the manufacturer's instructions) will reveal the presence of a translocation between chromosome 5 and chromosome 20, in the form of a two-color chromosome. In order to evaluate other rearrangements, spectral karyotyping (SKY) or multi-fluor FISH (Speicher et al., supra; Schrock et al., Science 273: 494 (1996) ) may be carried out. In these methods, a probe "cocktail"with every chromosome labeled with a unique combination of dyes is hybridized to metaphase preparations. Following washing and detection steps, the hybridized and counterstained preparations are imaged on a spectral imaging workstation (for SKY), which determines the spectrum (and hence the dye combination) at each point in the image, allowing unambiguous identification of complex chromosome rearrangements. Thus, any rearrangements involving chromosome 5 (and tramdL sequences) and chromosome 20 (and STK4/Mst-l/Krs-2 sequences) can be identified.

Although an understanding of the mechanism underlying the invention is not necessary to the practice of the invention, it is hypothesized that the STK4/Mst-l/Krs-2-tramdL fusion may be involved in the proliferation of parathyroid tumors, including but not limited to parathyroid adenomas. In order to investigate this role and create an animal model which can be used to investigate therapeutic regimens for parathyroid tumors harboring a STK4/Mst-l/Krs-2- tramdL fusion, transgenic mice in which the STK4/Mst-1/Krs-2-tramdL fusion construct (Figure 26) (SEQ ID NO : 38) is expressed under control of a promoter that is active in the parathyroid gland are contemplated. Gcm2 is a transcription factor that is expressed primarily, and perhaps exclusively, in parathyroid tissue [Kim et al., PNAS USA 95: 12364 (1998) ]. Mice will be generated in which the Gcm2 promoter controls expression of the STK4/Mst-l/Krs-2- tramdL fusion gene. These mice will be generated by"knock-in"targeted recombination in embryonic stem cells (ES cells).

The STK4/Mst-l/Krs-2-tramdL fusion protein (SEQ ID NO : 44) will be expressed in mice using 5.2kb of the human Pth promoter region. When overexpressed in parathyroid gland using this DNA fragment, the cyclin D 1 gene causes primary hyperparathyroidism in mice (Imanishi, Y. , Hosokawa, Y. , Yoshimoto, K. , Schipani, E. , Mallya, S. , Papanikolaou, A., Kifor, O., Tokura, T. , Sablosky, M. , Ledgard, F, Gronowicz, G, Wang, T. C. , Schmidt, E. V. , Hall, C., Brown, E. M. , Bronson, R. , and Arnold, A. 2001. Primary hyperparathyroidism caused by parathyroid-targeted overexpression of cyclin D1 in transgenic mice. Journal of Clinical Investigation 107 : 1093-1102.) Alternatively, the Gcm2 coding regions will is replaced with the STK4/Mst-l/Krs-2- tramdL fusion gene. The Gcm2 gene has been inactivated in mice [Gunther et al., Nature 406: 199-203, (2000) ], indicating that mice that carry a single knock-in allele will not be affected by the absence of a single Gcm2 allele. In an alternative embodiment, a"knock-in"of the STK4/Mst-l/Krs-2 into the parathyroid hormone locus is contemplated.

Parathyroid-specific expression of the STK4/Mst-l/Krs-2-tramdL fusion is expected to result in hyperparathyroidism (which can be detected by parathyroid enlargement and increased serum levels of calcium and parathyroid hormone), if, as hypothesized, the STK4/Mst-l/Krs-2- tramdL fusion plays a role in parathyroid tumors. Such mice can then be used in evaluation of therapeutics which may be useful in the management of parathyroid tumors harboring the STK4/Mst-l/Krs-2-tramdL fusion in human subjects VI. TRAMDORINS AND PERIPHERAL NEUROPATHIES As noted above, the human tramdorins map to chromosome 5q31-33. An autosomal recessive Charcot-Marie-Tooth Syndrome demyelinating neuropathy has been mapped to this region [Guilbot et al., Ann NYAcad Sci 883: 56 (1999a); Guilbot et al., Eur JHum Genet 7: 849 (1999b); LeGuern et al., Hum Mol Genet 5: 1685 (1996) ), suggesting that a human tramdorin could be a candidate peripheral neuropathy disease gene. As noted above, a crucial function of Oct-6 in myelination may be activating expression tramdorin 1, a gene that encodes a novel transmembrane domain protein. Of the six target genes for Oct-6 identified thus far, it is the only one that resides near a candidate peripheral neuropathy locus. A search for mutation in families that segregate the chromosome 5q32 peripheral neuropathy (Guilbot et al., Ann NY

Acad Sci 883 : 56 (1999a); Guilbot et al., Eur JHum Genet 7: 849 (1999b); LeGuern et al., Hum Mol Genet 5: 1685 (1996) ) is underway. Thus, by focusing on the tramdorins, the target area to look for disease-associated mutations is significantly reduced. If a consistent disease-associated mutation in a tramdorin gene can be identified, that gene can be used in a molecular diagnostic assay, and may also contribute to potential therapies. For example, the identification of the disease-associated mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene enabled the development of a diagnostic genetic test, an animal model, and potential gene therapies (see OMIM entry *602421, http ://www3. ncbi. nlm. nih. gov/htbin- post/Omim/dispmim ? 602421 and Chapter 3 of"Human Genetics, A Problem-based Approach", Bruce Korf, Blackwell Science, 1996 for a summary), and a similar outcome is expected should a peripheral neuropathy-associated mutation in a tramdorin be identified.

VII. REMYELINATION BY SCHWANN CELLS IN THE CNS FOLLOWING DISEASE OR INJURY Remyelination of axons in the central nervous system (CNS) by Schwann cells has been explored extensively as a possible treatment for demyelinating diseases such as multiple sclerosis, and as a treatment for nerve injury (reviewed in (Duncan et al., JAnat 190: 35 (1997); Scolding, Philos Trans R Soc Lond B Biol Sci 354 : 1711 (1999); Scolding et al., Baillieres Clin Neurol 6 : 525 (1997) ). MS affects 1/1000 Americans, and in people of northern European descent, the frequency is 1/800 (Lynch et al., Dis Mon 42 : 1-55 (1996) ) ; it represents a significant public health problem in the United States and Europe. An understanding of the genes that regulate myelination by Schwann cells is crucial for optimizing their use in remyelination in the CNS. The tramdorin gene may play a crucial role in regulating myelination for the following reasons: First, it is a putative target gene for Oct-6, a transcription factor that is essential for timely peripheral myelination (Bermingham et al., Gene Develop 10: 1751 (1996); Jaegle et al., Science 273: 507 (1996) ). Second, its expression following nerve injury indicates that it encodes a myelin-related protein. Tramdorin, and/or its putative ligands, may provide avenues for the development of therapeutic agents for the treatment of the effects of demyelination or nerve injury, both in the peripheral and central nervous systems. The ability to identify molecules that promote myelination will significantly

enhance the prospects for successfully using Schwann cells, and perhaps oligodendrocytes and olfactory ensheathing cells as well, to mitigate the damage caused by MS and other demyelinating diseases. A significant use of tramdorin will be in identifying such molecules.

VIII. USE OF TRAMDORIN TO RELIEVE NEUROGENIC AND NEUROPATHIC PAIN Pain that arises from the nervous system is termed"neurogenic."Peripheral neurogenic pain may follow transient pressure upon or stretching of a peripheral nerve or root, or reflect sustained damage to a nerve ("neuropathic pain") such as in polyneuropathy, entrapment neuropathy, or after herpes zoster. Neurogenic pain may have a central origin such as stroke, multiple sclerosis, or trauma, especially of the spinal cord.

Correct diagnosis allows tailoring treatment to the pathophysiologic mechanisms that trigger and maintain the painful condition. Assessing the pain location, intensity, quality, time course, precipitating and relieving factors, as well as its impact on physical and psychosocial function is the first step in clinical analysis. Diagnosis depends upon, first, the neuroanatomical distribution of the pain and, second, evidence of sensory dysfunction involving a peripheral nerve, plexus, nerve root or central pathway. If the affected nerve or pathway is mixed motor and sensory, then weakness, muscle atrophy, or reflex abnormalities may be additional clues to neural involvement. The diagnosis may be obvious but sometimes a thorough neurological examination is needed to uncover the neurogenic origin of the pain. A pain drawing made by the patient frequently gives a good indication of the neuroanatomic distribution and quality of the pain.

Impaired sensation is often evident during a careful examination. Sensory dysfunction may be manifested as hypo-and/or hyperesthesia for one or more modalities, increasing pain to normally painful stimuli (hyperalgesia) or pain due to normally nonpainful stimuli (allodynia).

Temporal and spatial sensory dysfunction are also common. Abnormal sensory function in neurogenic pain states can, regardless of modality, be described in terms of stimulus strength and sensation magnitude. Hypoesthesia consists of both increased perception threshold and reduced sensation magnitude at suprathreshold stimulus strengths. Occasionally, hypoesthesia occurs without an increase in stimulus detection threshold. Elevated stimulus perception

threshold, together with an increase in slope of the magnitude/stimulus relation is typical for the combination of hypo-and hyperesthesia often seen in neurogenic pain states. Evoked sensation hyperpathic syndrome often has a paresthetic or dysesthetic character or is frankly painful instead of the normal sensation evoked by the applied stimulus. Hyperesthesia can also occur separately with a steeper slope, and occasionally (thin line), with a lowered threshold.

Tramdorinl/mPAT2 has been shown to function as a proton-dependent transporter of small amino acids. (See, Boll, M. , Foltz, M. , Rubio-Aliaga, I., Kottra, G. , and Daniel, H.

(2002). Functional Characterization of Two Novel Mammalian Electrogenic Proton-dependent Amino Acid Cotransporters. J Biol Chem 277,22966-73.). It is related to yeast vacuolar and rat lysosomal amino acid transport proteins, but unlike tramdorin3/LYAAT-1, tramdorinl does not appear to be a lysosomal protein. Tramdorinl lacks a putative lysosomal targeting motif (an acidic residue located in the C-terminus,-4 to-6 relative to a dileucine, (LL (I, M, V); that is present in tramd3/LYAAT-1 consistent with its failure to co-localize with LAMpl. [see, Sandoval, I. V. , Martinez-Arca, S. , Valdueza, J. , Palacios, S. , and Holman, G. D.. J Biol Chem 275, 39874-85. (2000). ].

Tramdorinl/mPAT2 shows strong specificity for glycine, L-alanine and L-proline, with greatest inward currents generated by glycine. Although it is possible tramdorinl could provide additional amino acids required for metabolism in the paranodes and incisures, its restricted substrate specificity and expression pattern suggest other function (s). Glycine can act as an inhibitory neurotransmitter in the CNS, and as an agonist of the NMDA class of ionotropic glutamate receptors. Glycine receptors have not been reported in the PNS, but NMDA glutamate receptors have been found on both adult Schwann cells and peripheral axons (Coggeshall and Carlton, 1998; Fink et al. , 1999; Kinkelin et al. , 2000).

Therefore, in one embodiment, tramdorinl could inhibit glutaminergic signaling in peripheral nerve by sequestering extracellular glycine. Indeed it has been shown that axonal expression of all three types of ionotropic glutamate receptor increases following peripheral nerve inflammation; these receptors could contribute to the spontaneous discharges that have been observed in rat models of nerve injury and inflammatory pain. Additionally, an increased frequency of spontaneous discharges, with concomitant thermal hyperalgesia and mechanical allodynia, have been observed in saphenous nerves from dysmyelinatingprx mutant mice. See,

Gillespie, C. S. , Sherman, D. L. , Fleetwood-Walker, S. M., Cottrell, D. F., Tait, S. , Garry, E.

M. , Wallace, V. C. , Ure, J. , Griffiths, I. R. , Smith, A. , and Brophy, P. J Peripheral demyelination and neuropathic pain behavior in periaxin-deficient mice. Neuron 26, 523-31 (2001).

These mice possess altered Schmidt-Lanterman incisures, and therefore their expression of tramdorinl may be reduced. In selected embodiments, tramdorinl may dampen neuronal excitability thereby limiting pain sensitivity. In other embodiments, tramdorin may modulate NMDA glutamate receptor signaling between Schwann cells, or between adjacent membranes of a single Schwann cell.

In one embodiment, it is contemplated that tramdorins may be to a patient suffering at least one symptom of neurogenic and/or neuropathic pain. In another embodiment, said administration is by direct injection. Once again, however, Applicants note that a the present invention is not limited by tramdorins'precise mechanism of action or any specific mode of administration.

IX. TRAMDORIN LOCALIZATION USING ANTISERUM To characterize tramdorinl, an antiserum was raised against a sequence near the N- terminus that was divergent among the tramdorin gene family (e. g. antisera to a synthetic peptide: ESAKKLQSQDPSPANGTSC (SEQ ID NO: 74), containing amino acids 22-39 near the N-terminus of tramdorinl, as described in the section above.

In immunoblots of adult rat sciatic nerve, the antiserum binds to a single band close to the predicted molecular weight of unglycosylated tramdorinl (panel A of Fig. 29). A band of similar molecular weight is observed on immunoblots of cells transfected with full-length mouse tramdorinl cDNA (panel B of Fig. 29). Transfected cells were stained with the anti- tramdorinl antiserum, whereas parental cells were unstained (panel C of Figure 29). To localize tramdorinl, unfixed myelinated fibers from rat sciatic nerves were labeled with this antiserum.

Tramdorinl was localized in incisures and the paranodes, as shown by co-labeling with a monoclonal antibody against myelin associated glycoprotein (MAG; panels A-C of Figure 30).

To determine if tramdorinl, like tramdorin3/LYAAT-l, is associated with lysosomes, teased fibers were double-labeled for tramdorinl and LAMP1, a lysosomal marker. Panels D-F of

figure 30 shows that tramdorin and LAMP1 are found in paranodes, but do not co-localize.

While it is not intended that the present invention be limited to any particular motif of localization or a particular mechanism of action, this immunostaining suggest that tramdorinl is localized to non-compact myelin, but is not a component of lysosomes.

Again, while it is not intended the present invention be limited to any specific mechanism, Tramdorinl/mPAT2 has been shown to function as a proton-dependent transporter of small amino acid. See, Boll, M. , Foltz, M. , Rubio-Aliaga, I., Kottra, G. , and Daniel, H.

(2002). Functional Characterization of Two Novel Mammalian Electrogenic Proton-dependent Amino Acid Cotransporters. J Biol Chem 277,22966-73. It is related to yeast vacuolar and rat lysosomal amino acid transport proteins, but unlike tramdorin3/LYAAT-1, tramdorinl does not appear to be a lysosomal protein. Tramdorinl lacks a putative lysosomal targeting motif (an acidic residue located in the C-terminus,-4 to-6 relative to a dileucine, (LL (I, M, V) that is present in tramd3/LYAAT-l. See, Sandoval, I. V. , Martinez-Arca, S. , Valdueza, J. , Palacios, S. , and Holman, G. D. (2000). Distinct reading of different structural determinants modulates the dileucine-mediated transport steps of the lysosomal membrane protein LIMPII and the insulin-sensitive glucose transporter GLUT4. J Biol Chem 275, 39874-85. Tramdorinl/mPAT2 shows strong specificity for glycine, L-alanine and L-proline, with greatest inward currents generated by glycine (See, Boll supra). This restricted substrate specificity and localization suggest that glycine and glycine transport have a role in myelinating Schwann cells and modulating physiologies of the nervous system.

EXPERIMENTAL The following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.

In the experimental disclosure which follows, the following abbreviations apply: eq (equivalents); M (Molar); tiM (micromolar); N (Normal); mol (moles); mmol (millimoles); , umol (micromoles); nmol (nanomoles) ; g (grams); mg (milligrams) ; ug (micrograms); L (liters); ml (milliliters) ; jj. l (microliters); cm (centimeters); mm (millimeters) ; um (micrometers); nm (nanometers) ; °C (degrees Centigrade); RDA (representational difference analysis); nts (nucleotides).

Example 1 This example describes the identification of the cDNA for mouse tramdorin 1.

RDA on sciatic nerve from Oct-6 (-/-) and (+/+) sciatic nerves was performed as described (Bermingham et al., JNeurosci Res 63: 516 (2001); Erkman et al., Eur JPharmacol 393: 97 (2000) ). The RDA procedure was performed essentially as described elsewhere (Hubank et al., Nucleic Acids Res 22: 5640 (1994); Lisitsyn et al., Science 259: 946 (1995); Adman et al., Biochem J 323 : 113 (1997) ), with the following modifications: Carrier mRNA (Bermingham et al., manuscript in preparation) was added to the total RNA for most experiments, prior to the isolation of poly-A+ RNA and subsequent cDNA synthesis. cDNAs were digested with DpnII, which cleaves at the recognition site GATC; because the four base 5'overhang that results from its cleavage is identical to that generated by BglII, the J, N, and R oligonucleotides that were designed by (Lisitsyn et al., Methods Enzymol 254: 291 (1995) ) for use on genomic RDA with BgllI were used here to amplify Dpn-II generated fragments. The"N"adaptors were ligated to the DpnII ends. Those fragments with two appropriately spaced adaptors were amplified by two rounds of PCR. Typically 50-600 bp fragments were amplified. A small proportion of each of the drivers was digested with DpnII to remove the"N"adaptors, after which they were replaced with"R"adaptors. Each"tester"DNA was mixed with an excess of driver that was derived from the other source, denatured, and permitted to anneal. The reannealed DNA was amplified by PCR, digested with mung bean nuclease (New England Biolabs, Beverly, MA) to remove the single strands that result from priming at only one end of a template, then amplified again by PCR. DNA was digested with DpnlI to remove the"R"adaptors, after which they were replaced with"J"adaptors. This material was used as the tester for a second round of subtraction, after mixing with an excess of driver. The products of the second round of subtraction were subjected to two rounds of PCR amplification to produce the second difference products, DP2. The linkers were removed from the DP2 DNA, after which it was cloned. DpnII fragments were ligated into the BamHI site of pBKSIv (Stratagene, La Jolla, CA). Ligations were transfected into DH5a (Gibco-BRL, Grand Island, NY).

Because Oct-6 is transiently expressed in sciatic nerve, these experiments were carried out on nerves that were isolated at PO (day of birth), at or near the peak of Oct-6 expression.

Six RDA cDNA clones corresponded to genes that are downregulated in the absence of Oct-6,

and therefore are putatively activated by Oct-6 in sciatic nerve (Bermingham et al., manuscript in preparation). The differential abundance of these clones in cDNA from wild-type and Oct-6 mutant sciatic nerves was confirmed by hybridization to Southern blots of PCR-amplified cDNA ("Snorthern blots"). These blots are reliable indicators of differential expression (Bermingham et al., JNeurosci Res 63: 516 (2001); Bermingham et al., manuscript in preparation). As described in Bermingham et al., JNeurosci Res 63: 516 (2001), Snorthern blots use cDNA that was amplified by PCR for use as driver in the RDA experiments described above. PCR reactions were optimized to ensure that amplification was exponential, and parallel PCR reactions were pooled to minimize variation from individual samples and to obtain sufficient material. Driver DNA (0.5 or 1 ug) was electrophoresed through 3% NuSieve 3: 1 agarose (FMC) or 4% acrylamide gels, then transferred by conventional Southern blotting or electroblotting onto Hybond N+ membranes (Amersham, Arlington Heights, IL).

The results for one clone, 125, is shown in Figure 1A. RDA clone JBSN125 contains a cDNA fragment that corresponds to a putative Oct-6 regulated gene The panels show Southern blots of PCR-amplified cDNA ("Snorthern blots") from Oct-6 (+/+) (wild-type) and Oct-6 (-/-) (mutant) sciatic nerves. The left-hand panel shows a Snorthern blot that has been hybridized to RDA clone JBSN125; it indicates that one of two inserts in this clone corresponds to a gene that requires Oct-6, directly or indirectly, for normal activation. The larger insert is not differentially expressed, and serves as an internal control. A subclone that contains only the smaller insert, JBSN 125-10, also is differentially expressed (data not shown). The center and right panels are control blots that have been hybridized with JBSN11, a lacZ-containing clone, and Oct-6. No putative Oct-6 repressed genes were isolated, except for LacZ and Neo, which are expressed in the knockout mice. As expected, Oct-6 is expressed only in Oct-6 (+/+) sciatic nerves. The sizes of the fragments on the blots reflect the presence of 24bp linkers on the ends of the fragments. The differentially expressed subclone, JBSN 125-10, was used to isolate additional sequence for this gene.

Mouse cDNAs that correspond to clone 125-10 were isolated by obtaining and sequencing three homologous EST clones 1920302 (AI786604), 1380757 (AI035402), and 1363993 (AI005767) (Figure 1B). Figure 1B shows cDNAs that correspond to RDA clone JBSN125-10. Seven cDNA fragments, from two species (rat and mouse) are shown; they

include the RDA fragment JBSN125-10, two independent, nonidentical 5'RACE rat cDNAs, a 3'RACE rat cDNA, and three mouse EST cDNAs. Of these, the 2.45kb mouse EST cDNA 1920302 (SEQ ID NO : 13) appears to include the entire open reading frame, which is depicted in black. The organism and tissue of origin of each cDNA fragment is listed. The open reading frame commences with an ATG initiation codon that matches the Kozak consensus (Kozak, Nucleic Acids Res 15: 8125 (1987)). EST clone 1380757 (SEQ ID NO : 50) contains a polyA tract just following the stop codon; because no consensus polyadenylation signal exists in the cDNA near this site, this clone probably represents an artifactual polyadenylation event. EST cDNA clones 1920302 (SEQ ID NO : 13) and 1363993 (SEQ ID NO : 43) diverge at the 5'splice site at the 3'end of exon 7; comparison with genomic sequences indicates that EST cDNA 1363993 is unspliced. The intron-derived reading frame contains only three amino acids before a stop codon. Mouse EST 1363993 (AI005767) (SEQ ID NO : 43) thus appears to correspond to a partially spliced transcript.

The full-length mouse cDNA contains a 1.4 kb open reading frame (Figure 2) that encodes a protein of 52kd. The mouse cDNA and the putative protein that it encodes do not match any genes in the GenBank non-redundant database, or the SwissProt database.

Therefore, this cDNA is derived from a novel gene. Figure 2 also shows a putative rat tramdorin 1 cDNA, derived from 5'and 3'RACE sequences (see Example 2). Figure 2 shows the translated proteins; the mouse cDNA appears to encode a 478 amino acid protein, while the corresponding rat protein is 481 amino acids. Exon boundaries are superimposed on the mouse cDNA sequence. The boundaries of exons 8, 9 and 10 are defined by comparison to Ensembl mouse genomic sequence files, while the remainder are defined by sequencing of 129 mouse genomic DNA. The polyA tract of EST cDNA 1380757 (SEQ ID NO : 50) commences at the A at position 1603. A putative polyadenylation site is shown in bold; such AGUAAA (SEQ ID NO : 51) polyadenylation signals are processed in vitro at roughly 30% of the efficiency of the canonical AAUAAA (SEQ ID NO : 52) signal (Sheets et al., Nucleic Acids Res 18: 5799 (1990)).

The Niait restriction sites (CATG) that flank RDA clone 125-10 are shown in italics.

Example 2 This example shows the identification of a rat tramdorin 1 cDNA.

Rat cDNAs that correspond to clone 125-10 were isolated by 5'and 3'RACE (rapid isolation of cDNA ends (Frohman, Methods Enzymol 218 : 340 (1993); Frohman, PCR Methods Appl 4 : S40 (1994) ). RNA was isolated from sciatic nerves taken from one to two day old rat pups. First strand cDNA was synthesized using Superscript II reverse transcriptase (Gibco-BRL). RACE was performed using a kit from Clontech, according to the manufacturer's instructions. The 3' rat tramdl RACE primer has the following sequence: 5'GACTTCCCCTGGCTGTGAAGAATGCGGGC 3' (SEQ ID NO : 53) and the 5'rat tramdl RACE primer has the following sequence: 5'TcAGTCTGTGACAGAAGCGCTGGGCACATCtG 3' (SEQ ID NO : 54) The lower case nucleotides represent mismatches between the 5'RACE sequence and the final rat sequence. The final rat sequence has G's replacing the lower case letters in the 5' primer.

The 5'and 3'rat RACE fragments combined define a 2. 5kb cDNA (SEQ ID NO : 55).

The full-length rat cDNA contains a 1.4 kb open reading frame (SEQ ID NO : 6) (Figure 2) that encodes a protein of 52kd (SEQ ID NO : 22). The rat cDNA and the protein that it encodes does not match any genes in the GenBank non-redundant database, or the SwissProt database.

Therefore, this cDNA is derived from a novel gene. The sequence of a rat tramdorinl cDNA, derived from 5'and 3'RACE sequences is shown in Figure 2 (SEQ ID NO : 6), as is the translated protein. The rat protein is 481 amino acids.

Example 3 This example describes structural predictions for the protein encoded by mouse tramdorin 1 and related tramdorins in humans.

The protein encoded by mouse cDNA 1920302 (SEQ ID NO : 13) was predicted by several protein structure prediction programs to encode a transmembrane protein. These programs use aspects of protein structure such as hydropathy and charge distributions and/or utilize different algorithms to arrive at their predictions. Although all the programs used predicted the protein would be a polytopic transmembrane protein, they differed in the number of transmembrane domains predicted. PHDHtm (Rost et al., Protein Sci 4: 521 (1995) ) and TM finder (Deber et al., Protein Sci 10 : 212 (2001) ) predicted 8 transmembrane domains (Figure

3A and data not shown). PRED-TMR (Pasquier et al., Protein Eng 12: 381 (1999) ) predicted the presence of 10 putative transmembrane domains, and an Argos hydropathy plot (Argos et al., Eur JBiochem 128: 565 (1982) ) predicted 10 hydrophobic domains with a possible 11 (data not shown). Memsat2 (PSIPRED ; McGuffin et al., Bioinformatics 16: 404 (2000) ), and TMHMM (Sonnhammer et al., Proc Int Conflratell Syst Mol Biol 6: 175 (1998)) predict that the protein contains 11 transmembrane helices with the N-terminus facing the cytoplasm (Figure 3). Regardless of the differing predictions about the number of transmembrane domains, or their topology, this protein appears to contain multiple transmembrane helices. Therefore we have named the putative gene product tramdorin, for transmembrane domain rich protein.

Tramdorin has been assigned the human and mouse gene symbol tramdl.

The putative amino acid sequence encoded by the putative full length EST cDNA 1920302 (SEQ ID NO : 13) is shown in Figure 3A. The amino acid sequence of EST cDNA 1920302 (SEQ ID NO : 14) was analyzed using the transmembrane domain prediction programs PHDHtm (Rost et al., Protein Sci 4: 521 (1995) ), Memsat2 (McGuffin et al., Bioinformatics 16: 404 (2000) ), and TMHMM (Sonnhammer et al., Proc Int Conf Intell Syst Mol Biol 6 : 175 (1998) ). 11 putative transmembrane domains are numbered. Transmembrane domains predicted by PHDHtm and TMHMM are marked with Capital T's. For Memsat2, transmembrane domains are marked as follows: O : outside transmembrane helix cap; X: central transmembrane helix segment; I: inside transmembrane helix cap. For both Memsat2 and TMHMM, predicted cytoplasmic domains are marked by"+", while non-cytoplasmic domains are marked by"-". The partially spliced EST cDNA 1363993 (SEQ ID NO : 43) is truncated following putative transmembrane domain #6. Three consensus glycosylation sites within the protein sequence are shown in bold. Figure 3B shows a diagram depicting the predicted topology of tramdorinl with 11 transmembrane domains, as predicted by Memsat2 and TMHMM. The three extracellular/lumenal glycosylation sites are marked with branched structures. While it is not intended that the present invention be limited to any particular level of gylcosylation (if any) the electrophoretic mobility of tramdorin suggests that it is not glycosylated extensively. Moreover, glycosylation is shown in black at only one site, in accordance with the observation that in transmembrane domain proteins with multiple

glycosylation sites, only one is typically used (Landolt-Marticorena et al., Biocllem J302 : 253 (1994) ); the others are shown in gray.

While a precise understanding of the structure of tramdorin is not required for the practice of the present invention, the following considerations suggest that tramd proteins consist of 11 transmembrane domains, with the N-terminus facing the cytoplasm. However, the invention is not to be limited to such a structural prediction of tramdorin proteins. First, those programs that predicted 11 transmembrane domain helices more accurately predicted the number and topology of the transmembrane helices of a set of transmembrane proteins of known structure (Tusnady et al., J Chem Inf Comput Sci 41: 364 2001). Therefore those programs also may be better at predicting the structure of unknown proteins. Although PHDHtm, which predicted 8 transmembrane domains, has been shown to be up to 90% accurate in predicting transmembrane helices, the predicted helices often were too long.

Consistent with this observation, the first, third and seventh transmembrane domains predicted by PHDHtm are much longer than typical transmembrane domains (23-30 amino acids; reviewed in (von Heijne, Prog Biophys Mol Biol 66: 113 (1996) ), and are predicted to be two transmembrane domains by memsat2 and TMHMM. Second, analysis of human tramdl,-2, and-3 using Memsat2 and TMHMM produced 11 transmembrane predictions similar to those shown for mouse tramdl in Figure 3, except that TMHMM did not predict the first transmembrane domain for human tramdorin 1. Third, if the protein were to possess 10 transmembrane domains, analysis of charged amino acids flanking the first putative transmembrane domain (as predicted by either PHDHtm or PRED-TMR) suggests that the N- terminus will be outside the cytoplasm (Hartmann et al., PNAS USA 86: 5786 (1989)). Such a topology could arise from cleavage of an N-terminal signal sequence. However, a signal sequence prediction program (Nielsen et al., Protein Eng 10: 1 (1997) ) indicates that the protein does not appear to contain a cleaved N-terminal signal sequence. Fourth, membrane proteins are glycosylated only on their extracytoplamic faces, and the distribution of glycosylation consensus sites (NXS/T, [Asn Xaa Ser/Thr] (SEQ ID NO : 56) wherein X [Xaa] is any amino acid except proline) supports the Memsat2 and TMHMM topology. The mouse tramdl protein sequence contains five putative glycosylation sites. Of these, an N-terminal NGT [Asn Gly Thr] sequence is not conserved in human, and an NIS [Asn Ile Ser] sequence is buried in

predicted transmembrane helix 5. The three conserved, non-transmembrane domain consensus glycosylation sites are predicted to be extracytoplasmic in the structures predicted by Memsat2 and TMHMM. Thus the preponderance of evidence from protein structure prediction programs suggests that the protein contains 11 transmembrane domains, with the N-terminus facing the cytoplasm (Figure 3B).

However, as noted above, the invention is not limited to any particular tramdorin structure, and several considerations suggest alternative structures for tramdorins. The 11 transmembrane model for tramdorin structure. Proteins with 11 transmembrane domains are very rare (Jones, FEBS Lett 423: 281 (1998) ). Furthermore, the closest known relatives of tramdorins, the vesicular GABA transporters, are thought to consist of 10 transmembrane domains; they correspond to predicted transmembrane domains 2-11 of tramdorinl. For the C. elegans vesicular GABA transporter AF031935.1, the memsat2 program predicted 10 transmembrane domains, but the orientation in the membrane was opposite that shown in (Mchitire et al., Nature 389: 870 (1997) ), while TMHMM predicted 9 transmembrane domains (data not shown). These observations raise the possibility that current transmembrane domain structure prediction programs may have difficulty predicting correctly the structure of vesicular GABA transporters, and may have difficulty with tramdorins as well. Thus, the theoretical prediction that tramdorins consist of 11 transmembrane domain proteins with cytoplasmic N- termini should not be considered limiting for the purposes of the present invention cDNAs that encode truncated tramdorin proteins have been identified for both human and mouse tramdl, and for tramd3. In the case of mouse and human tramdl, the cDNAs that encode truncated proteins arise from the failure to splice out the intron that follows exon 7.

This intron is unusual in that it possesses 3 regions of homology between mouse and human, clustered at its 5'end (Figure 18). While not to be limited to any particular mechanism, if these conserved regions of homology are related to the splicing of exon 7, they may indicate a biological role for the truncated proteins. Such proteins would contain the first six putative transmembrane domains.

Example 4 This example is directed to the expression pattern of tramdorin 1 in the mouse.

To determine the size of tramdorin mRNA (s) and to determine if tramdorin expression is restricted to sciatic nerve, a northern blot with RNAs from adult mouse sciatic nerve and other tissues was hybridized to a radiolabelled probe synthesized from tramdorin cDNA 1920302 (SEQ ID NO : 13) (See, Figure 4). Specifically, Each lane contains an equal amount (10 pg) of total RNA isolated from the indicated mouse tissues. The blot was successively hybridized to a radiolabeled probe corresponding to tramdorinl, and exposed to film for 14 days after the hybridization. The sizes of the transcripts are estimated according to the sizes of 28S and 18S rRNAs (4712nt and 1869nt, respectively; (Hassouna et al. , 1984; Raynal et al. , 1984).

The probe hybridized strongly to two distinct transcript classes in sciatic nerve, of approximately 2kb and 2.8 kb in size. Of these, hybridization was strongest to the 2kb transcripts. The sizes of the transcripts were based on the sizes of 28S and 18S rRNAs (4712nt and 1869nt, respectively; (Hassouna et al., Nucleic Acids Res 12: 3563 (1984); Raynal et al., FEBS Lett 167: 263 (1984)). No expression was observed in spleen, and only a very faint band was seen at >6. 5kb in cerebrum, indicating that tramdorin 1 is not ubiquitously expressed. The 2.8kb species is the most prominent tramdorin 1 message in lung, adrenal gland and thymus.

This result indicates that EST cDNA 1920302 (SEQ ID NO : 13) defines a nearly full length tramdl message, and must correspond to the larger transcript class, because it is approximately 2.45kb, excluding the polyA tail. Therefore tramdorinl expression is neither ubiquitous, nor restricted to sciatic nerve. In contrast, a similar northern using rat tissue did not show hybridization to lung and thymus (data not shown), suggesting either variation in tramdorin expression among species, or cross-hybridization to related sequences.

Whether the expression of tramdl in sciatic nerve is modulated by nerve injury was of interest. If so, its pattern of expression would determine if the genes are expressed in myelinating or non-myelinating/immature Schwann cells. Axotomy leads to a sharp decline in the steady state mRNA levels for myelin-related genes, and an increase in the expression of genes associated with immature Schwann cells. Subsequently, in crushed nerves, Schwann cells re-express myelin-related genes in response to contact with regenerating axons (reviewed in Poduslo,"Regulation of myelin gene expression in the peripheral nervous system."In Dyck, P. J. et al. (eds), Peripheral Neuropathy, W. B. Saunders Co. , Philadelphia, pp. 282-289 (1993)).

A northern blot of RNA from sciatic nerves collected at various times following crush or transection injury was hybridized with a mouse tramdl probe (Figure 5). It should be noted that the mouse tramdl probe also hybridized with a northern blot of RNA from rat nerve tissue.

Adult Sprague Dawley rats were anesthetized with 50 mg/kg pentobarbitol i. p. , and the sciatic nerves were exposed at the obturator tendon. To prevent axonal regeneration, nerves were doubly ligated and transected between the ligatures. Nerves were crushed by compression with flattened forceps twice, each time for 10 seconds. Animals were allowed to survive for various periods of time prior to sacrifice by C02 inhalation. For RNA extraction, several millimeters of nerve adjacent to the lesion site were trimmed off, and the distal nerve-stumps were frozen in liquid nitrogen. Where indicated, the distal stumps of crushed nerves were subdivided into proximal and distal segments of equal lengths. Unlesioned nerves were taken from animals of various ages. Total RNA was isolated from the sciatic nerves by CsCl2 gradient centrifugation (Chirgwin et al., Biochem 18 : 5294 (1979) ). Equal samples (10 llg) of RNA were electrophoresed in 1% agarose, 2.2 M formaldehyde gels, transferred to nylon membranes in 6X SSC, and UV cross-linked (0.12 joules). Blots were prehybridized, hybridized, and washed using standard techniques (Sambrook et al., Molecular Cloning : a Laboratory manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, N. Y. [1989] pp. 31). cDNA probes were 32p labeled with specific activities of 2-5 x 109 cpmlllg, prepared by primer extension with random hexamers using the Prim-a-gene kit (Promega) according to the manufacturer's instructions.

Northern blots of RNA from segments of sciatic nerve distal to a transection injury (in which axons do not regenerate) and crush injury (in which axons regenerate and are remyelinated) were hybridized with probes for tramdorin, and Oct-6. The blots were hybridized also to probes for PO, p75 nerve growth factor receptor (NGFR) and GAPDH as controls. For the crushed nerves, RNA was made from two sciatic nerve segments, one immediately distal to the injury (P) and one more distal to the injury (D) (Figure 5). Myelination occurs more slowly in the (D) segments because contact with regenerating axons is delayed.

Tramdl expression is downregulated by transection injury, and remains low. Following a crush injury, tramdl is downregulated, but subsequently it is reexpressed as axons regenerate and are remyelinated (Figure 5). Therefore, the tramdorinl gene encodes a myelin-related protein. Several genes that are required for normal myelination show this pattern of expression;

these include Oct-6, PO, connexin32 and PMP-22. (Poduslo,"Regulation of myelin gene expression in the peripheral nervous system."In Dyck, P. J. et al. (eds), Peripheral Neuropathy, W. B. Saunders Co. , Philadelphia, pp. 282-289 (1993); Scherer et al., JNeurosci 14 : 1930 (1994); Sohl et al., Eur J Cell Biol 69 : 267 (1996) ). This result suggests that tramdl may play an important role in the timely formation of peripheral myelin.

Example 5 This example describes experiments to determine the mouse chromosomal location of tramdl.

The mouse chromosomal location of Tramdl was determined by interspecific backcross analysis using progeny derived from matings of [ (C57BL/6J x Mus spretus) F1 X C57BL/6J] mice. This interspecific backcross mapping panel has been typed for over 3100 loci that are well distributed among all the autosomes as well as the X chromosome (Copeland et al., Trends Genet 7: 113 (1991)). C57BL/6J and M. spretus DNAs were digested with several enzymes and analyzed by Southern blot hybridization for informative restriction fragment length polymorphisms (RFLPs) using a mouse cDNA probe specific for Tramdl. The 7.5 kb BglI M. spretus RFLP (see below) was used to follow the segregation of the Tramdl locus in backcross mice. The mapping results indicated that Tramdl is located in the proximal region of mouse chromosome 11 linked to 1113 and Handl.

Figure 6A shows a summary of the mapping results. Each column represents the chromosome identified in the backcross progeny that was inherited from the (C57BL/6J x M. spretus) F1 parent. The shaded boxes represent the presence of a C57BL/6J allele and white boxes represent the presence of a M. spretus allele. The number of offspring inheriting each type of chromosome is listed at the bottom of each column. A partial chromosome 11 linkage map showing the location of Tramdl in relation to linked genes is shown in Figure 6B.

Recombination distances between loci in centimorgans are shown to the left of the chromosome and the positions of loci in human chromosomes, where known, are shown to the right.

References for the human map positions of loci cited in this study can be obtained from GDB (Genome Data Base), a computerized database of human linkage information maintained by The William H. Welch Medical Library of The Johns Hopkins University (Baltimore, MD).

Although 136 mice were analyzed for every marker and are shown in the segregation analysis (Figure 6A), up to 179 mice were typed for some pairs of markers. Each locus was analyzed in pairwise combinations for recombination frequencies using the additional data. The ratios of the total number of mice exhibiting recombinant chromosomes to the total number of mice analyzed for each pair of loci and the most likely gene order are: centromere-I113-1/142 - Tramdl-6/179-Handl. The recombination frequencies (expressed as genetic distances in centiMorgans (cM) the standard error) are-m3-0. 7 +/-0. 7-Tramdl-3. 4 +/-1.4- Handl. This interspecific map of chromosome 11 has been compared with a composite mouse linkage map that reports the map location of many uncloned mouse mutations (provided from Mouse Genome Database, a computerized database maintained at The Jackson Laboratory, Bar Harbor, ME). Tramdl mapped in a region of the composite map that lacks mouse mutations with a phenotype that might be expected for an alteration in this locus (data not shown).

Interspecific backcross progeny were generated by mating (C57BL/6J x M. spretus) F females and C57BL/6J males as described (Copeland et al., Trends Genet 7: 113 (1991) ). A total of 205 N2 mice were used to map the Tramdl locus, as described above. DNA isolation, restriction enzyme digestion, agarose gel electrophoresis, Southern blot transfer and hybridization were performed essentially as described (Jenkins et aL, J Virol 43: 26 (1982) ). All blots were prepared with Hybond-N+ nylon membrane (Amersham). The probe, a 503 bp BssHII/EcoRI fragment from the 3'UTR of cDNA 1920302, was labeled with [a32P] dCTP using a random primed labeling kit (Stratagene); washing was done to a final stringency of 0.5 X SSCP, 0. 1% SDS, 65°C. A fragment of 10.0 kb was detected in BgII digested C57BL/6J DNA and a fragment of 7.5 kb was detected in BglI digested M. spretus DNA. The presence or absence of the 7.5 kb BglI M. spretus-specific fragment was followed in backcross mice. A description of the probes and RFLPs for the loci linked to Tramdl including 1113 and Handl has been reported previously (Cross et al., Development 121: 2513 (1995); McKenzie et al., J Immunol 150: 5436 (1993) ). Recombination distances were calculated using Map Manager, version 2.6. 5. Gene order was determined by minimizing the number of recombination events required to explain the allele distribution patterns.

The proximal region of mouse chromosome 11 in which tramdl maps, is a region that is syntenic with human chromosome 5q. These results indicate that tramdl is located on human

5q31-33, suggesting that the human homolog of Tramdl will map to 5q31-33 as well. An autosomal recessive Charcot-Marie-Tooth Syndrome demyelinating neuropathy has been mapped to this region (Guilbot et al., Ann NYAcad Sci 883: 56 (1999a); Guilbot et al., Eur J Hum Genet 7: 849 (1999b); LeGuern et al., Hum Mol Genet 5: 1685 (1996) ), suggesting that tramdorinl could be a candidate peripheral neuropathy disease gene.

Example 6 This example describes experiments directed at studying the human tramdorin gene.

As a first step toward determining if tramdorinl mutations are associated with human peripheral neuropathy, the human tramdorin gene was studied. Six human EST cDNAs that possess homology to mouse tramdorinl, I. M. A. G. E numbers 3184556 (BE501426), 1738130 (AI140615), 1837427 (AI208756), 1388139 (AA843982), and 2549054 (AI953890), as well as EST cDNA DKFZp434G1123 from the Deutsches Ressourcezentrum fur Genomforschung GmbH (RZPD) were obtained and sequenced in their entirety. The sequences were aligned on human genomic DNA from the International Human Genome Sequencing Consortium (IHGSC) Homo sapiens chromosome 5 working draft sequence segment Nit 006951. 4 and Celera databases (Lander et al., Nature 409: 860 (2001); Venter et al., Science 291: 1304 (2001)).

These cDNAs showed greatest homology to four discrete locations within 250 kb of chromosome 5q sequence (Figure 7), confirming the mapping of tramdorin in mouse, but in addition, suggesting the existence of multiple tramdorin genes. These genes have been assigned the names tramdl, tramd2, tramd3, and tramdL.

In Figure 7, the IHGSC sequences are shown; coordinates indicate kilobases of nucleotide sequence. The coordinates correspond to scaffold Nit 006951. 4 (1038-1290kb).

Human genomic sequences that are most homologous to mouse tramdorin 1 EST 1920302 reside between 70 and 102kb. Therefore this region, and EST cDNA 3184556, are likely to represent the human homolog of mouse tramdl.

The human tramdl gene has greatest homology to the original mouse tramdl gene, and is flanked on its 3'end by tramd2, and on its 5'end by tramd3, which is transcribed in the opposite orientation to tramdl and tramd2. Nested between tramdl and tramd3 resides a tramdorin gene fragment, tramdL, in the same orientation as tramdl and tramd2. It consists

only of 3'tramdorin sequences, and is defined by a single EST cDNA, 1388139. All four putative genes are found on both the Celera and IHGSC sequences. The structures of individual tramdorin genes are shown in greater detail as described below.

Figure 8 presents the organization of the human tramdorin loci. Sequences are from Celera human chromosome 5 scaffold sequence GA x2HTBL3TT27: 2500000-3000000, except where specified. The human tramdorin 1 locus is shown in Figure 8A. For cDNA htramdl#l, the dashed line represents an apparent non-splicing event has joined sequences that are located 15.7kb apart in the genome. The human tramdorin 2 locus is depicted in Figure 8B. Exons 1-7 were identified by homology to mouse tramdl and/or human tramd3. Exons 8-10 were identified by homology to EST cDNA 1837427. The human tramdorin 3 locus is depicted in Figure 8C. Sequence from Celera, except 1180nt of ambiguous sequences between exons 3 and 4 were replaced with corresponding sequence from IHGSC sequence Nit 006951. 3, as represented by the break marks between exons 3 and 4. Three EST cDNAs contain sequences that are homologous to mouse tramdl; a fourth, 1845574, contains sequence that are homologous to the 3'end of EST cDNA 1738130. These cDNAs reveal that expression of the tramd3 gene generates multiple alternative transcripts. EST cDNA 1388139 contains sequences that correspond to both chromosome 5q and to the STK4/Mst-l/Krs-2 locus on chromosome 20 (Figure 8D); the corresponding genomic regions are diagrammed. This EST encodes only one exon that is homologous to mouse tramdorin 1; on the chromosome 5q genomic sequence, no other exons with homology to. mouse or human tramdorin genes was found within 20kb of this exon. This locus does not encode a complete tramdorin protein, and therefore is called tramdorin L.

Figure 9 shows sequence between nucleotides 50001 and 300000 of Homo sapiens chromosome 5 working draft sequence segment Nit 006951. 4. Each line of the figure has 100 nucleotides and each page has 7000 nucleotides. At the top of each page, the coordinates on sequence Nit 006951. 4 of the first nucleotide on that page is given. Known exons for tramdorin 1, tramdorin 2, tramdorin 3 and tramdL are shown in bold type and are also indicated in the right margin. These exons were identified by their inclusion in human cDNAs or are inferred by homology to the homologous mouse gene, or to paralogous human sequences.

Initiation ATG and stop codons, and putative polyadenylation signal sites are shown in bold

italic. Note that tramd3 is in the opposite orientation relative to the other tramd genes, and is shown in the antisense orientation. Note that a single nucleotide polymorphism (C-T) in exon 2 of human tramdorin 1 at position 222682 has been confirmed, resulting in a synonymous change of a leucine codon. Other polymorphisms may also exist.

The structure of the human tramdorin 1 gene was determined by identifying regions of homology between the mouse and human tramdorin genes using the Pustell homology program (Pustell et al., Nucleic Acids Res 10: 4765 (1982b) ). This program identified both putative exons (Figures 8A and 9) as well as other conserved sequences that may define transcriptional or splicing enhancers. Additional putative transcribed regions of the tramdorin 1 gene were defined by the EST cDNA 3184556 and a cDNA that was isolated from a human sciatic nerve cDNA library kindly provided by Drs. James Lupski and Cornelius Berkoel of Baylor University.

Exons 1-10 of the tramdorin 2 gene are depicted in Figures 8B and 9; they are defined by a single EST cDNA, 1837427, and by homology with tramdorins 1 and 3, and mouse tramdorin 2 cDNA. It should be noted that mouse tramd2 cDNA was used to identify the first exon. The known and inferred exons span 26kb of genomic DNA.

The sequences of 4 alternatively spliced tramdorin 3 EST cDNAs were used to infer the structure of the tramdorin 3 gene (Figures 8C and 9). These cDNAs possess distinct sets of exons that together span 50 kb of genomic DNA. They demonstrate that expression of the tramd3 involves extensive alternative RNA processing to generate transcripts that encode tramd3 proteins with 3 distinct C termini. In addition, the distinct 5'ends of EST cDNAs 2549054 and 1738130 could indicate the presence of two tramd3 promoters.

A fourth tramdorin-related sequence, tramdL, is defined by the EST cDNA 1388139 (SEQ ID NO : 23) (Figures 8D, 9 and 25). tramdL contains only sequences that are homologous to exon 10 of tramdorinl. This cDNA is derived from a parathyroid adenoma, and is unusual in that its 5'end consists of sequences derived'from a member of the STE20 family of kinases, STK4/Mst-l/Krs-2 (Figure 12A; Creasy et al., JBiol Chem 270: 21695 (1995); Taylor et al., PNAS USA 93 : 10099 (1996) ), located on human chromosome 20. The STK4/Mst-l/Krs-2 cDNA fragment is fused to 3'sequences of tramdL (Figure 12B), located on chromosome 5.

Human STK4/Mst-l/Krs-2 and tramdL sequences. EST cDNA 1388139 encodes a fusion gene

product that fuses 5'MST kinase sequences to tramdorin 3'sequences at a splice junction.

Genomic sequence from nucleotides 8189548 to 8196472 of human chromosome 20 contig NT011382, which contain exons 6-9 of the serine-threonine kinase STK4/Mst-l/Krs2, are shown in Figure 12A. The 5'end of EST cDNA 1388139, within exon 6, is shown as an asterisk in the genomic sequence. The portion of the conserved kinase domain that is encoded by exons 6-8 is shown in bold. The caspase-3 cleavage site that activates the kinase is shown in bold italic. In EST cDNA 1833139, exon 9 is spliced to tramdL on human chromosome 5.

The nucleotide sequence of tramdL EST cDNA 1388139 (SEQ ID NO: 23) is shown in Figure 25. Vector sequences 5'to the EcoRI site (GAATTC) and 3'to the polyA stretch have been deleted, as have all but 11 A's in the polyA stretch. The point at which the cDNA diverges at its 5'end from STK4/Mst-l/Krs2 sequences is marked with a dot (.). Sequences between this point and the termination codon are translated (SEQ ID NO: 71). MscI (TGGCCA) and DraI (TTTAAA) sites that were used to generate a tramdL probe are underlined. Human chromosome 5 genomic sequence between nts 55336 and 56128 on Celera scaffold sequence GA x2HTBL3TT27: 2500000-3000000 is shown in Figure 12B, along with an open reading frame that is significantly conserved with the C-termini of tramd 1-3. An asterisk following the open reading frame denotes a termination codon, whereas the asterisk in the sequence denotes the location of the polyadenylation site of EST cDNA 1833139 (Figure 12B). The fusion between STK4/Mst-l/Krs-2 and tramd L involves the 5'splice site of STK4/Mst-l/Krs-2 exon 9, and a 3'splice site of tramdorin L (figure 12C). Such a fusion is unlikely to result from an artifact of cDNA synthesis. A comparison of proteins encoded by STK4/Mst-l/Krs-2 and EST cDNA 1833139 is shown in Figure 12D. Caspase-3 cleaves the peptide sequence DEMD (SEQ ID NO : 57), thereby separating active p36 kinase from the 3'inhibitory domain. In the putative STK4/Mst-l/Krs2-TramdL fusion protein (SEQ ID NO : 71), the C-terminus of STK4/Mst- l/Krs2 is replaced by tramdL peptide sequences that include a complete transmembrane domain, shown inserted in a membrane. The kinase activity of C-terminal deletions of STK4/Mst-l/Krs-2 (Creasy et al., JBiol Chem 271: 21049 (1996) ) suggests that an STK4/Mst- l/Krs-2-tramdL fusion protein would retain some kinase activity.

Comparison of the genomic sequences for STK4/Mst-l/Krs-2 (SEQ ID NO : 27) and tramdL (SEQ ID NO : 29) reveal that the fusion site corresponds to a 5'splice site in the

STK4/Mst-l/Krs-2 gene, and a 3'splice site in tramdL (Figure 12C). Such a fusion is unlikely to be an artifact of cDNA synthesis. This observation raises the possibility that tramdL resides near a parathyroid tumor translocation breakpoint, between chromosomes 20 and 5, that results in expression of a STK4/Mst-l/Krs-2-tramdorinL fusion protein. The predicted fusion protein encoded by EST cDNA 1388139 is depicted in Figure 25 (SEQ ID NO : 71), and a predicted fusion protein sequence containing additional 5'human STK4 sequence is depicted in Figure 26 (SEQ ID NO: 44). STK4/Mst-l/Krs-2 contains a C-terminal inhibitory domain (Creasy et al., 1996) that is cleaved by Caspase-3 to generate a cleavage product, p36, with kinase activity (Graves et al., Embo J 17: 2224 (1998); Lee et al., Oncogene 16 : 3029 (1998) ; Watabe et al., J Biol Chem 275: 8766 (2000) ). The STK4/Mst-l/Krs-2-tramdorinL fusion protein would replace STK4/Mst-l/Krs-2 C-terminal sequences with a C-terminal transmembrane domain (Figure 12D). Based on the activity of C-terminal deletions of STK4/Mst-l/Krs-2 (Creasy et al., JBiol Chem 271: 21049 (1996) ), the fusion protein may retain kinase activity, and may represent an important aspect of the etiology of parathyroid tumors. However, the present invention does not require an understanding of the underlying mechanism, and the invention is not to be limited to any particular mechanism.

Example 7 This example describes the expression pattern of human tramdorin genes.

To determine the tissues in which the various tramdorin genes are expressed, a northern blot of RNA derived from 12 human tissues was sequentially hybridized to radiolabelled cDNA probes that correspond to tramdl, 2,3 and tramdL (Figure 13). A human multitissue northern blot was purchased from OriGene Technologies, Rockville, MD, and probed sequentially with a 440bp EcoRI+Nco fragment of human tramd2 EST1837427, a 340bp MscI-DraI fragment of human tramdL EST1388139, a 850bp StuI+KpnI fragment of human tramd3 EST2549054, a 355bp EcoRI+PvuII fragment of human tramdl EST3184556, and GAPDH. Hybridization to a GAPDH probe shows that RNA is present in all lanes. Phosphorimager exposure times were 2 weeks for tramdorins 1 and 2, one week for tramd3 and overnight for GAPDH.

Each probe produced a distinct hybridization result (see Figure 13). The tramdl probe hybridized to a 4kb transcript that appears to be expressed only in muscle and kidney. Tramd2

appears to be encoded by a 3 kb transcript that is expressed only in testes. Tramd3 is the most widely expressed of the tramd genes ; it is encoded by a 7 kb transcript that is present in every tissue except liver and muscle. It is most highly expressed in brain, in which an alternative 9kb tramd3 transcript is seen also. Its pattern of expression is nearly complementary to that of tramdl ; both genes are expressed together only in kidney. In contrast to tramdl-3, no expression was observed for tramdL (data not shown). Either the gene is not expressed, consistent with the inability to find 5'sequences for it, or it is expressed only in tissues not represented on the northern blot. The differences in the sizes of transcripts that are hybridized by the various tramd probes also demonstrates that in spite of the considerable amino acid conservation between the tramd proteins, cross-hybridization between the tramd genes in minimal under the conditions used.

Example 8 This example demonstrates the conservation among human tramdorins.

Translation of the known putative coding exons for tramdorins 1,2 and 3 indicate that these proteins are highly conserved (Figure 14). Comparison was by the Clustal W algorithm (Thompson et al., Nucleic Acids Res 22: 4673 (1994) ). Tramdl, tramd 2 and tramd3 appear to have ten coding exons except for the N-terminus (first coding exon sequences). Consensus glycosylation sites in the putative second extracytoplasmic loop (Figure 3), amino acids 181- 183 and 190-192 are conserved in tramdorins 1 and 3, but not in tramdorin 2.

Example 9 This example demonstrates the conservation of tramdorin proteins between different species.

Clustal W analysis of putative tramdorin 1 proteins from human, rat, mouse and two invertebrate proteins that were identified by genome sequencing is shown in Figure 15. The most closely related sequences to tramdorins are putative proteins that have been derived from the Drosophila, Caenorhabditis elegans and Saccaromyces cerevisiae genome sequences (Figure 15). Mouse tramdl protein is 39% identical to Drosophila protein CG13384, and 32% identical to the T27A1. 5 protein from C. elegans. The functions of these proteins in

invertebrates may be related to tramdorin function in vertebrates. If so, tramdorins would be required for a function that is conserved by evolution, and predates the origin of myelin. The vertebrate proteins to which the tramdorins are most closely related are vesicular y- aminobutyric acid (GABA) transporters (VGATs ; Figure 16; (McIntire et al., Nature 389: 870 1997 ; Sagne et al., FEBS Lett 417: 177 (1997) ), to which mouse tramdl is 21-22% identical.

Note that transmembrane domains 1-10 of (McIntire et al., Nature 389: 870 (1997) ) correspond to predicted transmembrane domains 2-11 of tramdl. GABA is the principal inhibitory neurotransmitter in the brain (reviewed in Zigmond, Fundamental Neuroscience, Academic Press, San Diego Vol. , pp. xvi, 1600 (1999) ), and VGATs are thought to pack GABA into vesicles prior to exocytosis into the synapse. The similarity between VGATs and tramdorins suggests that the latter may also transport small molecule (s) with significant biological importance. Furthermore, the observation of neurons with GABA, but without VGAT, suggests the existence of additional GABA transporters (Chaudhry et al., JNeurosci 18: 9733 (1998)).

Example 10 This example presents the structure of the mouse tramdorin 1 and tramdorin 3 genes.

The organization of the mouse tramdl gene was studied to understand better the origin of the alternatively spliced tramdorin cDNAs, and as a preliminary step toward generating mice that lack tramdl function. A 129 mouse genomic library in bacteriophage k was screened with a probe consisting of a 720bp XhoI-NcoI fragment from cDNA 1920302 that contains sequence from exons 1-6. Three phage were isolated (Figure 17A). In addition, a 129/Sv bacterial artificial chromosome (BAC) library (Research Genetics, Huntsville, AL) was screened using the polymerase chain reaction (PCR) with primers 5'GCT GCC ACA AGA ACG AGA CG 3' (SEQ ID NO : 58) and 5'ATG ATG ACC AGG CTG ACC AGC 3' (SEQ ID NO : 59), that amplify 169bp of exon 6. A single tramdorin-containing BAC was isolated. Subclones from the BAC and from the lambda phage were sequenced to determine the structure of the 5'end of the tramdorin gene. A partial restriction map of mouse tramdl is shown at the top of the Figure 17A. The exact location of exon 5 relative to exons 4 and 6 is unknown, and the sizes of introns between exons 7-10 are unknown. Shown below the map are three tramdl EST cDNAs that

define tramdl exons. Genomic regions that have been sequenced are shown as thick lines below the cDNAs, and below them, select subcloned fragments are shown (Figure 17A). The genomic sequence 3'to exon 7 corresponds to sequence in EST cDNA 1363993, indicating that this cDNA is partially spliced. Exons 8, 9, and 10 are defined by sequences from the Ensembl database. Mouse tramdorin 1 genomic sequences are presented in Figure 18. Untranslated exon sequences are shown without spacing between the nucleotides, whereas the tramdorin open reading frame is marked by triplet nucleotide spacing.

Mouse and human tramdl genomic sequences were compared using the Pustell homology program (Pustell et al., Nucleic Acids Res 10: 51 (1982a)). The exons were highly conserved, but in addition, short stretches of intronic and 5'sequences were also conserved (Figure 18); these sequences may be important for tramdl RNA transcription or processing.

Notably, the intron 3'to exon 7, which is unspliced in mouse EST cDNA 1363993 and human cDNA htramdl#l, contains several such conserved sequences. In Figure 18, nucleotides that are conserved between mouse and human in orientation and spacing relative to one another are shown in bold. Nucleotides and exons are numbered to the right. Genomic sequence for the coding region of exon 10 has not been determined, and therefore exon 10 is listed in parentheses opposite a gap in the sequences that contains it. Restriction sites that correspond to the endpoints of subcloned fragments are shown in italics. Sequences between nucleotides 5188 and 5563, and 14823 and 17056 are derived from the Ensembl mouse genome sequence database and are derived from C57BL/6; all other sequences are derived from al29/Sv BAC.

To determine if the BAC contained genomic sequences for additional mouse tramdorin genes, plasmids that contained shotgun-cloned BAC DNA were arrayed on dot blots. Sequence analysis of clones that hybridized to radiolabelled tramdl cDNA probes revealed that in addition to tramdl sequences, the BAC also contained tramdorin 3 sequences (Figure 17B).

Exons 2-8 were defined by homology to mouse tramdl or human tramd3. Sequenced regions are shown below the exons; these sequences were obtained from the ends of the cloned fragments shown at the bottom of figure 17B, or from the Ensembl mouse genome sequence database.

Mouse tramdorin 3 genomic sequences (exons 1-11 and a possible 12th exon) are presented in Figure 19. Mouse genomic sequences encompassing the tramdorin3 gene are

shown. Tramd3 exons, as defined by inclusion in 5'and 3'RACE cDNA clones, and a PCR- amplified cDNA, are shown in bold. The initiation ATG codon, TAA stop codon and a putative AATAAA (SEQ ID NO : 60) polyadenylation site, are underlined. The sequence was assembled as follows: Unclipped sequences with homology to mouse tramd3 cDNA sequences were downloaded from the Ensembl trace server (http ://trace. ensembl. org), and assembled using the AssemblyLIGN software (Oxford Molecular). These sequences are derived from C57BL/6 mice. In addition, mouse tramd3 genomic sequences from strain 129/Sv were obtained from subclones of a mouse BAC clone that was isolated on the basis of its containing mouse tramdl sequences. These 129/Sv sequences are shown in the figure in italics. Several introns are not represented by complete sequence. The amount of missing sequence is estimated based on the size of the corresponding intron in human tramdorin3, with the exception of the distance between exon 11 and a possible 12th exon that was identified on the basis of homology to RACE cDNA mtramd3R3'58&num 37. In this case, the distance is defined by the estimated 4kb length of clone G10P619257, end sequences from which are homologous to exon 11 and the 3'end of RACE cDNA mtramd3R3'58&num 37. Note that the exon numbering differs between mouse and human. Mouse exonl is most homologous to human exonlb.

Mouse exon 8 is homologous to human exonlO. Two putative polymorphisms between C57BL/6 and 129/Sv mice were identified. A stretch of 9 C residues in intron 6, marked by an asterisk, is 12 C residues in C57BL/6 mice. The W at position 5539 may represent sequencing error or polymorphism; it is an A in 129Sv sequence, and a T in C57BL/6 sequence.

Discrepancies between cDNA and genomic sequence may reflect polymorphism between C57BL/6,129/Sv and Swiss Webster, the strain from which the cDNA that was used for RACE was derived; alternatively, they may be sequencing errors. These are listed below: 465 G-A in exon 1 12250 GTCA-TTTT, and 12272 G-T, in exon 11: likely genomic sequencing errors, based on comparison of the alternative translated sequences with human tramd3 and/or mouse tramdl A comparison between mouse and human tramdorin 3 is presented in Figure 20. The amino acid sequences of tramd3 are highly conserved between mouse and human, similarly to human and mouse tramdl (Figure 15). Additional comparisons between human tramdorins 1

and 2 and mouse tramd3 confirm that this mouse locus is the homolog of human tramd3 (data not shown).

Because BACs typically contain approximately 100kb of DNA, the presence of tramdl and tramd3 sequences in the same BAC suggests that these genes may reside more closely to one another in mice than they do in humans.

Example 11 This example provides putative and composite cDNAs for different tramdorins.

A. Putative cDNA for human tramdorin 1 (SEQ ID NO : 3) and its amino acid sequence (SEQ ID NO: 19) is shown in Figure 10. Exons 8 and 9 of this cDNA were identified by sequencing EST cDNA 3184556, whereas exons 7,8, and part of the 3'untranslated region were found in htramdl&num l, which was isolated from a human sciatic nerve cDNA library kindly provided by Dr. James Lupski of Baylor University. The remaining exons were identified as follows: A file that contains the sequence in the Celera human genome scaffold GA x2HTBL3TT27e2.5-3. 0 was generated using MacVector software (Oxford Molecular Ltd.).

This sequence was shown by BLAST searches of the Celera database to contain regions of homology to the mouse tramdl cDNA, and to human tramdl EST cDNA 3184556.30kb chunks of this sequence, from nucleotides 1 to 150,000 were analyzed for homology to mouse tramdl EST cDNA 1920302, human tramdl EST cDNA 3184556, human tramd2 EST cDNA 1837427, and human tramd3 EST cDNAs 1733180,2549054, and DFKZp434x1123 using the MacVector Pustell homology program (Pustell et al., Nucleic Acids Res 10: 51 (1982a)).

Multiple regions of homology on the human genomic sequence were found, but only those in the correct order and orientation relative to known exons 6,7 and 8 were considered further as candidate exons. The genomic DNA flanking candidate exons was examined manually for 5' and 3'splice sites with close matches to the consensus human splice sites (Senapathy et al, Methods Enzymol 183: 252 1990). Sequence between the splice sites was translated in all three reading frames. If the amino acid sequence in a reading frame displayed conservation to the amino acid sequence of mouse tramdl or human tramd3, the region of homology was considered a bona fide exon. In this manner, all ten coding exons of human tramdl were identified. Note that non-coding exons were not identified.

B. Putative cDNA for human tramdorin 2 (SEQ ID NO : 4), and the corresponding amino acid sequence (SEQ ID NO : 20) is shown in Figure 11. Exons 8-10 were identified by inclusion in EST cDNA 1837427. Remaining exons were identified as described above for human tramdl, or by homology to mouse tramd2 cDNA. This cDNA contains the entire human tramd2 protein coding region, but does not contain the entire 3'untranslated region. In the figure, vertical lines mark exon boundaries; exons are labeled to the right of the sequence.

C. Composite human tramd3 cDNA and the corresponding amino acid sequence is shown in Figure 21. Nucleotides 1-1311 are derived from EST cDNA 2549054, whereas nucleotides 641-2057 are derived from EST cDNA DFKZp434Gl 123. The two cDNAs overlap between the 5'end of DFKZp434G1123, which is marked by a dot (d) at nucleotide 641, and an exon boundary between nucleotides 1311 and 1312, at which their sequences diverge. A diagram of alternative human tramd3 cDNAs is shown in Figure 8, and the full genomic sequence for the human tramdorin 3 gene is contained within the sequence presented in Figure 9. The locations of exon boundaries at which alternative cDNAs diverge are marked with vertical slashes (1). An additional alternative human tramd3 EST cDNA, 1738130, possesses different 5'and 3'ends from EST cDNA 2549054 and diverges from it at the exon boundaries shown between nucleotides 146 and 147, and 875 and 876. Both EST cDNAs 1738130 and 2549054 diverge at their 3'termini from DFKZp434G1123, and encode truncated proteins. The putative initiation ATG is shown in bold and is a good match to the Kozak consensus (Kozak, Nucleic Acids Res 15: 8125 (1987) ). Two ATG sequences reside 5'to the consensus start site in EST cDNA 2549054, as does one in EST cDNA 1738130; they do not match the Kozak consensus and therefore are not likely to be used. The sequences of EST cDNAs 2549054 and 1738130 converge at a splice site five nucleotides 5'to the putative initiation ATG; the context for this ATG in EST cDNA 1738130 (CTGACTGCCATGT) is a slightly weaker match to the Kozak consensus start site than its context in EST cDNA 2549054.

Thus it is possible that mRNAs corresponding to EST cDNA 2549054 are translated more efficiently than those that correspond to EST cDNA 1738130. The number of A residues at the 3'end of EST cDNA DFKZp434Gl 123 has been arbitrarily shortened to 12.

D. Composite mouse tramdorin2 cDNA and corresponding amino acid sequence is shown in Figure 22. Mouse tramdorin2 cDNAs were isolated from a mouse testicle cDNA

library (Ambion) by 5'and 3'RLM-RACE (Maruyama et al., Gene 138 : 171 (1994); Schaefer, Anal Biochem 227: 255 (1995) ), using a FirstChoiceTM RACE-ready cDNA Kit (Ambion), and the following primers, located in exon 10: 5'AGGCTGTGCGGACAGTCAGGTCTA 3' (SEQ ID NO : 61) 5'AGACGACATAGGGGACGATGATCTCAGC 3' (SEQ ID NO : 62) 5'GCTGTACCAGTCAGTCAAGCTGAT 3' (SEQ ID NO : 63) Sequences from the 5'and 3'RACE cDNAs were used to generate the composite DNA sequence. Vertical bars in the nucleotide sequence denote exon boundaries. The absence of a consensus polyadenylation site, and the observation that the poly (A) sequences at the 3'end are found in genomic DNA, indicate that the 3'RACE cDNA is not full length.

E. Composite mouse tramdorin 3 cDNA and corresponding amino acid sequence is shown in Figure 23A. Mouse tramdorin 3 cDNAs were obtained as follows: 5'and 3'RACE cDNAs were amplified from Swiss Webster mouse brain RACE-ready cDNA (Ambion) by 5' and 3'RLM-RACE (Maruyama et al., Gene 138: 171 (1994); Schaefer, Anal Biochem 227: 255 (1995) ), using a FirstChoice RACE-ready cDNA Kit (Ambion), and the following primers: 5'RACE primers: 5'-CAGCAGGGAGAAGATGGACAACACAC-3' (SEQ ID NO : 64) 5'-AGCTGAGTGACGATGAGGAAGAAGTCCAC-3' (SEQ ID NO : 65) 3'RACE primers: 5'-CTATGGGGACACGGTGATGTATG 3'. (SEQ ID NO : 66) 5'-GGGGAAGGCGCATCGTGGA 3'. (SEQ ID NO : 67) A mouse tramd3 cDNA clone that contains sequences from exon 3 to exon 11 was obtained by PCR amplification of the same brain RACE-ready cDNA using primers 5' CCTCTCAGCCTGCTGGTGATTG 3' (SEQ ID NO : 68) and 5'GGACACTACTGGGAGACACACAGG 3'. (SEQ ID NO : 69) All cDNAs were cloned into pCRII (Invitrogen) resulting in GAATTCGGCT (SEQ ID NO : 70) sequences flanking the cDNAs. In figures 23A-23C, vertical lines mark exon boundaries; exons are labeled to the right of the figure

The composite mouse tramd 3 cDNA shown in Figure 23A contains the entire tramd3 protein coding region, but does not contain the entire 3'untranslated region. It was made by combining sequences from 5'RACE cDNA clone mtramd3R5'50&num 22, a 580bp cDNA which contains exons 1-5 and part of exon 6, and clone 61-45, a 1250bp cDNA which was PCR amplified from mouse brain cDNA using primers that are specific for sequences in exon 3 and 11.

Figure 23 B shows mouse tramd3 3'RACE cDNA 58#37. This cDNA may represent an internally deleted tramd3 cDNA. It fuses sequences from exon 7, shown in parentheses because its 3'sequences are missing, to putative tramd3 3'untranslated sequences that include a polyadenylation site. These sequences are denoted with a question mark, because it is unknown whether they represent a separate exon, or the 3'end of exon 11.

Figure 23C shows mouse tramd3 3'RACE cDNA 56#22. Similar alternatively spliced cDNAs that truncate the tramd reading frame have been found for mouse tramdl and human trand3. These observations suggest that such splicing events may have biological importance, perhaps by generating a truncated protein that could function as a dominant negative regulator of tramd activity. This cDNA contains a splice junction between exon 10 and an intracistemal A particle (IAP) repeat sequence (Aota et al. ,"Nucleotide sequence and molecular evolution of mouse retrovirus-like IAP elements. "Gene 56: 1-12 (1987); Ono, "Molecular biology of type A endogenous retrovirus"Kitasato Arch Exp Med 63: 77-90 (1990) ). Due to the repetitive nature of the IAP sequences, the location of the putative tramd3 IAP sequence could not be determined.

Example 12 This example describes an assay to identify the mouse and human tramdorin ligands.

COS-7 cells are transfected with Lipofectin (Life Technologies, Grand Island, NY), as described in Sagne et al. (2001; supra). Briefly, 1 day before transfection, 50,000 cells/well are plated in 24-well dishes. The day of transfection, cells are washed once with 0.5 ml of serum- free medium and then incubated 16-20 h with 250 J. l of serum-free medium containing a complex formed by 3 all of Lipofectin and 1 jj. g ofpcDNA3 or pcDNA3-tramdorin. The tramdorins to be tested will include mouse tramdorin 1, mouse tramdorin 2, human tramdorin 1,

human tramdorin 2 and human tramdorin 3. pcDNA3.1 is suitable for expression of cDNAs.

One milliliter of medium supplemented with FCS is added on the following day, and transport assays are performed 36-48 h after the beginning of transfection. Cells are washed twice with 0.5 ml of Krebs-Ringer (KR) phosphate buffer (146 mM NaCI/3 mM KCI/1 mM Cal2/1 mM MgCl2/lOmM KH2PO4/K2HP04) adjusted at pH 7.5 and then incubated for 15 min at 26°C in KR buffer adjusted at pH 5.5, supplemented with 0. 5-1, uCi of [3H] GABA or [3H] labeled amino acids of interest and 100 pM GABA. Reaction is terminated by two washes with ice-cold KR buffer at pH 7.5. Cells are lysed in 0.1 N NaOH, and their radioactivity is measured after neutralization by scintillation counting in Aquasol (Packard). [3H] GABA, L- [3H] glutamine and L- [3H] glutamate are available from Amersham Pharmacia ; L- [3H] proline and L-[3H] alanine are available from NEN.

Example 13 Transfection of Mouse Tramdorinl A 1566bp BglII-XmnI fragment of mouse EST cDNA 1920302 [which defines a 2.5 kb cDNA (SEQ ID NO : 13/GenBank Acession No.: AI 1780664], containing the entire coding region, was cloned between the BamHl and EcoRV sites in the expression plasmid pcDNA3.1 (Invitrogen) and transiently transfected into Cos, HeLa, and 293 cells. The cells were grown in low-glucose Dulbecco's modified Eagle's Medium (DMEM) supplemented by 10% fetal bovine serum (FBS) and antibiotics (100 zg/ml penicillinlstreptomycin) in a humidified atmosphere containing 5% C02 at 37°C. Both Lipofectin (Gibco-BRL) and plasmid DNA were incubated in Optimen for 30 min in RT, then combined for another 15 min. The cells (approximately 80% confluent) were washed with Optimen, then incubated with the combined Lipofectin/DNA solution for 6 h at 37°C. After 6 h the cells were washed once with Hank's BSS (calcium or magnesium free) and incubated for 3 days in DMEM at 37°C, then replated for immunoblotting or immunostaining.

Example 14 Immunoblotting Plates of confluent cells in 100 mm plates were harvested in cold Dulbecco's PBS lacking calcium and magnesium (Life Technologies). The cell pellet was lysed in ice-cold 50 mM Tris, pH 7.0, 1% SDS, and 0.017 mg/ml phenylmethylsulfonyl fluoride (Sigma), followed by a brief sonication on ice with a dismembrator (Fisher Scientific). Protein concentration was determined using the BioRad kit (Bio-Rad Laboratories) according to manufacturers instructions. For each sample, after a 5-15 min incubation in loading buffer at RT, 100 ug of protein lysate were loaded onto a 12% SDS-polyacrylamide gel, electrophoresed, and transferred to an Immobilon-polyvinylidene fluoride membrane (Millipore) over 1 hr, using a semidry transfer unit (Fisher). The blots were blocked (5% powdered skim milk and 0.5% Tween-20 in Tris-buffered saline) overnight at 4°C and incubated for 24 h at 4°C in a rabbit antiserum against tramdorinl (diluted 1: 1,000). After washing in blocking solution and Tris- buffered saline containing 0.5% Tween-20, blots were visualized by enhanced chemiluminescence (Amersham) according to the manufacturer's protocols.

Example 15 Immunostaining Transfected cells were plated onto plated onto 4-chamber glass slides (Nalge Nunc Intl) and incubated for 2-3 days to approximately 60% confluency. The cells were washed in PBS, and then fixed in acetone at-20°C for 10 min, then blocked with 5% fish skin gelatin in PBS containing 0.1% Triton for 1 hr in RT. Cells were labeled with rabbit anti-tramdorinl (1: 500), and processed as described below. Unfixed rat sciatic nerves were embedded in OCT and immediately frozen in a dry ice-acetone bath. Five micron thick cryostat sections were thaw- mounted on SuperFrost Plus glass slides (Fisher Scientific) and stored at-20°C. Teased nerve fibers were prepared from adult rat sciatic nerves, and dried on SuperFrost Plus glass slides overnight at room temperature and stored at-20°C. Sections and teased fibers were post-fixed and permeabilized by immersion in-20°C acetone for 10 minutes, blocked at room temperature for at least 1 hour in 5% fish skin gelatin containing 0.5% Triton X100 in PBS, and incubated 16-48 hours at 4°C with various combinations of primary antibodies: rabbit anti-tramdorinl

(1: 500); mouse arat MAG (clone 513, Boehringer Mannheim, 1: 100); mouse aLAMPl (Developmental Hydridoma Bank 1: 10). After incubating with the primary antibodies, the slides were washed, incubated with the appropriate fluorescein-and rhodamine-conjugated donkey cross-affinity purified secondary antibodies (diluted 1: 100; Jackson ImmunoResearch Laboratories, West Grove, PA). Slides were mounted with Vectashield (Vector Laboratories, <BR> <BR> <BR> <BR> Inc. , Burlingame, CA) and examined by epifluorescence with TRITC and FITC optics on a Leica DMR light microscope and photographed with a cooled Hamamatsu camera or followed by image manipulation with Adobe Photoshop.

All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in the art are intended to be within the scope of the following claims.