Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS OF DIAGNOSING & TREATING DIABETES AND INSULIN RESISTANCE
Document Type and Number:
WIPO Patent Application WO/2006/010051
Kind Code:
A3
Abstract:
The present invention provides compositions and methods for diagnosing and treating diabetes and insulin resistance. In particular, the invention provides methods of identifying modulators of the polynucleotides or polypeptides of the invention and using those modulators to treat diabetes, as well as methods of diagnosing diabetes by measuring the levels of the polynucleotides or polypeptides of the invention in a patient.

Inventors:
MOODIE SHONNA A (US)
PEREZ GRACE (US)
ZHANG FANG (US)
Application Number:
PCT/US2005/024365
Publication Date:
February 22, 2007
Filing Date:
July 07, 2005
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
METABOLEX INC (US)
MOODIE SHONNA A (US)
PEREZ GRACE (US)
ZHANG FANG (US)
International Classes:
G09B9/00; G09B9/56
Foreign References:
US5811242A1998-09-22
US6074822A2000-06-13
Other References:
WILKIN T. ET AL.: "Markers for insulin dependent diabetes: towards early detection", BRIT. MED. J., vol. 293, 22 November 1986 (1986-11-22), pages 1323 - 1326, XP003008378
Attorney, Agent or Firm:
HINSCH, Matthew, E. et al. (2 Embarcadero Center 8th Floo, San Francisco CA, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A method for identifying an agent for treating a diabetic or pre-diabetic individual, the method comprising the steps of: (i) contacting an agent to a polypeptide encoded by a polynucleotide that hybridizes to a nucleic acid encoding SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28 in 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1 % SDS at 550C; and (ii) selecting an agent that modulates the expression or activity of the polypeptide or that binds to the polypeptide, thereby identifying an agent for treating a diabetic or pre-diabetic individual.
2. The method of claim 1 , wherein the polypeptide comprises an amino acid sequence at least 95% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or a protein domain thereof.
3. The method of claim 1, the method further comprising selecting an agent that modulates insulin sensitivity.
4. The method of claim 1, wherein step (ii) comprises selecting an agent that modulates expression of the polypeptide.
5. The method of claim 1, wherein step (ii) comprises selecting an agent that modulates the activity of the polypeptide.
6. The method of claim 1 , wherein step (ii) comprises selecting an agent that specifically binds to the polypeptide.
7. The method of claim 1 , wherein the polypeptide is expressed in a cell and the cell is contacted with the agent.
8. The method of claim 1 , wherein the polypeptide comprises SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28.
Description:
Methods of Diagnosing & Treating Diabetes and Insulin Resistance

CROSS-REFERENCE TO RELATED APPLICATIONS [01] This application claims benefit of U.S. Provisional Application No. 60/586,358, filed July 7, 2004, which is herein incorporated by reference.

BACKGROUND OF THE INVENTION [02] Diabetes mellitus can be divided into two clinical syndromes, Type 1 and Type 2 diabetes mellitus. Type 1, or insulin-dependent diabetes mellitus (IDDM), is a chronic autoimmune disease characterized by the extensive loss of beta cells in the pancreatic Islets of Langerhans, which produce insulin. As these cells are progressively destroyed, the amount of secreted insulin decreases, eventually leading to hyperglycemia (abnormally high level of glucose in the blood) when the amount of secreted insulin drops below the level required for euglycemia (normal blood glucose level). Although the exact trigger for this immune response is not known, patients with IDDM have high levels of antibodies against proteins expressed in pancreatic beta cells. However, not all patients with high levels of these antibodies develop IDDM. [03] Type 2 diabetes (also referred to as non-insulin dependent diabetes mellitus (NIDDM)) develops when muscle, fat and liver cells fail to respond normally to insulin. This failure to respond (called insulin resistance) may be due to reduced numbers of insulin receptors on these cells, or a dysfunction of signaling pathways within the cells, or both. The beta cells initially compensate for this insulin resistance by increasing insulin output. Over time, these cells become unable to produce enough insulin to maintain normal glucose levels, indicating progression to Type 2 diabetes. [04] Type 2 diabetes is brought on by a combination of genetic and acquired risk factors - including a high-fat diet, lack of exercise, and aging. Worldwide, Type 2 diabetes has become an epidemic, driven by increases in obesity and a sedentary lifestyle, widespread adoption of western dietary habits, and the general aging of the population in many countries. In 1985, an estimated 30 million people worldwide had diabetes ~ by 2000, this figure had increased 5-fold, to an estimated 154 million people. The number of people with diabetes is expected to double between now and 2025, to about 300 million. [05] Type 2 diabetes is a complex disease characterized by defects in glucose and lipid metabolism. Typically there are perturbations in many metabolic parameters including increases in fasting plasma glucose levels, free fatty acid levels and triglyceride levels, as well as a decrease in the ratio of HDL/LDL. As discussed above, one of the principal underlying causes of diabetes is thought to be an increase in insulin resistance in peripheral tissues, principally muscle and fat. [06] Therapies aimed at reducing peripheral insulin resistance are available. The most relevant to this invention are drugs of the thiazolidinedione (TZD) class namely troglitazone, pioglitazone, and rosiglitazone. In the US these have been marketed under the names Rezulin™, Avandia™ and Actos™, respectively. The principal effect of these drugs is to improve glucose homeostasis. Notably in diabetics treated with TZDs there are increases in peripheral glucose disposal rates indicative of increased insulin sensitivity in both muscle and fat. [07] The molecular target of TZDs is a member of the PPAR family of ligand-activated transcription factors called PPAR gamma. This transcription factor is highly expressed in adipose tissue with much lower levels being observed in muscle. Binding of TZDs to PPAR gamma in target cells and tissues such as fat and muscle brings about a change in gene expression. The link between TZD-altered gene expression in fat and muscle and increased insulin sensitivity is unknown. The present invention addresses this and other problems.

BRIEF SUMMARY OF THE INVENTION [08] The present invention provides methods for identifying an agent for treating a diabetic or pre-diabetic individual. In some embodiments, the methods comprise the steps of: (i) contacting an agent to a polypeptide encoded by a polynucleotide that is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1% SDS at 550C, wherein the polypeptide optionally has the activity listed in Table 1 ; and (ii) selecting an agent that modulates the expression or activity of the polypeptide or that binds to the polypeptide, thereby identifying an agent for treating a diabetic or pre-diabetic individual. In some embodiments, the polypeptide comprises an amino acid sequence at least 95% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or a protein domain thereof. In some embodiments, the methods further comprise selecting an agent that modulates insulin sensitivity. Table 1: List of Polypeptides, SEQ ID numbers and Proposed Activity

[09] In some embodiments, step (ii) comprises selecting an agent that modulates expression of the polypeptide. In some embodiments, step (ii) comprises selecting an agent that modulates the activity of the polypeptide. In some embodiments, step (ii) comprises selecting an agent that specifically binds to the polypeptide. In some embodiments, the polypeptide is expressed in a cell and the cell is contacted with the agent. hi some embodiments, the polypeptide comprises SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [10] The present invention also provides methods of treating a diabetic or pre-diabetic animal. In some embodiments, the methods comprise administering to the animal a therapeutically effective amount of an agent that modulates the activity or expression of SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [11] In some embodiments, the agent is identified by a method comprising (i) contacting an agent to a polypeptide encoded by a polynucleotide that is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1 % SDS at 550C; and (ii) selecting an agent that modulates the expression or activity of the polypeptide or that binds to the polypeptide. In some embodiments, the polypeptide comprises an amino acid sequence at least 95% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or a protein domain thereof, hi some embodiments, the agent is an antibody. In some embodiments, the antibody is a monoclonal antibody, hi some embodiments, the animal is a human. [12] The present invention also provides methods of introducing an expression cassette into a cell, hi some emodiments, the methods comprise introducing into the cell an expression cassette comprising a promoter operably linked to a polynucleotide encoding a polypeptide, wherein the polynucleotide is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1% SDS at 550C, and the polypeptide optionally has the activity listed in Table 1. In some embodiments, the polypeptide comprises an amino acid sequence at least 95% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or a protein domain thereof. [13] In some embodiments, the polypeptide comprises SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. In some embodiments, the cell is selected from the group consisting of an adipocyte and a skeletal muscle cell. [14] In some embodiments, the methods further comprising introducing the cell into a human. In some embodiments, the human is diabetic. In some embodiments, the human is prediabetic. In some embodiments, the cell is from the human. [15] The present invention also provides methods of diagnosing an individual who has Type 2 diabetes or is prediabetic. hi some embodiments, the method comprises, detecting in a sample from the individual the level of a polypeptide or the level of a polynucleotide encoding the polypeptide, wherein the polynucleotide is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1 % SDS at 550C, wherein a modulated level of the polypeptide or polynucleotide in the sample compared to a level of the polypeptide or polynucleotide in either a lean individual or a previous sample from the individual indicates that the individual is diabetic or prediabetic, and the polypeptide optionally has the activity listed in Table 1. In some embodiments, the detecting step comprises contacting the sample with an antibody that specifically binds to the polypeptide. In some embodiments, the amino acid sequence comprises SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [16] In some embodiments, the detecting step comprises quantifying mRNA encoding the polypeptide. In some embodiments, the mRNA is reverse transcribed and amplified in a polymerase chain reaction. [17] In some embodiments, the sample is a blood, urine or tissue sample. [18] The present invention also provides isolated nucleic acids that is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1% SDS at 550C. [19] In some embodiments, the nucleic acid comprises SEQ ID NO: 1, 3, 5, 7, 11, 13, 15, 17, 19, 21, 23, 25 or 27. In some embodiments, the nucleic acid encodes SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [20] The present invention also provides an expression cassette comprising a heterologous promoter operably linked to a polynucleotide that is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1% SDS at 550C. [21] The present invention also provides host cells transfected with a polynucleotide that is substantially identical to or hybridizes to a nucleic acid encoding a polypeptide listed in Table 1 under hybridization conditions of 50% formamide, 5X SSC, and 1% SDS at 420C followed by a wash in 0.2X SSC, and 0.1% SDS at 550C. In some embodiments, the host cell is a human cell. In some embodiments, the host cell is a bacterium. In some embodiments, the cell is selected from the group consisting of an adipocyte and a skeletal muscle cell. [22] The present invention also provides isolated polypeptides comprising an amino acid sequence at least 70% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. In some embodiments, the polypeptide comprises SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [23] The present invention also provides pharmaceutical compositions comprising polypeptides comprising an amino acid sequence at least 70% identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or fragments thereof, and a pharmaceutically-acceptable excipient.

DEFINITIONS [24] "Insulin sensitivity" refers to the ability of a cell or tissue to respond to insulin. Responses include, e.g., glucose uptake of a cell or tissue in response to insulin stimulation. Sensitivity can be determined at an organismal, tissue or cellular level. For example, blood or urine glucose levels following a glucose tolerance test are indicative of insulin sensitivity. Other methods of measuring insulin sensitivity include, e.g., measuring glucose uptake (see, e.g., Garcia de Herreros, A., and Birnbaum, M. J. J. Biol. Chem. 264, 19994-19999 (1989); Klip, A., Li, G., and Logan, WJ. Am. J. Physiol. 247, E291-296 (1984)), measuring the glucose infusion rate (GINF) into tissue such as the skeletal muscle (see, e.g., Ludvik et ah, J. Clin. Invest. 100:2354 (1997); Frias et ah, Diabetes Care 23:64, (2000)) and measuring sensitivity of GLUT4 translocation (e.g., as described herein) in response to insulin. [25] The term "lipid metabolism" refers to the in vivo process of catabolism (decomposition) and anabolism (accumulation) of lipids (e.g., triglycerides derived from food) and is intended to include, in the broad sense, reactions for transforming lipids into energy, biosynthesis of fatty acids, acylglycerol, phospholipid metabolism and cholesterol metabolism. [26] "Activity" of a polypeptide of the invention refers to structural, regulatory, or biochemical functions of a polypeptide in its native cell or tissue. Examples of activity of a polypeptide include both direct activities and indirect activities. Exemplary direct activities are the result of firect interaction with the polypeptide, , e.g., enzymatic activity, ligand binding, production or depletion of second messengers (e.g., cAMP, cGMP, IP3, DAG, or Ca2+), ion flux, phosphorylation levels, transcription levels, and the like. Exemplary indirect activities are observed as a change in phenotype or response in a cell or tissue to a polypeptide's directed activity, e.g., modulating insulin sensitivity of a cell as a result of the interaction of the polypeptide with other cellular or tissue components. [27] "Predisposition for diabetes" occurs in a person when the person is at high risk for developing diabetes. A number of risk factors are known to those of skill in the art and include: genetic factors (e.g., carrying alleles that result in a higher occurrence of diabetes than in the average population or having parents or siblings with diabetes); overweight (e.g., body mass index (BMI) greater or equal to 25 kg/m2); habitual physical inactivity, race/ethnicity (e.g., African- American, Hispanic- American, Native Americans, Asian-Americans, Pacific Islanders); previously identified impaired fasting glucose or impaired glucose tolerance, hypertension (e.g., greater or equal to 140/90 mmHg in adults); HDL cholesterol less than or equal to 35 mg/dl; triglyceride levels greater or equal to 250 mg/dl; a history of gestational diabetes or delivery of a baby over nine pounds; and/or polycystic ovary syndrome. See, e.g., "Report of the Expert Committee on the Diagnosis and Classification of Diabetes Mellitus" and "Screening for Diabetes" Diabetes Care 25(1): S5- S24 (2002). [28] A "lean individual," when used to compare with a sample from a patient, refers to an adult with a fasting blood glucose level less than 100 mg/dl or a 2 hour PG reading of 140 mg/dl. "Fasting"refers to no caloric intake for at least 8 hours. A "2 hour PG" refers to the level of blood glucose after challenging a patient to a glucose load containing the equivalent of 75g anhydrous glucose dissolved in water. The overall test is generally referred to as an oral glucose tolerance test (OGTT). See, e.g., Diabetes Care, 2003, 26(11): 3160-3167 (2003). The level of a polypeptide in a lean individual can be a reading from a single individual, but is typically a statistically relevant average from a group of lean individuals. The level of a polypeptide in a lean individual can be represented by a value, for example in a computer program. [29] A "pre-diabetic individual," when used to compare with a sample from a patient, refers to an adult with a fasting blood glucose level greater than 110 mg/dl but less than 126 mg/dl or a 2 hour PG reading of greater than 140 mg/dl but less than 200mg/dl. A "diabetic individual," when used to compare with a sample from a patient, refers to an adult with a fasting blood glucose level greater than 126 mg/dl or a 2 hour PG reading of greater than 200 mg/dl. [30] A "diabetes-related nucleic acid" or "diabetes-related polynucleotide" (also referred to as a "nucleic acid of the invention" or a "polynucleotide of the invention") of the invention is a subsequence or full-length polynucleotide sequence of a gene that encodes a polypeptide, whose activity modulates diabetes or insulin sensitivity, or whose presence or absence is indicative of diabetes or altered insulin sensitivity. Exemplary nucleic acids of the invention include those sequences substantially identical to SEQ ID NO: 1, 3, 5, 7, 11, 13, 15, 17, 19, 21, 23, 25, 27 or encoding polypeptides substantially identical to SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26 or 28. [31] An "agonist" refers to an agent that binds to, stimulates, increases, activates, facilitates, enhances activation, sensitizes or up regulates the activity or expression of a polypeptide of the invention. [32] An "antagonist" refers to an agent that binds to, partially or totally blocks stimulation, decreases, prevents, delays activation, inactivates, desensitizes, or down regulates the activity or expression of a polypeptide of the invention. [33] "Antibody" refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof which specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. [34] An exemplary immunoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kD) and one "heavy" chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively. [35] Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2; a dimer of Fab which itself is a light chain joined to VH-CHI by a disulfide bond. The F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)'2 dimer into an Fab' monomer. The Fab' monomer is essentially an Fab with part of the hinge region (see, Paul (Ed.) Fundamental Immunology, Third Edition, Raven Press, NY (1993)). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv). [36] The terms "peptidomimetic" and "mimetic" refer to a synthetic chemical compound that has substantially the same structural and functional characteristics of the antagonists or agonists of the invention. Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed "peptide mimetics" or "peptidomimetics" (Fauchere, J. Adv. Drug Res. 15:29 (1986); Veber and Freidinger TINS p. 392 (1985); and Evans et al. J. Med. Chem. 30:1229 (1987), which are incorporated herein by reference). Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent or enhanced therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biological or pharmacological activity), such as apolypeptide exemplified in this application, but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of, e.g., -CH2NH-, -CH2S-, -CH2-CH2-, - CH=CH- (cis and trans), -COCH2-, -CH(OH)CH2-, and -CH2SO-. The mimetic can be either entirely composed of synthetic, non-natural analogues of amino acids, or, is a chimeric molecule of partly natural peptide amino acids and partly non-natural analogs of amino acids. The mimetic can also incorporate any amount of natural amino acid conservative substitutions as long as such substitutions also do not substantially alter the mimetic' s structure and/or activity. For example, a mimetic composition is within the scope of the invention if it is capable of carrying out the binding or other activities of an agonist or antagonist of a polypeptide of the invention. [37] The term "gene" means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and , trailer) as well as intervening sequences (introns) between individual coding segments (exons). [38] The term "isolated," when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It is preferably in a homogeneous state although it can be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. In particular, an isolated gene is separated from open reading frames that flank the gene and encode a protein other than the gene of interest. The term "purified" denotes that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. Particularly, it means that the nucleic acid or protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure. [39] The term "nucleic acid" or "polynucleotide" refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double- stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et ah, Nucleic Acid Res. 19:5081 (1991); Ohtsuka et ah, J. Biol. Chem. 260:2605-2608 (1985); and Cassol et al. (1992); Rossolini et al, MoI. Cell. Probes 8:91-98 (1994)). The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene. [40] The terms "polypeptide," "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. As used herein, the terms encompass amino acid chains of any length, including full-length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds. [41] The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, γ -carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an α carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. "Amino acid mimetics" refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but which functions in a manner similar to a naturally occurring amino acid. [42] Amino acids may be referred to herein by either the commonly known three letter symbols or by the one-letter symbols recommended by the RJPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes. [43] "Conservatively modified variants" applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, "conservatively modified variants" refers to those nucleic acids that encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid that encodes a polypeptide is implicit in each described sequence. [44] As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention. [45] The following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)). [46] "Percentage of sequence identity" is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (e.g., a polypeptide of the invention), which does not comprise additions or deletions, for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. [47] The terms "identical" or percent "identity," in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same sequences. Sequences are "substantially identical" if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region, or, when not specified, over the entire sequence), when compared and aligned for maximum correspondence over a comparison window, designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection, or across the entire sequence where not indicated. The invention provides polypeptides or polynucleotides that are substantially identical to the polypeptides or polynucleotides, respectively, exemplified herein (e.g., SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28). This definition also refers to the complement of a test sequence. Optionally, the identity exists over a region that is at least about 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides in length. [48] For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. [49] A "comparison window", as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. MoI. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat 'I. Acad. ScL USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by manual alignment and visual inspection {see, e.g., Ausubel et ah, Current Protocols in Molecular Biology (1995 supplement)). [50] Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nuc. Acids Res. 25:3389-3402, and Altschul et ah (1990) J MoI. Biol. 215:403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive- valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et ah, supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative- scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11 , an expectation (E) or 10, M=5, N=-4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix {see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sd USA 89:10915) alignments (B) of 50, expectation (E) of 10, M=5, N=-4, and a comparison of both strands. [51] The BLAST algorithm also performs a statistical analysis of the similarity between two sequences {see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sd. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001. [52] An indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below. Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence. [53] The phrase "selectively (or specifically) hybridizes to" refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (e.g., total cellular or library DNA or RNA). [54] The phrase "stringent hybridization conditions" refers to conditions under which a probe will hybridize to its target subsequence, typically in a complex mixture of nucleic acid, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Probes, "Overview of principles of hybridization and the strategy of nucleic acid assays" (1993). Generally, stringent conditions are selected to be about 5-10° C lower than the thermal melting point (T1n) for the specific sequence at a defined ionic strength pH. The Tm is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at Tm, 50% of the probes are occupied at equilibrium). Stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 3O0C for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. For selective or specific hybridization, a positive signal is at least two times background, optionally 10 times background hybridization. Exemplary stringent hybridization conditions can be as following: 50% formamide, 5X SSC, and 1% SDS, incubating at 420C, or 5X SSC, 1 % SDS, incubating at 650C, with wash in 0.2X SSC, and 0.1% SDS at 550C, 6O0C, or 650C. Such washes can be performed for 5, 15, 30, 60, 120, or more minutes. [55] Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides that they encode are substantially identical. This occurs, for example, when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code. In such cases, the nucleic acids typically hybridize under moderately stringent hybridization conditions. Exemplary "moderately stringent hybridization conditions" include a hybridization in a buffer of 40% formamide, 1 M NaCl, 1% SDS at 370C, and a wash in IX SSC at 450C. Such washes can be performed for 5, 15, 30, 60, 120, or more minutes. A positive hybridization is at least twice background. Those of ordinary skill will readily recognize that alternative hybridization and wash conditions can be utilized to provide conditions of similar stringency. [56] The phrase "a nucleic acid sequence encoding" refers to a nucleic acid which contains sequence information for a structural RNA such as rRJSf A, a tRNA, or the primary amino acid sequence of a specific protein or peptide, or a binding site for a trans¬ acting regulatory agent. This phrase specifically encompasses degenerate codons (i.e., different codons which encode a single amino acid) of the native sequence or sequences that may be introduced to conform with codon preference in a specific host cell. [57] The term "recombinant" when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under-expressed or not expressed at all. [58] The term "heterologous" when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not found in the same relationship to each other in nature. For instance, the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid, e.g., a promoter from one source and a coding region from another source. Similarly, a heterologous protein indicates that the protein comprises two or more subsequences that are not found in the same relationship to each other in nature (e.g., a fusion protein). [59] An "expression vector" is a nucleic acid construct, generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a host cell. The expression vector can be part of a plasmid, virus, or nucleic acid fragment. Typically, the expression vector includes a nucleic acid to be transcribed operably linked to a promoter. [60] The phrase "specifically (or selectively) binds to an antibody" or "specifically (or selectively) immunoreactive with", when referring to a protein or peptide, refers to a binding reaction which is determinative of the presence of the protein in the presence of a heterogeneous population of proteins and other biologies. Thus, under designated immunoassay conditions, the specified antibodies bind to a particular protein and do not bind in a significant amount to other proteins present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein. For example, antibodies raised against a protein having an amino acid sequence encoded by any of the polynucleotides of the invention can be selected to obtain antibodies specifically immunoreactive with that protein and not with other proteins, except for polymorphic variants. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid- ' phase ELISA immunoassays, Western blots, or immunohistochemistry are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, Harlow and Lane Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, NY (1988) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity. Typically, a specific or selective reaction will be at least twice the background signal or noise and more typically more than 10 to 100 times background. [61] "Inhibitors," "activators," and "modulators" of expression or of activity are used to refer to inhibitory, activating, or modulating molecules, respectively, of expression of the polypeptides of the invention as determined using in vitro or in vivo assays to monitor expression or activity. Modulators encompass e.g., ligands, agonists, antagonists, their homologs and mimetics, as well as the polypeptides of the invention, or fragments thereof with antagonist activity or that act to increase overall polypeptide activity (i.e., fragments that have at least some of the activity of the full-length protein). In some cases, fragments of the polypeptides of the invention are at least 20, 50, 75 or 100 amino acids in length. The term "modulator" includes inhibitors and activators. Inhibitors are agents that, e.g., inhibit expression of a polypeptide of the invention or bind to, partially or totally block stimulation, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of a polypeptide of the invention, e.g., antagonists. Activators are agents that, e.g., induce or activate the expression of a polypeptide of the invention or bind to, stimulate, increase, open, activate, facilitate, or enhance activation, sensitize or up regulate the activity of a polypeptide of the invention, e.g., agonists. Modulators include naturally occurring and synthetic ligands, antagonists, agonists, small chemical molecules and the like. Such assays for inhibitors and activators include, e.g., applying putative modulator compounds to cells expressing a polypeptide of the invention and then determining the functional effects on a polypeptide of the invention activity, as described above. Samples or assays comprising a polypeptide of the invention that are treated with a potential activator, inhibitor, or modulator are compared to control samples without the inhibitor, activator, or modulator to examine the extent of effect. Control samples (untreated with modulators) are assigned a relative activity value of 100%. Inhibition of a polypeptide of the invention is achieved when the polypeptide activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%. Activation of the polypeptide is achieved when the polypeptide activity value relative to the control is 110%, optionally 150%, optionally 200, 300%, 400%, 500%, or 1000-3000% or more higher.

DETAILED DESCRIPTION OF THE INVENTION I. INTRODUCTION [62] The present application demonstrates that, surprisingly, modulated levels of mRNA comprising sequences of the invention occur in muscle from tissue either insulin resistant obese non-diabetics or from type 2 diabetic individuals treated compared to levels of the mRNA in the lean, non-diabetic individuals. Additional muscle from tissue from type 2 diabetic individuals treated with thiazolidinedione (TZD) compared to levels of mRNA in the type 2 diabetic individuals before TZD treatment. Insulin resistant obese are generally pre-diposed to become type II diabetics. Therefore, the modulation of the sequences in the study described herein indicates the sequences' involvement in diabetes and pre¬ diabetes. Without intending to limit the invention to a particular mechanism of action, it is believed that modulation of the expression or activity of the polypeptides or polynucleotides of the invention is beneficial in treating diabetic, pre-diabetic or obese insulin resistant, non- diabetic patients. Furthermore, modulated levels of the polypeptides of the invention are indicative of insulin resistance. Thus, the detection of a polypeptide of the invention is useful for diagnosis of diabetes and insulin resistance. [63] This invention also provides methods of using polypeptides of the invention and modulators of the polypeptides of the invention to diagnose and treat diabetes, pre-diabetes (including insulin resistant individuals) and related metabolic diseases. The present method also provides methods of identifying modulators of expression or activity of the polypeptides of the invention. Such modulators are useful for treating Type 2 diabetes as well as the pathological aspects of diabetes (e.g., insulin resistance).

II. GENERAL RECOMBINANT NUCLEIC ACID METHODS FOR USE WITH THE INVENTION [64] In numerous embodiments of the present invention, nucleic acids encoding a polypeptide of the present invention will be isolated and cloned using recombinant methods. Such embodiments are used, e.g., to isolate polynucleotides identical or substantially identical to SEQ ID NO: 1, 3, 5, 7, 11, 13, 15, 17, 19, 21, 23, 25 or 27 for protein expression or during the generation of variants, derivatives, expression cassettes, or other sequences derived from an polypeptide or polynucleotide of the invention, to monitor gene expression, for the isolation or detection of sequences in different species, for diagnostic purposes in a patient, e.g., to detect mutations in a polypeptide or polynucleotide of the invention or to detect expression levels of nucleic acids or polypeptides. . In some embodiments, the sequences encoding the polypeptides of the invention (or polypeptides comprising fragments of the polypeptides of the invention) are operably linked to a heterologous promoter. In some cases, fragments of the polypeptides of the invention are at least 20, 50, 75 or 100 amino acids in length. The polypeptides of the invention can be linked to heterologous amino acid sequences using recombinant DNA technology. In one embodiment, the nucleic acids of the invention are from any mammal, including, in particular, e.g., a human, a mouse, a rat, etc. [65] Polynucleotides, including expression cassettes, encoding polypeptides of the invention can be introduced into cells and optionally expressed in the cells. Polynucleotides of the invention can be introduced into eukaryotic or prokaryotic cells, including adipocyte or muscle cells. The cells can be primary cells or cell lines.

A. General Recombinant Nucleic Acid Methods [66] This invention relies on routine techniques in the field of recombinant genetics. Basic texts disclosing the general methods of use in this invention include Sambrook et al., Molecular Cloning, A Laboratory Manual (3rd ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al. , eds., 1994)). [67] For nucleic acids, sizes are given in either kilobases (kb) or base pairs (bp). These are estimates derived from agarose or acrylamide gel electrophoresis, from sequenced nucleic acids, or from published DNA sequences. For proteins, sizes are given in kilodaltons (kDa) or amino acid residue numbers. Proteins sizes are estimated from gel electrophoresis, from sequenced proteins, from derived amino acid sequences, or from published protein sequences. [68] Oligonucleotides that are not commercially available can be chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage & Caruthers, Tetrahedron Letts. 22:1859-1862 (1981), using an automated synthesizer, as described in Van Devanter et. al. , Nucleic Acids Res. 12:6159-6168 (1984). Purification of oligonucleotides is by either native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson & Reanier, J. Chrom. 255:137-149 (1983). [69] The sequence of the cloned genes and synthetic oligonucleotides can be verified after cloning using, e.g., the chain termination method for sequencing double- stranded templates of Wallace et al., Gene 16:21-26 (1981).

B. Cloning Methods for the Isolation of Nucleotide Sequences Encoding Desired Proteins [70] In general, the nucleic acids encoding the subject proteins are cloned from DNA sequence libraries that are made to encode cDNA or genomic DNA. The particular sequences can be located by hybridizing with an oligonucleotide probe, the sequence of which can be derived from the sequences disclosed herein, which provide a reference for PCR primers and defines suitable regions for isolating probes pecific for the polypeptides or polynucleotides of the invention. Alternatively, where the sequence is cloned into an expression library, the expressed recombinant protein can be detected immunologically with antisera or purified antibodies made against a polypeptide of interest, including those disclosed herein. [71] Methods for making and screening genomic and cDNA libraries are well known to those of skill in the art (see, e.g., Gubler and Hoffman Gene 25:263-269 (1983); Benton and Davis Science, 196:180-182 (1977); and Sambrook, supra). [72] Briefly, to make the cDNA library, one should choose a source that is rich in mRNA. The mRNA can then be made into cDNA, ligated into a recombinant vector, and transfected into a recombinant host for propagation, screening and cloning. For a genomic library, the DNA is extracted from a suitable tissue and either mechanically sheared or enzymatically digested to yield fragments of preferably about 5-100 kb. The fragments are then separated by gradient centrifugation from undesired sizes and are constructed in bacteriophage lambda vectors. These vectors and phage are packaged in vitro, and the recombinant phages are analyzed by plaque hybridization. Colony hybridization is carried out as generally described in Grunstein et al., Proc. Natl. Acad. ScL USA., 72:3961-3965 (1975). [73] An alternative method combines the use of synthetic oligonucleotide primers with polymerase extension on an mRNA or DNA template. Suitable primers can be designed from specific sequences disclosed herein. This polymerase chain reaction (PCR) method amplifies the nucleic acids encoding the protein of interest directly from mRNA, cDNA, genomic libraries or cDNA libraries. Restriction endonuclease sites can be incorporated into the primers. Polymerase chain reaction or other in vitro amplification methods may also be useful, for example, to clone nucleic acids encoding specific proteins and express said proteins, to synthesize nucleic acids that will be used as probes for detecting the presence of mRNA encoding a polypeptide of the invention in physiological samples, for nucleic acid sequencing, or for other purposes (see, U.S. Patent Nos. 4,683,195 and 4,683,202). Genes amplified by a PCR reaction can be purified from agarose gels and cloned into an appropriate vector. [74] Appropriate primers and probes for identifying the genes encoding a polypeptide of the invention from mammalian tissues can be derived from the sequences provided herein. For a general overview of PCR, see, Innis et al. PCR Protocols: A Guide to Methods and Applications, Academic Press, San Diego (1990). [75] Synthetic oligonucleotides can be used to construct genes. This is done using a series of overlapping oligonucleotides, usually 40-120 bp in length, representing both the sense and anti-sense strands of the gene. These DNA fragments are then annealed, ligated and cloned. [76] A polynucleotide encoding a polypeptide of the invention can be cloned using intermediate vectors before transformation into mammalian cells for expression. These intermediate vectors are typically prokaryote vectors or shuttle vectors. The proteins etal

can be expressed in either prokaryotes or eukaryotes, using standard methods well known to those of skill in the art.

III. PURIFICATION OF PROTEINS OF THE INVENTION

[77] Either naturally occurring or recombinant polypeptides of the invention can be purified for use in functional assays. Naturally occurring polypeptides of the invention can be purified from any source (e.g., tissues of an organism expressing an ortholog). Recombinant polypeptides can be purified from any suitable expression system. [78] The polypeptides of the invention may be purified to substantial purity by standard techniques, including selective precipitation with such substances as ammonium sulfate; column chromatography, immunopurification methods, and others (see, e.g., Scopes, Protein Purification: Principles and Practice (1982); U.S. Patent No. 4,673,641; Ausubel et ah, supra; and Sambrook et ah, supra). [79] A number of procedures can be employed when recombinant polypeptides are being purified. For example, proteins having established molecular adhesion properties can be reversibly fused to a polypeptide of the invention. With the appropriate ligand, either protein can be selectively adsorbed to a purification column and then freed from the column in a relatively pure form. The fused protein may be then removed by enzymatic activity. Finally polypeptides can be purified using immunoaffinity columns.

A. Purification of Proteins from Recombinant Bacteria [80] When recombinant proteins are expressed by the transformed bacteria in large amounts, typically after promoter induction, although expression can be constitutive, the proteins may form insoluble aggregates. There are several protocols that are suitable for purification of protein inclusion bodies. For example, purification of aggregate proteins (hereinafter referred to as inclusion bodies) typically involves the extraction, separation and/or purification of inclusion bodies by disruption of bacterial cells typically, but not limited to, by incubation in a buffer of about 100-150 μg/ml lysozyme and 0.1% Nonidet P40, a non-ionic detergent. The cell suspension can be ground using a Polytron grinder (Brinkman Instruments, Westbury, NY). Alternatively, the cells can be sonicated on ice. Alternate methods of lysing bacteria are described in Ausubel and Sambrook et ah, both supra, and will be apparent to those of skill in the art. [81] The cell suspension is generally centrifuged and the pellet containing the inclusion bodies resuspended in buffer which does not dissolve but washes the inclusion bodies, e.g., 20 niM Tris-HCl (pH 7.2), 1 mM EDTA, 150 mM NaCl and 2% Triton-X 100, a non-ionic detergent. It may be necessary to repeat the wash step to remove as much cellular debris as possible. The remaining pellet of inclusion bodies may be resuspended in an appropriate buffer {e.g., 20 mM sodium phosphate, pH 6.8, 150 mM NaCl). Other appropriate buffers will be apparent to those of skill in the art. [82] Following the washing step, the inclusion bodies are solubilized by the addition of a solvent that is both a strong hydrogen acceptor and a strong hydrogen donor (or a combination of solvents each having one of these properties). The proteins that formed the inclusion bodies may then be renatured by dilution or dialysis with a compatible buffer. Suitable solvents include, but are not limited to, urea (from about 4 M to about 8 M), formamide (at least about 80%, volume/volume basis), and guanidine hydrochloride (from about 4 M to about 8 M). Some solvents that are capable of solubilizing aggregate-forming proteins, such as SDS (sodium dodecyl sulfate) and 70% formic acid, are inappropriate for use in this procedure due to the possibility of irreversible denaturation of the proteins, accompanied by a lack of immunogenicity and/or activity. Although guanidine hydrochloride and similar agents are denaturants, this denaturation is not irreversible and renaturation may occur upon removal (by dialysis, for example) or dilution of the denaturant, allowing re-formation of the immunologically and/or biologically active protein of interest. After solubilization, the protein can be separated from other bacterial proteins by standard separation techniques. [83] Alternatively, it is possible to purify proteins from bacteria periplasm. Where the protein is exported into the periplasm of the bacteria, the periplasmic fraction of the bacteria can be isolated by cold osmotic shock in addition to other methods known to those of skill in the art (see, Ausubel et al, supra). To isolate recombinant proteins from the periplasm, the bacterial cells are centrifuged to form a pellet. The pellet is resuspended in a buffer containing 20% sucrose. To lyse the cells, the bacteria are centrifuged and the pellet is resuspended in ice-cold 5 mM MgSO4 and kept in an ice bath for approximately 10 minutes. The cell suspension is centrifuged and the supernatant decanted and saved. The recombinant proteins present in the supernatant can be separated from the host proteins by standard separation techniques well known to those of skill in the art. B. Purification of Proteins from Insect Cells [84] Proteins can also be purified from eukaryotic gene expression systems as described in, e.g., Fernandez and Hoeffler, Gene Expression Systems (1999). In some embodiments, baculo virus expression systems are used to isolate proteins of the invention. Recombinant baculoviruses are generally generated by replacing the polyhedrin coding sequence of a baculovirus with a gene to be expressed (e.g., encoding a polypeptide of the invention). Viruses lacking the polyhedrin gene have a unique plaque morphology making them easy to recognize. In some embodiments, a recombinant baculovirus is generated by first cloning a polynucleotide of interest into a transfer vector (e.g., a pUC based vector) such that the polynucleotide is operably linked to a polyhedrin promoter. The transfer vector is transfected with wildtype DNA into an insect cell (e.g., Sf9, Sf21 or BT1-TN-5B1-4 cells), resulting in homologous recombination and replacement of the polyhedrin gene in the wildtype viral DNA with the polynucleotide of interest. Virus can then be generated and plaque purified. Protein expression results upon viral infection of insect cells. Expressed proteins can be harvested from cell supernatant if secreted, or from cell lysates if intracellular. See, e.g., Ausubel et al. and Fernandez and Hoeffler, supra.

C. Purification of secreted proteins from mammalian cells [85] Polypeptides of the invention, and in particular, secreted proteins of the invention can be readily purified from mammalian cells expressing the polypeptides. Expression of the polypeptides can be the result of either transient or stable expression of the protein from a recombinant expression cassette introduced into the cells. Secreted proteins can generally be isolated using standard procedures to purify the proteins from the cell culture medium.

D. Standard Protein Separation Techniques For Purifying Proteins 1. Solubility Fractionation [86] Often as an initial step, and if the protein mixture is complex, an initial salt fractionation can separate many of the unwanted host cell proteins (or proteins derived from the cell culture media) from the recombinant protein of interest. The preferred salt is ammonium sulfate. Ammonium sulfate precipitates proteins by effectively reducing the amount of water in the protein mixture. Proteins then precipitate on the basis of their solubility. The more hydrophobic a protein is, the more likely it is to precipitate at lower ammonium sulfate concentrations. A typical protocol is to add saturated ammonium sulfate to a protein solution so that the resultant ammonium sulfate concentration is between 20- 30%. This will precipitate the most hydrophobic proteins. The precipitate is discarded (unless the protein of interest is hydrophobic) and ammonium sulfate is added to the supernatant to a concentration known to precipitate the protein of interest. The precipitate is then solubilized in buffer and the excess salt removed if necessary, through either dialysis or diafiltration. Other methods that rely on solubility of proteins, such as cold ethanol precipitation, are well known to those of skill in the art and can be used to fractionate complex protein mixtures. 2. Size Differential Filtration [87] Based on a calculated molecular weight, a protein of greater and lesser size can be isolated using ultrafiltration through membranes of different pore sizes (for example, Amicon or Millipore membranes). As a first step, the protein mixture is ultrafiltered through a membrane with a pore size that has a lower molecular weight cut-off than the molecular weight of the protein of interest. The retentate of the ultrafiltration is then ultrafiltered against a membrane with a molecular cut off greater than the molecular weight of the protein of interest. The recombinant protein will pass through the membrane into the filtrate. The filtrate can then be chromatographed as described below. 3. Column Chromatography [88] The proteins of interest can also be separated from other proteins on the basis of their size, net surface charge, hydrophobicity and affinity for ligands. hi addition, antibodies raised against proteins can be conjugated to column matrices and the proteins immunopurified. All of these methods are well known in the art. [89] Immunoaffinity chromatography using antibodies raised to a variety of affinity tags such as hemagglutinin (HA), FLAG, Xpress, Myc, hexahistidine (His), glutathione S transferase (GST) and the like can be used to purify polypeptides. The His tag will also act as a chelating agent for certain metals (e.g., Ni) and thus the metals can also be used to purify His-containing polypeptides. After purification, the tag is optionally removed by specific proteolytic cleavage. [90] It will be apparent to one of skill that chromatographic techniques can be performed at any scale and using equipment from many different manufacturers (e.g., Pharmacia Biotech).

IV. DETECTION OF POLYNUCLEOTIDES OF THE INVENTION

[91] Those of skill in the art will recognize that detection of expression of polynucleotides and polypeptides of the invention has many uses. For example, as discussed herein, detection of levels of polynucleotides and polypeptides of the invention in a patient is useful for diagnosing diabetes or a predisposition for at least some of the pathological effects of diabetes. Moreover, detection of gene expression is useful to identify modulators of expression of polynucleotides and polypeptides of the invention. [92] A variety of methods of specific DNA and RNA measurement that use nucleic acid hybridization techniques are known to those of skill in the art (see, Sambrook, supra). Some methods involve an electrophoretic separation (e.g., Southern blot for detecting DNA, and Northern blot for detecting RNA), but measurement of DNA and RNA can also be carried out in the absence of electrophoretic separation (e.g., by dot blot). Southern blot of genomic DNA (e.g., from a human) can be used for screening for restriction fragment length polymorphism (RFLP) to detect the presence of a genetic disorder affecting a polypeptide of the invention. [93] The selection of a nucleic acid hybridization format is not critical. A variety of nucleic acid hybridization formats are known to those skilled in the art. For example, common formats include sandwich assays and competition or displacement assays. Hybridization techniques are generally described in Hames and Higgins Nucleic Acid Hybridization, A Practical Approach, IRL Press (1985); Gall and Pardue, Proc. Natl. Acad. Sd. U.S.A., 63:378-383 (1969); and John et al. Nature, 223:582-587 (1969). [94] Detection of a hybridization complex may require the binding of a signal-generating complex to a duplex of target and probe polynucleotides or nucleic acids. Typically, such binding occurs through ligand and anti-ligand interactions as between a ligand-conjugated probe and an anti-ligand conjugated with a signal. The binding of the signal generation complex is also readily amenable to accelerations by exposure to ultrasonic energy. [95] The label may also allow indirect detection of the hybridization complex. For example, where the label is a hapten or antigen, the sample can be detected by using antibodies. In these systems, a signal is generated by attaching fluorescent or enzyme molecules to the antibodies or in some cases, by attachment to a radioactive label (see, e.g., Tijssen, "Practice and Theory of Enzyme Immunoassays," Laboratory Techniques in Biochemistry and Molecular Biology, Burdon and van Knippenberg Eds., Elsevier (1985), pp. 9-20). [96] The probes are typically labeled either directly, as with isotopes, chromophores, lumiphores, chromogens, or indirectly, such as with biotin, to which a streptavidin complex may later bind. Thus, the detectable labels used in the assays of the present invention can be primary labels (where the label comprises an element that is detected directly or that produces a directly detectable element) or secondary labels (where the detected label binds to a primary label, e.g., as is common in immunological labeling). Typically, labeled signal nucleic acids are used to detect hybridization. Complementary nucleic acids or signal nucleic acids may be labeled by any one of several methods typically used to detect the presence of hybridized polynucleotides. The most common method of detection is the use of autoradiography with 3H, 1251, 35S, 14C, or 32P -labeled probes or the like. [97] Other labels include, e.g., ligands that bind to labeled antibodies, fluorophores, chemiluminescent agents, enzymes, and antibodies that can serve as specific binding pair members for a labeled ligand. An introduction to labels, labeling procedures and detection of labels is found in Polak and Van Noorden Introduction to Immunocytochemistry, 2nd ed., Springer Verlag, NY (1997); and in Haugland Handbook of Fluorescent Probes and Research Chemicals, a combined handbook and catalogue Published by Molecular Probes, Inc. (1996). [98] In general, a detector that monitors a particular probe or probe combination is used to detect the detection reagent label. Typical detectors include spectrophotometers, phototubes and photodiodes, microscopes, scintillation counters, cameras, film and the like, as well as combinations thereof. Examples of suitable detectors are widely available from a variety of commercial sources known to persons of skill in the art. Commonly, an optical image of a substrate comprising bound labeling moieties is digitized for subsequent computer analysis. [99] The amount of, for example, an RNA is measured by quantitating the amount of label fixed to the solid support by binding of the detection reagent. Typically, the presence of a modulator during incubation will increase or decrease the amount of label fixed to the solid support relative to a control incubation that does not comprise the modulator, or as compared to a baseline established for a particular reaction type. Means of detecting and quantitating labels are well known to those of skill in the art. [100] In some embodiments, the target nucleic acid or the probe is immobilized on a solid support. Solid supports suitable for use in the assays of the invention are known to those of skill in the art. As used herein, a solid support is a matrix of material in a substantially fixed arrangement. [101] A variety of automated solid-phase assay techniques are also appropriate. For instance, very large scale immobilized polymer arrays (VLSIPS™), i.e. Gene Chips or microarrays, available from Affymetrix, Inc. in Santa Clara, CA can be used to detect changes in expression levels of a plurality of genes involved in the same regulatory pathways simultaneously. See, Tijssen, supra., Fodor etal. (1991) Science, 251: 767- 777; Sheldon etal. (1993) Clinical Chemistry 39(4): 718-719, and Kozal et al (\996)Nature Medicine 2(7): 753-759. Similarly, spotted cDNA arrays (arrays of cDNA sequences bound to nylon, glass or another solid support) can also be used to monitor expression of a plurality of genes. [102] Typically, the array elements are organized in an ordered fashion so that each element is present at a specified location on the substrate. Because the array elements are at specified locations on the substrate, the hybridization patterns and intensities (which together create a unique expression profile) can be interpreted in terms of expression levels of particular genes and can be correlated with a particular disease or condition or treatment. See, e.g., Schena etal, Science 270: 467-470 (1995)) and (Lockhart et al, Nature Biotech. 14: 1675-1680 (1996)). [103] Hybridization specificity can be evaluated by comparing the hybridization of specificity-control polynucleotide sequences to specificity-control polynucleotide probes that are added to a sample in a known amount. The specificity-control target polynucleotides may have one or more sequence mismatches compared with the corresponding polynucleotide sequences, hi this manner, whether only complementary target polynucleotides are hybridizing to the polynucleotide sequences or whether mismatched hybrid duplexes are forming is determined. [104] Hybridization reactions can be performed in absolute or differential hybridization formats. In the absolute hybridization format, polynucleotide probes from one sample are hybridized to the sequences in a microarray format and signals detected after hybridization complex formation correlate to polynucleotide probe levels in a sample. In the differential hybridization format, the differential expression of a set of genes in two biological samples is analyzed. For differential hybridization, polynucleotide probes from both biological samples are prepared and labeled with different labeling moieties. A mixture of the two labeled polynucleotide probes is added to a microarray. The microarray is then examined under conditions in which the emissions from the two different labels are individually detectable. Sequences in the microarray that are hybridized to substantially equal numbers of polynucleotide probes derived from both biological samples give a distinct combined fluorescence (Shalon et al. PCT publication WO95/35505). In some embodiments, the labels are fluorescent labels with distinguishable emission spectra, such as Cy3 and Cy5 fluorophores. [105] After hybridization, the microarray is washed to remove nonhybridized nucleic acids and complex formation between the hybridizable array elements and the polynucleotide probes is detected. Methods for detecting complex formation are well known to those skilled in the art. In some embodiments, the polynucleotide probes are labeled with a fluorescent label and measurement of levels and patterns of fluorescence indicative of complex formation is accomplished by fluorescence microscopy, such as confocal fluorescence microscopy. [106] In a differential hybridization experiment, polynucleotide probes from two or more different biological samples are labeled with two or more different fluorescent labels with different emission wavelengths. Fluorescent signals are detected separately with different photomultipliers set to detect specific wavelengths. The relative abundances/expression levels of the polynucleotide probes in two or more samples are obtained. [107] Typically, microarray fluorescence intensities can be normalized to take into account variations in hybridization intensities when more than one microarray is used under similar test conditions. In some embodiments, individual polynucleotide probe/target complex hybridization intensities are normalized using the intensities derived from internal normalization controls contained on each microarray. [108] Detection of nucleic acids can also be accomplished, for example, by using a labeled detection moiety that binds specifically to duplex nucleic acids (e.g., an antibody that is specific for RNA-DNA duplexes). One example uses an antibody that recognizes DNA-RNA heteroduplexes in which the antibody is linked to an enzyme (typically by recombinant or covalent chemical bonding). The antibody is detected when the enzyme reacts with its substrate, producing a detectable product. Coutlee et al. (1989) Analytical Biochemistry 181:153-162; Bogulavski (1986) et al. J. Immunol. Methods 89:123- 130; Prooijen-Knegt (1982) Exp. Cell Res. 141:397-407; Rudkin (1976) Nature 265:472-473, Stollar (1970) PNAS 65:993-1000; Ballard (1982) MoI. Immunol. 19:793-799; Pisetsky and Caster (1982) MoI. Immunol. 19:645-650; Viscidi et al. (1988) J. Clin. Microbial. 41:199- 209; and Kiney et al. (1989) J. Clin. Microbiol. 27:6-12 describe antibodies to RNA duplexes, including homo and heteroduplexes. Kits comprising antibodies specific for DNA:RNA hybrids are available, e.g., from Digene Diagnostics, Inc. (Beltsville, MD). [109] In addition to available antibodies, one of skill in the art can easily make antibodies specific for nucleic acid duplexes using existing techniques, or modify those antibodies that are commercially or publicly available. In addition to the art referenced above, general methods for producing polyclonal and monoclonal antibodies are known to those of skill in the art (see, e.g., Paul (ed) Fundamental Immunology, Third Edition Raven Press, Ltd., NY (1993); Coligan Current Protocols in Immunology Wiley/Greene, NY (1991); Harlow and Lane Antibodies: A Laboratory Manual Cold Spring Harbor Press, NY (1989); Stites et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, CA, and references cited therein; Goding Monoclonal Antibodies: Principles and Practice (2d ed.) Academic Press, New York, NY, (1986); and Kohler and Milstein Nature 256: 495-497 (1975)). Other suitable techniques for antibody preparation include selection of libraries of recombinant antibodies in phage or similar vectors (see, Huse et al. Science 246:1275-1281 (1989); and Ward et al. Nature 341 :544-546 (1989)). Specific monoclonal and polyclonal antibodies and antisera will usually bind with a KD of at least about 0.1 μM, preferably at least about 0.01 μM or better, and most typically and preferably, 0.001 μM or better. [110] The nucleic acids used in this invention can be either positive or negative probes. Positive probes bind to their targets and the presence of duplex formation is evidence of the presence of the target. Negative probes fail to bind to the suspect target and the absence of duplex formation is evidence of the presence of the target. For example, the use of a wild type specific nucleic acid probe or PCR primers may serve as a negative probe in an assay sample where only the nucleotide sequence of interest is present. [Ill] The sensitivity of the hybridization assays may be enhanced through use of a nucleic acid amplification system that multiplies the target nucleic acid being detected. Examples of such systems include the polymerase chain reaction (PCR) system and the ligase chain reaction (LCR) system. Other methods recently described in the art are the nucleic acid sequence based amplification (NASBA, Cangene, Mississauga, Ontario) and Q Beta Replicase systems. These systems can be used to directly identify mutants where the PCR or LCR primers are designed to be extended or ligated only when a selected sequence is present. Alternatively, the selected sequences can be generally amplified using, for example, nonspecific PCR primers and the amplified target region later probed for a specific sequence indicative of a mutation. It is understood that various detection probes, including Taqman and molecular beacon probes can be used to monitor amplification reaction products, e.g., in real time. [112] An alternative means for determining the level of expression of the nucleic acids of the present invention is in situ hybridization. In situ hybridization assays are well known and are generally described in Angerer et al., Methods Enzymol. 152:649-660 (1987). In an in situ hybridization assay, cells, preferentially human cells from the cerebellum or the hippocampus, are fixed to a solid support, typically a glass slide. IfDNA is to be probed, the cells are denatured with heat or alkali. The cells are then contacted with a hybridization solution at a moderate temperature to permit annealing of specific probes that are labeled. The probes are preferably labeled with radioisotopes or fluorescent reporters. [113] Single nucleotide polymorphism (SNP) analysis is also useful for detecting differences between alleles of the polynucleotides (e.g., genes) of the invention. SNPs linked to genes encoding polypeptides of the invention are useful, for instance, for diagnosis of diseases (e.g., diabetes) whose occurrence is linked to the gene sequences of the invention. For example, if an individual carries at least one SNP linked to a disease- associated allele of the gene sequences of the invention, the individual is likely predisposed for one or more of those diseases. If the individual is homozygous for a disease-linked SNP, the individual is particularly predisposed for occurrence of that disease (e.g., diabetes). In some embodiments, the SNP associated with the gene sequences of the invention is located within 300,000; 200,000; 100,000; 75,000; 50,000; or 10,000 base pairs from the gene sequence. [114] Various real-time PCR methods including, e.g., Taqman or molecular beacon-based assays (e.g., U.S. Patent Nos. 5,210,015; 5,487,972; Tyagi et al., Nature Biotechnology 14:303 (1996); and PCT WO 95/13399 are useful to monitor for the presence of absence of a SNP. Additional SNP detection methods include, e.g., DNA sequencing, sequencing by hybridization, dot blotting, oligonucleotide array (DNA Chip) hybridization analysis, or are described in, e.g., U.S. Patent No. 6,177,249; Landegren et al, Genome Research, 8:769-776 (1998); Botstein et al, Am J Human Genetics 32:314-331 (1980); Meyers et al, Methods in Enzymology 155:501-527 (1987); Keen et al, Trends in Genetics 7:5 (1991); Myers et al, Science 230:1242-1246 (1985); and Kwok et al., Genomics 23:138- 144 (1994).

V. IMMUNOLOGICAL DETECTION OF POLYPEPTIDES OF THE INVENTION [115] hi addition to the detection of polynucloetides of the invention and gene expression using nucleic acid hybridization technology, one can also use immunoassays to detect polypeptides of the invention. Immunoassays can be used to qualitatively or quantitatively analyze polypeptides of the invention. A general overview of the applicable technology can be found in Harlow & Lane, Antibodies: A Laboratory Manual (1988).

A. Antibodies to Target Proteins or other immunogens [116] Methods for producing polyclonal and monoclonal antibodies that react specifically with a protein of interest or other immunogen are known to those of skill in the art {see, e.g., Coligan, supra; and Harlow and Lane, supra; Stites et al, supra and references cited therein; Goding, supra; and Kohler and Milstein Nature, 256:495-497 (1975)). Such techniques include antibody preparation by selection of antibodies from libraries of recombinant antibodies in phage or similar vectors {see, Huse et al, supra; and Ward et al, supra). For example, in order to produce antisera for use in an immunoassay, the protein of interest or an antigenic fragment thereof, is isolated as described herein. For example, a recombinant protein is produced in a transformed cell line. An inbred strain of mice or rabbits is immunized with the protein using a standard adjuvant, such as Freund's adjuvant, and a standard immunization protocol. Alternatively, a synthetic peptide derived from the sequences disclosed herein is conjugated to a carrier protein and used as an immunogen. [117] Polyclonal sera are collected and titered against the immunogen in an immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support. Polyclonal antisera with a titer of 104 or greater are selected and tested for their crossreactivity against proteins other than the polypeptides of the invention or even other homologous proteins from other organisms, using a competitive binding immunoassay. Specific monoclonal and polyclonal antibodies and antisera will usually bind with a KD of at least about 0.1 mM, more usually at least about 1 μM, preferably at least about 0.1 μM or better, and most preferably, 0.01 μM or better. [118] A number of proteins of the invention comprising immunogens may be used to produce antibodies specifically or selectively reactive with the proteins of interest. Recombinant protein is the preferred immunogen for the production of monoclonal or polyclonal antibodies. Naturally occurring protein may also be used either in pure or impure form. Synthetic peptides made using the protein sequences described herein may also be used as an immunogen for the production of antibodies to the protein. Recombinant protein can be expressed in eukaryotic or prokaryotic cells and purified as generally described supra. The product is then injected into an animal capable of producing antibodies. Either monoclonal or polyclonal antibodies may be generated for subsequent use in immunoassays to measure the protein. [119] Methods of production of polyclonal antibodies are known to those of skill in the art. In brief, an irnmunogen, preferably a purified protein, is mixed with an adjuvant and animals are immunized. The animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to polypeptides of the invention. When appropriately high titers of antibody to the immunogen are obtained, blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the protein can be done if desired (see, Harlow and Lane, supra). [120] Monoclonal antibodies may be obtained using various techniques familiar to those of skill in the art. Typically, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see, Kohler and Milstein, Eur. J. Immunol. 6:511-519 (1976)). Alternative methods of immortalization include, e.g., transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host. Alternatively, one may isolate DNA sequences that encode a monoclonal antibody or a binding fragment thereof by screening a DNA library from human B cells according to the general protocol outlined by Huse et al., supra. [121] Once target immunogen-specifϊc antibodies are available, the immunogen can be measured by a variety of immunoassay methods with qualitative and quantitative results available to the clinician. For a review of immunological and immunoassay procedures in general see, Stites, supra. Moreover, the immunoassays of the present invention can be performed in any of several configurations, which are reviewed extensively in Maggio Enzyme Immunoassay, CRC Press, Boca Raton, Florida (1980); Tijssen, supra; and Harlow and Lane, supra. [122] Immunoassays to measure target proteins in a human sample may use a polyclonal antiserum that was raised to full-length polypeptides of the invention or a fragment thereof. This antiserum is selected to have low cross-reactivity against other proteins and any such cross-reactivity is removed by immunoabsorption prior to use in the immunoassay.

B. Immunological Binding Assays [123] In some embodiments, a protein of interest is detected and/or quantified using any of a number of well-known immunological binding assays (see, e.g., U.S. Patents 4,366,241; 4,376,110; 4,517,288; and 4,837,168). For a review of the general immunoassays, see also Asai Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. NY (1993); Stites, supra. Immunological binding assays (or immunoassays) typically utilize a "capture agent" to specifically bind to and often immobilize the analyte (e.g., full-length polypeptides of the present invention, or antigenic subsequences thereof). The capture agent is a moiety that specifically binds to the analyte. The antibody may be produced by any of a number of means well known to those of skill in the art and as described above. [124] Immunoassays also often utilize a labeling agent to bind specifically to and label the binding complex formed by the capture agent and the analyte. The labeling agent may itself be one of the moieties comprising the antibody/analyte complex. Alternatively, the labeling agent may be a third moiety, such as another antibody, that specifically binds to the antibody/protein complex. [125] In a preferred embodiment, the labeling agent is a second antibody bearing a label. Alternatively, the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. The second antibody can be modified with a detectable moiety, such as biotin, to which a third labeled molecule can specifically bind, such as enzyme-labeled streptavidin. [126] Other proteins capable of specifically binding immunoglobulin constant regions, such as protein A or protein G, can also be used as the label agents. These proteins are normal constituents of the cell walls of streptococcal bacteria. They exhibit a strong non-immunogenic reactivity with immunoglobulin constant regions from a variety of species (see, generally, Kronval, et al. J. Immunol, 111 : 1401 -1406 (1973); and Akerstrom, et al l Immunol, 135:2589-2542 (1985)). [127] Throughout the assays, incubation and/or washing steps may be required after each combination of reagents. Incubation steps can vary from about 5 seconds to several hours, preferably from about 5 minutes to about 24 hours. The incubation time will depend upon the assay format, analyte, volume of solution, concentrations, and the like. Usually, the assays will be carried out at ambient temperature, although they can be conducted over a range of temperatures, such as 10°C to 400C. 1. Non-Competitive Assay Formats [128] Immunoassays for detecting proteins or analytes of interest from tissue samples may be either competitive or noncompetitive. Noncompetitive immunoassays are assays in which the amount of captured protein or analyte is directly measured. In one preferred "sandwich" assay, for example, the capture agent (e.g., antibodies specific for the polypeptides of the invention) can be bound directly to a solid substrate where it is immobilized. These immobilized antibodies then capture the polypeptide present in the test sample. The polypeptide of the invention thus immobilized is then bound by a labeling agent, such as a second labelled antibody specific for the polypeptide. Alternatively, the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. The second can be modified with a detectable moiety, such as biotin, to which a third labeled molecule can specifically bind, such as enzyme-labeled streptavidin. 2. Competitive Assay Formats [129] In competitive assays, the amount of protein or analyte present in the sample is measured indirectly by measuring the amount of an added (exogenous) protein or analyte displaced (or competed away) from a specific capture agent (e.g., antibodies specific for a polypeptide of the invention) by the protein or analyte present in the sample. The amount of immunogen bound to the antibody is inversely proportional to the concentration of immunogen present in the sample. In a particularly preferred embodiment, the antibody is immobilized on a solid substrate. The amount of analyte may be detected by providing a labeled analyte molecule. It is understood that labels can include, e.g., radioactive labels as well as peptide or other tags that can be recognized by detection reagents such as antibodies. [130] Immunoassays in the competitive binding format can be used for cross- reactivity determinations. For example, the protein encoded by the sequences described herein can be immobilized on a solid support. Proteins are added to the assay and compete with the binding of the antisera to the immobilized antigen. The ability of the above proteins to compete with the binding of the antisera to the immobilized protein is compared to that of the protein encoded by any of the sequences described herein. The percent cross-reactivity for the above proteins is calculated, using standard calculations. Those antisera with less than 10% cross-reactivity with each of the proteins listed above are selected and pooled. The cross-reacting antibodies are optionally removed from the pooled antisera by immunoabsorption with the considered proteins, e.g., distantly related homologs. [131] The immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second protein, thought to be perhaps a protein of the present invention, to the immunogen protein. In order to make this comparison, the two proteins are each assayed at a wide range of concentrations and the amount of each protein required to inhibit 50% of the binding of the antisera to the immobilized protein is determined. If the amount of the second protein required is less than 10 times the amount of the protein partially encoded by a sequence herein that is required, then the second protein is said to specifically bind to an antibody generated to an immunogen consisting of the target protein. 3. Other Assay Formats [132] In some embodiments, western blot (immunoblot) analysis is used to detect and quantify the presence of a polypeptide of the invention in the sample. The technique generally comprises separating sample proteins by gel electrophoresis on the basis of molecular weight, transferring the separated proteins to a suitable solid support (such as, e.g., a nitrocellulose filter, a nylon filter, or a derivatized nylon filter) and incubating the sample with the antibodies that specifically bind the protein of interest. For example, antibodies are selected that specifically bind to the polypeptides of the invention on the solid support. These antibodies may be directly labeled or alternatively may be subsequently detected using labeled antibodies (e.g., labeled sheep anti-mouse antibodies) that specifically bind to the antibodies against the protein of interest. [133] Other assay formats include liposome immunoassays (LIA), which use liposomes designed to bind specific molecules (e.g., antibodies) and release encapsulated reagents or markers. The released chemicals are then detected according to standard techniques (see, Monroe et al. (1986) Amer. Clin. Prod. Rev. 5:34-41). 4. Labels [134] The particular label or detectable group used in the assay is not a critical aspect of the invention, as long as it does not significantly interfere with the specific binding of the antibody used in the assay. The detectable group can be any material having a detectable physical or chemical property. Such detectable labels have been well-developed in the field of immunoassays and, in general, most labels useful in such methods can be applied to the present invention. Thus, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Useful labels in the present invention include magnetic beads (e.g., Dynabeads™), fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 1251, 35S, 14C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. [135] The label may be coupled directly or indirectly to the desired component of the assay according to methods well known in the art. As indicated above, a wide variety of labels may be used, with the choice of label depending on the sensitivity required, the ease of conjugation with the compound, stability requirements, available instrumentation, and disposal provisions. [136] Non-radioactive labels are often attached by indirect means. The molecules can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorescent compound. A variety of enzymes and fluorescent compounds can be used with the methods of the present invention and are well-known to those of skill in the art (for a review of various labeling or signal producing systems which may be used, see, e.g., U.S. Patent No. 4,391,904). [137] Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter or photographic film as in autoradiography. Where the label is a fluorescent label, it may be detected by exciting the fiuorochrome with the appropriate wavelength of light and detecting the resulting fluorescence. The fluorescence may be detected visually, by means of photographic film, by the use of electronic detectors such as charge coupled devices (CCDs) or photomultipliers and the like. Similarly, enzymatic labels may be detected by providing the appropriate substrates for the enzyme and detecting the resulting reaction product. Finally simple colorimetric labels may be detected directly by observing the color associated with the label. Thus, in various dipstick assays, conjugated gold often appears pink, while various conjugated beads appear the color of the bead. [138] Some assay formats do not require the use of labeled components. For instance, agglutination assays can be used to detect the presence of the target antibodies. In this case, antigen-coated particles are agglutinated by samples comprising the target antibodies. In this format, none of the components need to be labeled and the presence of the target antibody is detected by simple visual inspection. VI. IDENTIFICATION OF MODULATORS OF POLYPEPTIDES OF THE INVENTION [139] Modulators of a polypeptide of the invention, i.e. agonists or antagonists of a polypeptide's activity, or polypeptide's or polynucleotide's expression of full-length polypetides of the invention or fragments thereof, are useful for treating a number of human diseases, including diabetes. For example, administration of modulators can be used to treat diabetic patients or prediabetic individuals to prevent progression, and therefore symptoms, associated with diabetes (including insulin resistance).

A. Agents that Modulate Polypeptides of the Invention [140] The agents tested as modulators of polypeptides of the invention can be any small chemical compound, or a biological entity, such as a protein, sugar, nucleic acid or lipid. Essentially any chemical compound can be used as a potential modulator or ligand in the assays of the invention, although most often compounds that can be dissolved in aqueous or organic (especially DMSO-based) solutions are used. Modulators include agents designed to reduce the level of mRNA encoding a polypeptide of the invention (e.g. antisense molecules, ribozymes, DNAzymes, small inhibitory RNAs and the like) or the level of translation from an mRNA (e.g., translation blockers such as an antisense molecules that are complementary to translation start or other sequences on an mRNA molecule). Modulators of the invention also include antibodies that specific bind to and/or inhibit or activate the polypeptides of the invention. Other modulators include the polypeptides of the invention themselves, fragments thereof, or fusion proteins comprising the polypeptides or fragments thereof (e.g., in some embodiments, comprising at least 25, 50, or 100 amino acids of the polypeptide). For polypeptides of the invention that are receptors, soluble fragments of the polypeptides (i.e., lacking a transmembrane domain) can act as modulators of polypeptide signaling activity. For polypeptides of the invention that are secreted, both full length and fragments with biological activity can act as modulators. It will be appreciated that there are many suppliers of chemical compounds, including Sigma (St. Louis, MO), Aldrich (St. Louis, MO), Sigma- Aldrich (St. Louis, MO), Fluka Chemika-Biochemica Analytika (Buchs, Switzerland) and the like. [141] In some embodiments, high throughput screening methods involve providing a combinatorial chemical or peptide library containing a large number of potential therapeutic compounds (potential modulator compounds). Such "combinatorial chemical libraries" or "ligand libraries" are then screened in one or more assays, as described herein, to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional "lead compounds" or can themselves be used as potential or actual therapeutics. [142] A combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis, by combining a number of chemical "building blocks" such as reagents. For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in every possible way for a given compound length {i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks. [143] Preparation and screening of combinatorial chemical libraries is well known to those of skill in the art. Such combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Patent 5,010,175, Furka, Int. J. Pept. Prot. Res. 37:487-493 (1991) and Houghton et al, Nature 354:84-88 (1991)). Other chemistries for generating chemical diversity libraries can also be used. Such chemistries include, but are not limited to: peptoids (e.g., PCT Publication No. WO 91/19735), encoded peptides (e.g., PCT Publication WO 93/20242), random bio-oligomers (e.g., PCT Publication No. WO 92/00091), benzodiazepines (e.g., U.S. Pat. No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al, Proc. Nat. Acad. ScL USA 90:6909-6913 (1993)), vinylogous polypeptides (Hagihara et al, J. Amer. Chem. Soc. 114:6568 (1992)), nonpeptidal peptidomimetics with glucose scaffolding (Hirschmann et al, J. Amer. Chem. Soc. 114:9217-9218 (1992)), analogous organic syntheses of small compound libraries (Chen et al, J. Amer. Chem. Soc. 116:2661 (1994)), oligocarbamates (Cho et al, Science 261 :1303 (1993)), and/or peptidyl phosphonates (Campbell et al, J. Org. Chem. 59:658 (1994)), nucleic acid libraries (see Ausubel, Berger and Sambrook, all supra), peptide nucleic acid libraries (see, e.g., U.S. Patent 5,539,083), antibody libraries (see, e.g., V raughn et al, Nature Biotechnology, 14(3):309-314 (1996) and PCT/US96/10287), carbohydrate libraries (see, e.g., Liang et al, Science, 274:1520-1522 (1996) and U.S. Patent 5,593,853), small organic molecule libraries (see, e.g., benzodiazepines, Baum C&EN, Jan 18, page 33 (1993); isoprenoids, U.S. Patent 5,569,588; thiazolidinones and metathiazanones, U.S. Patent 5,549,974; pyrrolidines, U.S. Patents 5,525,735 and 5,519,134; morpholino compounds, U.S. Patent 5,506,337; benzodiazepines, 5,288,514, and the like). [144] Devices for the preparation of combinatorial libraries are commercially available (see, e.g., 357 MPS, 390 MPS, Advanced Chem Tech, Louisville KY, Symphony, Rainin, Woburn, MA, 433 A Applied Biosystems, Foster City, CA, 9050 Plus, Millipore, Bedford, MA). In addition, numerous combinatorial libraries are themselves commercially available {see, e.g., ComGenex, Princeton, NJ., Tripos, Inc., St. Louis, MO, 3D Pharmaceuticals, Exton, PA, Martek Biosciences, Columbia, MD, etc.).

B. Methods of Screening for Modulators of the Polypeptides of the Invention [145] A number of different screening protocols can be utilized to identify agents that modulate the level of expression or activity of a polynucleotide of a polypeptide of the invention in cells, particularly mammalian cells, and especially human cells. In general terms, the screening methods involve screening a plurality of agents to identify an agent that modulates the activity of a polypeptide of the invention by, e.g., binding to the polypeptide, preventing an inhibitor or activator from binding to the polypeptide, increasing association of an inhibitor or activator with the polypeptide, or activating or inhibiting expression of the polypeptide. [146] Any cell expressing a full-length polypeptide of the invention or a fragment thereof can be used to identify modulators. In some embodiments, the cells are eukaryotic cells lines (e.g., CHO or HEK293) transformed to express a heterologous polypeptide of the invention. In some embodiments, a cell expressing an endogenous polypeptide of the invention is used in screens. In other embodiments, modulators are screened for their ability to effect insulin responses. In other embodiments, modulators are screened for their ability to effect lipid metabolism. [147] In some embodiments, modulators of BC001573 (e.g., comprising the amino acid sequence of SEQ ID NO: 2, 4 or 6), maybe identified using, e.g., expression assays, promoter-reporter assays, or modulator binding assays. [148] In some embodiments, modulators of CILP (e.g., comprising the amino acid sequence of SEQ ID NO: 8, 9, 10, 12 or 14), maybe identified using, e.g., expression assays, promoter-reporter assays, or modulator binding assays. [149] In some embodiments, modulators of FLJ45434 (e.g., comprising the amino acid sequence of SEQ ID NO: 16), maybe identified using, e.g., expression assays, promoter-reporter assays, or modulator binding assays. Kinase assays can be carried out after contacting either purified recombinant FLJ45434 protein, or an intact cell with the modulator using e.g. the generic peptide casein or myosin as substrates. [150] In some embodiments, modulators of UBE2E3 (e.g., comprising the amino acid sequence of SEQ ID NO: 18, 20 or 22), may be identified using, e.g., expression assays, promoter-reporter assays, or modulator binding assays or enzyme assays. Enzyme assays can be carried out after contacting either purified recombinant UBE2E3 protein, or an intact cell with a modulator using conjugation of one or more molecules of ubiquitin to target proteins. Binding assays can be carried out after contacting either purified recombinant UBE2E3 protein, or an intact cell with a modulator using e.g. Nedd4-2 as an interacting protein [151] In some embodiments, modulators of USP38 (e.g., comprising the amino acid sequence of SEQ ID NO: 24, 26 or 28), may be identified using, e.g., expression assays, promoter-reporter assays, or modulator binding assays, or enzyme assays. Enzyme assays can be carried out after contacting either purified recombinant USP38 protein or an intact cell with a modulator using e.g. an polyubiquinated peptide as a substrate.

1. Polypeptide Binding Assays [152] Preliminary screens can be conducted by screening for agents capable of binding to polypeptides of the invention, as at least some of the agents so identified are likely modulators of a polypeptide of the invention. Binding assays are also useful, e.g., for identifying endogenous proteins that interact with polypeptides of the invention. For example, antibodies, receptors or other molecules that bind polypeptides of the invention can be identified in binding assays. [153] Binding assays usually involve contacting a polypeptide of the invention with one or more test agents and allowing sufficient time for the protein and test agents to form a binding complex. Any binding complexes formed can be detected using any of a number of established analytical techniques. Protein binding assays include, but are not limited to, methods that measure co-precipitation or co-migration on non-denaturing SDS- polyacrylamide gels, and co-migration on Western blots (see, e.g., Bennet, J.P. and Yamamura, H.I. (1985) "Neurotransmitter, Hormone or Drug Receptor Binding Methods," in Neurotransmitter Receptor Binding (Yamamura, H. L, etal., eds.), pp. 61-89. Other binding assays involve the use of mass spectrometry or NMR techniques to identify molecules bound to a polypeptide of the invention or displacement of labeled substrates. The polypeptides of the invention utilized in such assays can be naturally expressed, cloned or synthesized. [154] In addition, mammalian or yeast two-hybrid approaches {see, e.g., Bartel, P.L. et. al. Methods Enzymol, 254:241 (1995)) can be used to identify polypeptides or other molecules that interact or bind when expressed together in a host cell.

2. Polypeptide Activity [155] The activity of polypeptides of the invention can be assessed using a variety of in vitro and in vivo assays to determine functional, chemical, and physical effects, e.g., measuring ligand binding (e.g., radioactive or otherwise labeled ligand binding), second messengers (e.g., cAMP, cGMP, IP3, DAG, or Ca2+), ion flux, phosphorylation levels, transcription levels, and the like. Furthermore, such assays can be used to test for inhibitors and activators of the polypeptides of the invention. Modulators can also be genetically altered versions of polypeptides of the invention. [156] The polypeptide of the assay will be selected from a polypeptide with substantial identity to a sequence of SEQ ID NO: 2, 4, 6, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28 or other conservatively modified variants thereof. Generally, the amino acid sequence identity will be at least 70%, optionally at least 85%, optionally at least 90, or optionally at least 95% to the polypeptides exemplified herein. Optionally, the polypeptide of the assays will comprise a fragment of a polypeptide of the invention, such as an extracellular domain, transmembrane domain, cytoplasmic domain, ligand binding domain, subunit association domain, active site, and the like. Either a polypeptide of the invention or a domain thereof can be covalently linked to a heterologous protein to create a chimeric protein used in the assays described herein. [157] Modulators of polypeptide activity are tested using either recombinant or naturally occurring polypeptides of the invention. The protein can be isolated, expressed in a cell, expressed in a membrane derived from a cell, expressed in tissue or in an animal, either recombinant or naturally occurring. For example, tissue slices, dissociated cells, e.g., from tissues expressing polypeptides of the invention, transformed cells, or membranes can be used. Modulation is tested using one of the in vitro or in vivo assays described herein. [158] Modulator binding to polypeptides of the invention, a domain, or chimeric protein can be tested in solution, in a bilayer membrane, attached to a solid phase, in a lipid monolayer, or in vesicles. Binding of a modulator can be tested using, e.g., changes in spectroscopic characteristics (e.g., fluorescence, absorbance, refractive index), hydrodynamic (e.g., shape), chromatographic, or solubility properties. [159] Samples or assays that are treated with a potential moulator (e.g., a "test compound") are compared to control samples without the test compound, to examine the extent of modulation. Control samples (untreated with activators or inhibitors) are assigned a relative activity value of 100. Inhibition of the polypeptides of the invention is achieved when the activity value relative to the control is about 90%, optionally 50%, optionally 25- 0%. Activation of the polypeptides of the invention is achieved when the activity value relative to the control is 110%, optionally 150%, 200%, 300%, 400%, 500%, or 1000-2000%.

3. ' Expression Assays [160] Screening for a compound that modulates the expression of a polynucleotide or a polypeptide of the invention is also provided. Screening methods generally involve conducting cell-based assays in which test compounds are contacted with one or more cells expressing a polynucleotide or a polypeptide of the invention, and then detecting an increase or decrease in expression (either transcript or translation product). Assays can be performed with any cells that express a polynucleotide or a polypeptide of the invention. [161] Expression can be detected in a number of different ways. As described infra, the expression level of a polynucleotide of the invention in a cell can be determined by probing the mRNA expressed in a cell with a probe that specifically hybridizes with a transcript (or complementary nucleic acid derived therefrom) of a polynucleotide of the invention. Probing can be conducted by lysiiig the cells and conducting Northern blots or without lysing the cells using in szYw-hybridization techniques. Alternatively, a polypeptide of the invention can be detected using immunological methods in which a cell lysate is probed with antibodies that specifically bind to the polypeptide. [162] Promoter-reporter assays can be carried out using mammalian cells transfected with a reporter gene operably linked to sequences derived from the promoter regions of genes encoding the polypeptides of the invention. The increased or decreased expression of the reporter gene can be detected in the presence and absence of the modulator. Expression of reporter genes may be detected by hybridization to a complementary nucleic acid, by using an immunological reagent, by assaying for an activity of the reporter gene product, or other methods known to those in the art [163] The level of expression or activity of a polynucleotide or a polypeptide of the invention can be compared to a baseline value. The baseline value can be a value for a control sample or a statistical value that is representative of expression levels of a polynucleotide or a polypeptide of the invention for a control population (e.g., lean individuals as described herein) or cells (e.g., tissue culture cells not exposed to a modulator). Expression levels can also be determined for cells that do not express the polynucleotide or a polypeptide of the invention as a negative control. Such cells generally are otherwise substantially genetically the same as the test cells. [164] A variety of different types of cells can be utilized in the reporter assays. Cells that do not endogenously express a polypeptide of the invention can be prokaryotic, but are preferably eukaryotic. The eukaryotic cells can be any of the cells typically utilized in generating cells that harbor recombinant nucleic acid constructs. Exemplary eukaryotic cells include, but are not limited to, yeast, and various higher eukaryotic cells such as the HEK293, HeρG2, COS, CHO and HeLa cell lines. [165] Various controls can be conducted to ensure that an observed activity is authentic including running parallel reactions with cells that lack the reporter construct or by not contacting a cell harboring the reporter construct with test compound. Compounds can also be further validated as described below.

4. Validation [166] Agents that are initially identified by any of the foregoing screening methods can be further tested to validate the apparent activity. Modulators that are selected for further study can be tested on the "classic" insulin responsive cell line, mouse 3T3-L1 adipocytes, muscle cells such as L6 cells and the like. Cells (e.g., adipocytes or muscle cells) are pre-incubated with the modulators and tested for acute (up to 4 hours) and chronic (overnight) effects on basal and insulin-stimulated GLUT4 translocation and glucose uptake. [167] Following such studies, validity of the modulators is tested in suitable animal models. The basic format of such methods involves administering a lead compound identified during an initial screen to an animal that serves as a model for humans and then determining if expressionof activity of a polypeptide of the invention is in fact modulated. [168] The effect of the compound will be assessed in either diabetic animals or in diet induced insulin resistant animals. The blood glucose and insulin levels will be determined. The animal models utilized in validation studies generally are mammals of any kind. Specific examples of suitable animals include, but are not limited to, primates, mice and rats. For example, monogenic models of diabetes (e.g., ob/ob and db/db mice, Zucker rats and Zucker Diabetic Fatty rats etc) or polygenic models of diabetes (e.g., OLETF rats, GK rats, NSY mice, and KK mice) can be useful for validating modulation of a polypeptide of the invention in a diabetic or insulin resistant animal. In addition, transgenic animals expressing human polypeptides of the invention can be used to further validate drug candidates.

C. Solid Phase and Soluble High Throughput Assays [169] In the high throughput assays of the invention, it is possible to screen up to several thousand different modulators or ligands in a single day. In particular, each well of a microtiter plate can be used to run a separate assay against a selected potential modulator, or, if concentration or incubation time effects are to be observed, every 5-10 wells can test a single modulator. Thus, a single standard microtiter plate can assay about 100 (e.g., 96) modulators. If 1536 well plates are used, then a single plate can easily assay from about 100 to about 1500 different compounds. It is possible to assay several different plates per day; assay screens for up to about 6,000-20,000 or more different compounds are possible using the integrated systems of the invention. In addition, microfluidic approaches to reagent manipulation can be used. [170] A molecule of interest (e.g., a polypeptide or polynucleotide of the invention, or a modulator thereof) can be bound to the solid-state component, directly or indirectly, via covalent or non-covalent linkage, e.g., via a tag. The tag can be any of a variety of components. In general, a molecule that binds the tag (a tag binder) is fixed to a solid support, and the tagged molecule of interest is attached to the solid support by interaction of the tag and the tag binder. [171] A number of tags and tag binders can be used, based upon known molecular interactions well described in the literature. For example, where a tag has a natural binder, for example, biotin, protein A, or protein G, it can be used in conjunction with appropriate tag binders (avidin, streptavidin, neutravidin, the Fc region of an immunoglobulin, poly-His, etc.) Antibodies to molecules with natural binders such as biotin are also widely available and appropriate tag binders (see, SIGMA Immunochemicals 1998 catalogue SIGMA, St. Louis MO). [172] Similarly, any haptenic or antigenic compound can be used in combination with an appropriate antibody to form a tag/tag binder pair. Thousands of specific antibodies are commercially available and many additional antibodies are described in the literature. For example, in one common configuration, the tag is a first antibody and the tag binder is a second antibody that recognizes the first antibody. In addition to antibody- antigen interactions, receptor-ligand interactions are also appropriate as tag and tag-binder pairs, such as agonists and antagonists of cell membrane receptors (e.g., cell receptor-ligand interactions such as transferrin, c-kit, viral receptor ligands, cytokine receptors, chernokine receptors, interleukin receptors, immunoglobulin receptors and antibodies, the cadherin family, the integrin family, the selectin family, and the like; see, e.g., Pigott & Power, The Adhesion Molecule Facts Book I (1993)). Similarly, toxins and venoms, viral epitopes, hormones (e.g., opiates, steroids, etc.), intracellular receptors (e.g., which mediate the effects of various small ligands, including steroids, thyroid hormone, retinoids and vitamin D; peptides), drugs, lectins, sugars, nucleic acids (both linear and cyclic polymer configurations), oligosaccharides, proteins, phospholipids and antibodies can all interact with various cell receptors. ' [173] Synthetic polymers, such as polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, and polyacetates can also form an appropriate tag or tag binder. Many other tag/tag binder pairs are also useful in assay systems described herein, as would be apparent to one of skill upon review of this disclosure. [174] Common linkers such as peptides, polyethers, and the like can also serve as tags, and include polypeptide sequences, such as poly-gly sequences of between about 5 and 200 amino acids. Such flexible linkers are known to those of skill in the art. For example, poly(ethelyne glycol) linkers are available from Shearwater Polymers, Inc., Huntsville, Alabama. These linkers optionally have amide linkages, sulfhydryl linkages, or heterofunctional linkages. [175] Tag binders are fixed to solid substrates using any of a variety of methods currently available. Solid substrates are commonly derivatized or functionalized by exposing all or a portion of the substrate to a chemical reagent that fixes a chemical group to the surface that is reactive with a portion of the tag binder. For example, groups that are suitable for attachment to a longer chain portion would include amines, hydroxyl, thiol, and carboxyl groups. Amino alkylsilanes and hydroxyalkylsilanes can be used to functionalize a variety of surfaces, such as glass surfaces. The construction of such solid phase biopolymer arrays is well described in the literature (see, e.g., Merrifield, J. Am. Chem. Soc. 85:2149- 2154 (1963) (describing solid phase synthesis of, e.g., peptides); Geysen et al., J. Immun. Meth. 102:259-274 (1987) (describing synthesis of solid phase components on pins); Frank and Doring, Tetrahedron 44:60316040 (1988) (describing synthesis of various peptide sequences on cellulose disks); Fodor et al, Science, 251:161-111 (1991); Sheldon et al, Clinical Chemistry 39(4):718-719 (1993); and Kozal et al, Nature Medicine 2(7):753759 (1996) (all describing arrays of biopolymers fixed to solid substrates). Non-chemical approaches for fixing tag binders to substrates include other common methods, such as heat, cross-linking by UV radiation, and the like. [176] The invention provides in vitro assays for identifying, in a high throughput format, compounds that can modulate the expression or activity of a polypeptide of the invention. Control reactions that measure activity of a polypeptide of the invention in a cell in a reaction that does not include a potential modulator are optional, as the assays are highly uniform. Such optional control reactions are appropriate and increase the reliability of the assay. Accordingly, in some embodiments, the methods of the invention include such a control reaction. For each of the assay formats described, "no modulator" control reactions that do not include a modulator provide a background level of binding activity. [177] hi some assays it will be desirable to have positive controls. At least two types of positive controls are appropriate. First, a known activator of a polypeptide or a polynucleotide of the invention can be incubated with one sample of the assay, and the resulting increase in signal resulting from an increased expression level or activity of a polypeptide or a polynucleotide of the invention are determined according to the methods herein. Second, a known inhibitor of a polypeptide or a polynucleotide of the invention can be added, and the resulting decrease in signal for the expression or activity of a polypeptide or a polynucleotide of the invention can be similarly detected. It will be appreciated that modulators can also be combined with activators or inhibitors to find modulators that inhibit the increase or decrease that is otherwise caused by the presence of the known modulator of a polypeptide or a polynucleotide of the invention.

VII. COMPOSITIONS, KITS AND INTEGRATED SYSTEMS

[178] The invention provides compositions, kits and integrated systems for practicing the assays described herein using nucleic acids or polypeptides of the invention, antibodies, etc. [179] The invention provides assay compositions for use in solid phase assays; such compositions can include, for example, one or more nucleic acids encoding a polypeptide of the invention immobilized on a solid support, and a labeling reagent, hi each case, the assay compositions can also include additional reagents that are desirable for hybridization. Modulators of expression or activity of a polypeptide of the invention can also be included in the assay compositions. [180] The invention also provides kits for carrying out the assays of the invention. The kits typically include a probe that comprises an antibody that specifically binds to a polypeptide of the invention or a polynucleotide sequence encoding such polypeptides, and a label for detecting the presence of the probe. The kits may include at least one polynucleotide sequence encoding a polypeptide of the invention. Kits can include any of the compositions noted above, and optionally further include additional components such as instructions to practice a high-throughput method of assaying for an effect on expression of the genes encoding a polypeptide of the invention, or on activity of a polypeptide of the invention, one or more containers or compartments (e.g., to hold the probe, labels, or the like), a control modulator of the expression or activity of a polypeptide of the invention, a robotic armature for mixing kit components or the like. [181] The invention also provides integrated systems for high-throughput screening of potential modulators for an effect on the expression or activity of a polypeptide of the invention. The systems can include a robotic armature which transfers fluid from a source to a destination, a controller which controls the robotic armature, a label detector, a data storage unit which records label detection, and an assay component such as a microliter dish comprising a well having a reaction mixture or a substrate comprising a fixed nucleic acid or immobilization moiety. [182] A number of robotic fluid transfer systems are available, or can easily be made from existing components. For example, a Zymate XP (Zymark Corporation; Hopkinton, MA) automated robot using a Microlab 2200 (Hamilton; Reno, NV) pipetting station can be used to transfer parallel samples to 96 well microtiter plates to set up several parallel simultaneous binding assays. [183] Optical images viewed (and, optionally, recorded) by a camera or other recording device (e.g., a photodiode and data storage device) are optionally further processed in any of the embodiments herein, e.g., by digitizing the image and storing and analyzing the image on a computer. A variety of commercially available peripheral equipment and software is available for digitizing, storing and analyzing a digitized video or digitized optical image. [184] One conventional system carries light from the specimen field to a cooled charge-coupled device (CCD) camera, in common use in the art. A CCD camera includes an array of picture elements (pixels). The light from the specimen is imaged on the CCD. Particular pixels corresponding to regions of the specimen (e.g., individual hybridization sites on an array of biological polymers) are sampled to obtain light intensity readings for each position. Multiple pixels are processed in parallel to increase speed. The apparatus and methods of the invention are easily used for viewing any sample, e.g., by fluorescent or dark field microscopic techniques.

VIII. ADMINISTRATION AND PHARMACEUTICAL COMPOSITIONS [185] Modulators of the polypeptides of the invention (e.g., antagonists or agonists) can be administered directly to the mammalian subject for modulation of activity of a polypeptide of the invention in vivo. Administration is by any of the routes normally used for introducing a modulator compound into ultimate contact with the tissue to be treated and is well known to those of skill in the art. Although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. [186] The pharmaceutical compositions of the invention may comprise a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there are a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington 's Pharmaceutical Sciences, 17th ed. 1985)). [187] The modulators (e.g., agonists or antagonists) of the expression or activity of the a polypeptide of the invention, alone or in combination with other suitable components, can be prepared for injection or for use in a pump device. Pump devices (also known as "insulin pumps") are commonly used to administer insulin to patients and therefore can be easily adapted to include compositions of the present invention. Manufacturers of insulin pumps include Animas, Disetronic and MiniMed. [188] The modulators (e.g., agonists or antagonists) of the expression or activity of a polypeptide of the invention, alone or in combination with other suitable components, can be made into aerosol formulations (i.e., they can be "nebulized") to be administered via inhalation. Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. [189] Formulations suitable for administration include aqueous and non- aqueous solutions, isotonic sterile solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives, hi the practice of this invention, compositions can be administered, for example, orally, nasally, topically, intravenously, intraperitoneally, or intrathecally. The formulations of compounds can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials. Solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. The modulators can also be administered as part of a prepared food or drug. [190] The dose administered to a patient, in the context of the present invention should be sufficient to induce a beneficial response in the subject over time. The optimal dose level for any patient will depend on a variety of factors including the efficacy of the specific modulator employed, the age, body weight, physical activity, and diet of the patient, on a possible combination with other drugs, and on the severity of the case of diabetes. It is recommended that the daily dosage of the modulator be determined for each individual patient by those skilled in the art in a similar way as for known insulin compositions. The size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular compound or vector in a particular subject. [191] In determining the effective amount of the modulator to be administered a physician may evaluate circulating plasma levels of the modulator, modulator toxicity, and the production of anti-modulator antibodies, m general, the dose equivalent of a modulator is from about 1 ng/kg to 10 mg/kg for a typical subject. [192] For administration, modulators of the present invention can be administered at a rate determined by the LD-50 of the modulator, and the side-effects of the modulator at various concentrations, as applied to the mass and overall health of the subject. Administration can be accomplished via single or divided doses. [193] The compounds of the present invention can also be used effectively in combination with one or more additional active agents depending on the desired target therapy (see, e.g., Turner, N. et al. Prog. Drug Res. (1998) 51: 33-94; Haffher, S. Diabetes Care (1998) 21: 160-178; and DeFronzo, R. et al. (eds.), Diabetes Reviews (1997) Vol. 5 No. 4). A number of studies have investigated the benefits of combination therapies with oral agents (see, e.g., Mahler, R., J. Clin. Endocrinol Metab. (1999) 84: 1165-71; United Kingdom Prospective Diabetes Study Group: UKPDS 28, Diabetes Care (1998) 21 : 87-92; Bardin, C. W.,(ed.), Current Therapy In Endocrinology And Metabolism, 6th Edition (Mosby - Year Book, Inc., St. Louis, MO 1997); Chiasson, J. et al., Ann. Intern. Med. (1994) 121: 928-935; Coniff, R. et al., Clin. Ther. (1997) 19: 16-26; Coniff, R. et ύ., Am. J. Med. (1995) 98: 443-451; and Iwamoto, Y. et al., Diabet. Med. (1996) 13 365-370; Kwiterovich, P. Am. J. Cardiol (1998) 82(12A): 3U- 17U). These studies indicate that modulation of diabetes, among other diseases, can be further improved by the addition of a second agent to the therapeutic regimen. Combination therapy includes administration of a single pharmaceutical dosage formulation that contains a modulator of the invention and one or more additional active agents, as well as administration of a modulator and each active agent in its own separate pharmaceutical dosage formulation. For example, a modulator and a thiazolidinedione can be administered to the human subject together in a single oral dosage composition, such as a tablet or capsule, or each agent can be administered in separate oral dosage formulations. Where separate dosage formulations are used, a modulator and one or more additional active agents can be administered at essentially the same time (i.e., concurrently), or at separately staggered times (i.e., sequentially). Combination therapy is understood to include all these regimens. [194] One example of combination therapy can be seen in treating pre- diabetic individuals (e.g., to prevent progression into type 2 diabetes) or diabetic individuals (or treating diabetes and its related symptoms, complications, and disorders), wherein the modulators can be effectively used in combination with, for example, sulfonylureas (such as chlorpropamide, tolbutamide, acetohexamide, tolazamide, glyburide, gliclazide, glynase, glimepiride, and glipizide); biguanides (such as metformin); a PPAR beta delta agonist; a ligand or agonist of PPAR gamma such as thiazolidinediones (such as ciglitazone, pioglitazone {see, e.g., U.S. Patent No. 6,218,409), troglitazone, and rosiglitazone {see, e.g., U.S. Patent No. 5,859,037)); PPAR alpha agonists such as clofibrate, gemfibrozil, fenofibrate, ciprofibrate, and bezafibrate; dehydroepiandrosterone (also referred to as DHEA or its conjugated sulphate ester, DHEA-SO4); antiglucocorticoids; TNFα inhibitors; α- glucosidase inhibitors (such as acarbose, miglitol, and voglibose); amylin and amylin derivatives (such as pramlintide, (see, also, U.S. Patent Nos. 5,902,726; 5,124,314; 5,175,145 and 6,143,718.)); insulin secretogogues (such as repaglinide, gliquidone, and nateglinide (see, also, U.S. Patent Nos. 6,251,856; 6,251,865; 6,221,633; 6,174,856)), and insulin.

IX. GENE THERAPY [195] Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids encoding engineered polypeptides of the invention in mammalian cells or target tissues. Such methods can be used to administer nucleic acids encoding polypeptides of the invention to cells in vitro. IQ some embodiments, the nucleic acids encoding polypeptides of the invention are administered for in vivo or ex vivo gene therapy uses. Non- viral vector delivery systems include DNA plasmids, naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. For a review of gene therapy procedures, see Anderson, Science 256:808-813 (1992); Nabel & Feigner, TIBTECH 11:211-217 (1993); Mitani & Caskey, TIBTECH 11:162-166 (1993); Dillon, TIBTECH 11 :167-175 (1993); Miller, Nature 357:455- 460 (1992); Van Brunt, Biotechnology 6(10):l 149-1154 (1988); Vigne, Restorative Neurology and Neuroscience 8:35-36 (1995); Kremer & Perricaudet, British Medical Bulletin 51(1):31-44 (1995); Haddada et ah, in Current Topics in Microbiology and Immunology Doerfler and Bδhm (eds) (1995); and Yu et al, Gene Therapy 1:13-26 (1994). [196] Methods of non- viral delivery of nucleic acids encoding engineered polypeptides of the invention include lipofection, microinjection, biolistics, virosomes, liposomes, immunoliposom.es, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection is described in e.g., US 5,049,386, US 4,946,787; and US 4,897,355) and lipofection reagents are sold commercially (e.g., Transfectam™ and Lipofectin™). Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those of Feigner, WO 91/17424, WO 91/16024. Delivery can be to cells (ex vivo administration) or target tissues (in vivo administration). [197] The preparation of liρid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science 270:404-410 (1995); Blaese et al, Cancer Gene Ther. 2:291-297 (1995); Behr et ah, Bioconjugate Chem. 5:382-389 (1994); Remy et al, Bioconjugate Chem. 5:647- 654 (1994); Gao et al, Gene Therapy 2:710-722 (1995); Ahmad et al, Cancer Res. 52:4817- 4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787). [198] The use of RNA or DNA viral based systems for the delivery of nucleic acids encoding engineered polypeptides of the invention take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus. Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo). Conventional viral based systems for the delivery of polypeptides of the invention could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. Viral vectors are currently the most efficient and versatile method of gene transfer in target cells and tissues. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues. [199] The tropism of a retrovirus can be altered by incorporating foreign envelope proteins, expanding the potential target population of target cells. Lentiviral vectors are retroviral vectors that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum ex¬ acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian Immuno deficiency virus (SIV), human irnrnuno deficiency virus (HIV), and combinations thereof (see, e.g., Buchscher et al, J. Virol. 66:2731-2739 (1992); Johann et α/., J. Virol. 66:1635-1640 (1992); Sommerfelt et α/., Virol. 176:58-59 (1990); Wilson et al, J. Virol 63:2374-2378 (1989); Miller et al, J. Virol. 65:2220-2224 (1991); PCT/US94/05700). [200] In applications where transient expression of the polypeptides of the invention is preferred, adenoviral based systems are typically used. Adenoviral based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system. Adeno-associated virus ("AAV") vectors are also used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acids and peptides, and for in vivo and ex vivo gene therapy procedures {see, e.g., West et al, Virology 160:38-47 (1987); U.S. Patent No. 4,797,368; WO 93/24641; Kotin, Human Gene Therapy 5:793-801 (1994); Muzyczka, J. Clin. Invest. 94:1351 (1994)). Construction of recombinant AAV vectors are described in a number of publications, including U.S. Pat. No. 5,173,414; Tratschin et al., Mol Cell. Biol. 5:3251-3260 (1985); Tratschin, et al, MoI Cell. Biol. 4:2072-2081 (1984); Hermonat & Muzyczka, PNAS 81:6466-6470 (1984); and Samulski et al, J. Virol. 63:03822-3828 (1989). [201] pLASN and MFG-S are examples are retroviral vectors that have been used in clinical trials (Dunbar etal, Blood 85:3048-305 (1995); Kohn et al, Nat. Med. 1:1017-102 (1995); Malech et al, PNAS 94:22 12133-12138 (1997)). PA317/pLASN was the first therapeutic vector used in a gene therapy trial. (Blaese et ah, Science 270:475-480 (1995)). Transduction efficiencies of 50% or greater have been observed for MFG-S packaged vectors. (Ellem et ah, Immunol Immunother. 44(l):10-20 (1997); Dranoff et ah, Hum. Gene Ther. 1:111-2 (1997). [202] Recombinant adeno-associated virus vectors (rAAV) are a promising alternative gene delivery systems based on the defective and nonpathogenic parvovirus adeno-associated type 2 virus. All vectors are derived from a plasmid that retains only the AAV 145 bp inverted terminal repeats flanking the transgene expression cassette. Efficient gene transfer and stable transgene delivery due to integration into the genomes of the transduced cell are key features for this vector system. (Wagner et ah, Lancet 351:9117 1702- 3 (1998), Kearns et ah, Gene Ther. 9:748-55 (1996)). [203] Replication-deficient recombinant adenoviral vectors (Ad) can be engineered such that a transgene replaces the Ad EIa, EIb, and E3 genes; subsequently the replication defector vector is propagated in human 293 cells that supply deleted gene function in trans. Ad vectors can transduce multiply types of tissues in vivo, including nondividing, differentiated cells such as those found in the liver, kidney and muscle system tissues. Conventional Ad vectors have a large carrying capacity. An example of the use of an Ad vector in a clinical trial involved polynucleotide therapy for antitumor immunization with intramuscular injection (Sterman et ah, Hum. Gene Ther. 7:1083-9 (1998)). Additional examples of the use of adenovirus vectors for gene transfer in clinical trials include Rosenecker et ah, Infection 24:1 5-10 (1996); Sterman et ah, Hum. Gene Ther. 9:7 1083- 1089 (1998); Welsh et ah, Hum. Gene Ther. 2:205-18 (1995); Alvarez et ah, Hum. Gene Ther. 5:597-613 (1997); Topf et al, Gene Ther. 5:507-513 (1998); Sterman et al, Hum. Gene Ther. 7:1083-1089 (1998). [204] Packaging cells are used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and ψ2 cells or PA317 cells, which package retrovirus. Viral vectors used in gene therapy are usually generated by producer cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences being replaced by an expression cassette for the protein to be expressed. The missing viral functions are supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome which are required for packaging and integration into the host genome. Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences. The cell line is also infected with adenovirus as a helper. The helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid. The helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV. [205] In many gene therapy applications, it is desirable that the gene therapy vector be delivered with a high degree of specificity to a particular tissue type. A viral vector is typically modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the viruses outer surface. The ligand is chosen to have affinity for a receptor known to be present on the cell type of interest. For example, Han et al, PNAS 92:9747-9751 (1995), reported that Moloney murine leukemia virus can be modified to express human heregulin fused to gp70, and the recombinant virus infects certain human breast cancer cells expressing human epidermal growth factor receptor. This principle can be extended to other pairs of virus expressing a ligand fusion protein and target cell expressing a receptor. For example, filamentous phage can be engineered to display antibody fragments (e.g., FAB or Fv) having specific binding affinity for virtually any chosen cellular receptor. Although the above description applies primarily to viral vectors, the same principles can be applied to nonviral vectors. Such vectors can be engineered to contain specific uptake sequences thought to favor uptake by specific target cells. [206] Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravenous, intraperitoneal, intramuscular, subdermal, or intracranial infusion) or topical application, as described below. Alternatively, vectors can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells which have incorporated the vector. [207] Ex vivo cell transfection for diagnostics, research, or for gene therapy (e.g., via re-infusion of the transfected cells into the host organism) is well known to those of skill in the art. hi some embodiments, cells are isolated from the subject organism, transfected with a nucleic acid (gene or cDNA) encoding a polypeptides of the invention, and re-infused back into the subject organism (e.g., patient). Various cell types suitable for ex vivo transfection are well known to those of skill in the art (see, e.g., Freshney et al., Culture of Animal Cells, A Manual of Basic Technique (3rd ed. 1994)) and the references cited therein for a discussion of how to isolate and culture cells from patients). [208] In one embodiment, stem cells are used in ex vivo procedures for cell transfection and gene therapy. The advantage to using stem cells is that they can be differentiated into other cell types in vitro, or can be introduced into a mammal (such as the donor of the cells) where they will engraft in the bone marrow. Methods for differentiating CD34+ cells in vitro into clinically important immune cell types using cytokines such a GM- CSF, IFN-γ and TNF-α are known {see Inaba et al, J. Exp. Med. 176:1693-1702 (1992)). [209] Stem cells are isolated for transduction and differentiation using known methods. For example, stem cells are isolated from bone marrow cells by panning the bone marrow cells with antibodies which bind unwanted cells, such as CD4+ and CD 8+ (T cells), CD45+ (panB cells), GR-I (granulocytes), and lad (differentiated antigen presenting cells) {see Inaba et al, J. Exp. Med. 176:1693-1702 (1992)). [210] Vectors (e.g., retroviruses, adenoviruses, liposomes, etc.) containing therapeutic nucleic acids can be also administered directly to the organism for transduction of cells in vivo. Alternatively, naked DNA can be administered. Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. [211] Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention, as described below {see, e.g., Remington 's Pharmaceutical Sciences, 17th ed., 1989).

X. DIAGNOSIS OF DIABETES [212] The present invention also provides methods of diagnosing diabetes or a predisposition of at least some of the pathologies of diabetes. Diagnosis can involve determination of a genotype of an individual (e.g., with SNPs) and comparison of the genotype with alleles known to have an association with the occurrence of diabetes. Alternatively, diagnosis also involves determining the level of a polypeptide or polynucleotide of the invention in a patient and then comparing the level to a baseline or range. Typically, the baseline value is representative of a polypeptide or polynucleotide of the invention in a healthy (e.g., lean) person. [213] As discussed above, variation of levels (e.g., low or high levels) of a polypeptide or polynucleotide of the invention compared to the baseline range indicates that the patient is either diabetic or at risk of developing at least some of the pathologies of diabetes (e.g., pre-diabetic). The level of a polypeptide in a lean individual can be a reading from a single individual, but is typically a statistically relevant average from a group of lean individuals. The level of a polypeptide in a lean individual can be represented by a value, for example in a computer program. [214] In some embodiments, the level of polypeptide or polynucleotide of the invention is measured by taking a blood, urine or tissue sample from a patient and measuring the amount of a polypeptide or polynucleotide of the invention in the sample using any number of detection methods, such as those discussed herein. For instance, fasting and fed blood or urine levels can be tested. [215] In some embodiments, the baseline level and the level in a lean sample from an individual, or at least two samples from the same individual differ by at least about 5%, 10%, 20%, 50%, 75%, 100%, 150%, 200%, 300%, 400%, 500%, 1000% or more. In some embodiments, the sample from the individual is greater by at least one of the above- listed percentages relative to the baseline level. In some embodiments, the sample from the individual is lower by at least one of the above-listed percentages relative to the baseline level. [216] In some embodiments, the level of a polypeptide or polynucleotide of the invention is used to monitor the effectiveness of antidiabetic therapies such as thiazolidinediones, metformin, sulfonylureas and other standard therapies. In some embodiments the activity or expression of a polypeptide or polynucleotide of the invention will be measured prior to and after treatment of diabetic or pre-diabetic patients with antidiabetic therapies as a surrogate marker of clinical effectiveness. For example, the greater the reduction in expression or activity of a polypeptide of the invention indicates greater effectiveness. [217] Glucose/insulin tolerance tests can also be used to detect the effect of glucose levels on levels of a polypeptide or polynucleotide of the invention. In glucose tolerance tests, the patient's ability to tolerate a standard oral glucose load is evaluated by assessing serum and urine specimens for glucose levels. Blood samples are taken before the glucose is ingested, glucose is given by mouth, and blood or urine glucose levels are tested at set intervals after glucose ingestion. Similarly, meal tolerance tests can also be used to detect the effect of insulin or food, respectively, on levels of a polypeptide or polynucleotide of the invention.

[218] All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. [219] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

EXAMPLES [220] The following examples are offered to illustrate, but not to limit the claimed invention. [221] The molecular mechanism by which thiazolidinediones (TZDs) cause an increase in peripheral insulin sensitivity was studied. Genes in muscle or fat whose expression is altered by TZDs may lie on a pathway leading from TZD treatment to increased insulin sensitivity. Modulators of such genes can elicit the same effect as TZD treatment. Moreover, such modulators can lack some of the side effects of TZD. The majority of glucose disposal occurs in muscle. For this reason, gene expression profiling in human muscle from diabetics treated with troglitazone was used to identify genes important for TZD action and therefore treatment of diabetes and insulin resistance. In (obese individuals) type II diabetics, peripheral tissues, especially muscle and fat, are known to have a (moderately) severely impaired ability to respond to insulin and hence to take up glucose. This defect in glucose metabolism is usually compensated for by increased secretion of insulin from the pancreas, thereby maintaining normal glucose levels. The majority of glucose disposal occurs in the muscle. A number of obese insulin resistant patients will progress to overt diabetics with time. The molecular defects underlying this peripheral insulin resistance in (obese individuals) diabetics are not well defined. Genes in muscle or fat whose expression is altered in (obese individuals) diabetics when compared to lean individuals may be causative genes for insulin resistance and also may predict the transition to diabetes. Modulators of such genes have the ability to reverse insulin resistance and restore normal insulin sensitivity, thereby improving whole body glucose homeostasis including for example insulin secretion. Modulators of such genes also have the ability to be used to pre-empt the transition from obesity-induced insulin resisatnce to diabetes. The majority of glucose disposal occurs in the muscle. For these reasons, gene expression profiling was performed in muscle from lean, obese and diabetic individuals. [222] Gene expression profiling was performed on tissue samples (muscle) obtained from lean, obese and diabetic individuals. Two studies were performed. In the first study, basal samples were isolated from all individuals at the beginning of a 5 hour hyperinsulinemic euglycemic clamp. Clamp samples were isolated at the end of this procedure. Similar basal and clamp samples were taken 3 months later after all patients had taken the insulin sensitizing drug troglitazone (tro). [223] In the second study, samples were obtained from lean, obese and diabetic individuals before and after a hyperinsulinemic euglycemic clamp. No troglitazone treatment was used. For all tissue samples mRNA was isolated from these muscle samples and converted to cRNA by standard procedures. The gene expression profile for each individual was determined by hybridization of cRNA to custom synthesized Affymetrix chips. [224] Gene expression profile differences were calculated as follows. The expression level of a particular gene is indicated by its 'average difference score'. The raw data was analyzed by a statistical test to remove Outliers'. The mean 'average difference score' was then calculated from the average difference scores for all individuals in a particular treatment group. Genes were determined to be changed in condition 1 (such as basal diabetics before trogliazone (tro) treatment) versus condition 2 (such as basal diabetics after troglitazone treatment), condition 1 (such as basal leans) versus condition 2 (such as basal diabetics) or condition 1 (such as basal leans) versus condition 2 (such as basal obese) by calculating the Students t test statistic between the two conditions and selecting those with t less than or equal to 0.05. Fold change was determined as the ratio of mean average difference score in condition 2 to the mean average difference score in condition 1. BC001573 [225] Probe set MBXHUMMUS36548 detects BC001573 nucleic acid sequences. Expression of BCOO 1573 transcripts was increased in diabetic compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of diabetic in comparison to lean patients. [226] Probe set MBXHUMMUS36548 detects BCOOl 573 nucleic acid sequences. Expression of BCOO 1573 transcripts was increased in obese compared to lean patients in the gene profiling experiment.

B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of obese in comparison to lean patients. [227] BCOOl 573 was also evaluated using real-time PCR. The results further show that BCOOl 573 is significantly over-expressed in muscle from obese individuals when compared to muscle from lean individuals.

"Fold Change" indicates the fold expression calculated as the ratio of the mean obese expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [228] BCOOl 573 contains the following protein domains (designated with reference to SEQ ID NO:2): Dienelactone hydrolase family (PF01738) at amino acids 30 to 245. It is possible that this enzyme may play a role in catalyzing the hydrolysis of long chain fatty acyl-CoA thioesters. CILP

[229] Probe set MBXHUMMUS08162 detects CILP nucleic acid sequences. Expression of CILP transcripts was increased in diabetic compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of diabetic in comparison to lean patients. [230] CILP was also evaluated using real-time PCR. The results further show that CILP is significantly over-expressed in muscle from diabetic individuals when compared to muscle from lean individuals.

"Fold Change" indicates the fold expression calculated as the ratio of the mean diabetic expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [231] Probe set MBXHUMMUS08162 detects CILP nucleic acid sequences. Expression of CILP transcripts was increased in obese compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of obese in comparison to lean patients. [232] CILP was also evaluated using real-time PCR. The results further show that CILP is significantly over-expressed in muscle from obese individuals when compared to muscle from lean individuals. "Fold Change" indicates the fold expression calculated as the ratio of the mean obese expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [233] CILP contains the following protein domains (designated with reference to SEQ ID NO:8): Thrombospondin type 1 domain (PF00090) at amino acids 153 to 200; and Immunoglobulin domain (PF00047) at amino acids 323 to 378. Soluble secreted forms of CILP has been detected (Lorenzo, P. et al, J Biol Chem. 1998 Sep 4;273(36):23469-75) and these are displayed in SEQ ID NO:9 and 10. CILP could impair chondrocyte growth and matrix repair (Johnson, K. et al., Arthritis Rheum. 48:1302-14. (2003)). An immune response to CILP has been observed and this is proposed to play a role in the pathogenesis of inflammatory joint destruction (Tsuruha, J. et al., Arthritis Rheum. 44:838-45 (2001)).

FLJ45434 [234] Probe set MBXHUMMUS 05222 detects FLJ45434 nucleic acid sequences. Expression of FLJ45434 transcripts was increased in diabetic compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of diabetic in comparison to lean patients. [235] FLJ45434 was also evaluated using real-time PCR. The results further show that FLJ45434 is significantly over-expressed in muscle from diabetic individuals when compared to muscle from lean individuals.

"Fold Change" indicates the fold expression calculated as the ratio of the mean diabetic expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [236] Probe set MBXHUMMUS05222 detects FLJ45434 nucleic acid sequences. Expression of FLJ45434 transcripts was increased in obese compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of obese in comparison to lean patients. [237] FLJ45434 was also evaluated using real-time PCR. The results further show that FLJ45434 is significantly over-expressed in muscle from obese individuals when compared to muscle from lean individuals.

"Fold Change" indicates the fold expression calculated as the ratio of the mean obese expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [238] FLJ45434 contains the following protein domains (designated with reference to SEQ ID NO: 16): Lipopolysaccharide kinase (Kdo/WaaP) family (PF06293) at amino acids 91 to 294; RIOl family (PFOl 163) at amino acids 119 to 266; and Protein kinase domain (PF00069) at amino acids 106 to 361.

UBE2E3 [239] Probe set MBXHUMMUS 11919 detects UBE2E3 nucleic acid sequences. Expression of UBE2E3 transcripts was increased in diabetic compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of diabetic in comparison to lean patients. [240] UBE2E3 was also evaluated using real-time PCR. The results further show that UBE2E3 is significantly over-expressed in muscle from diabetic individuals when compared to muscle from lean individuals.

"Fold Change" indicates the fold expression calculated as the ratio of the mean diabetic expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [241] Probe set MBXHUMMUS 11919 detects UBE2E3 nucleic acid sequences. Expression of UBE2E3 transcripts was increased in obese compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of obese in comparison to lean patients. [242] UBE2E3 contains the following protein domains (designated with reference to SEQ ID NO: 18): Ubiquitin-conjugating enzyme (PFOO 179) at amino acids 65 to 202. Overexpression of UBE2E3 has been reported to affect Na+ transport activity of the heteromeric protein complex of the epithelial Na+ channel (ENaC). This channel is known to play a fundamental role in Na+ homeostasis and blood pressure regulation (Debonneville, C. and Staub, O., MoI Cell Biol. 24:2397-409 (2004)). USP38 [243] Probe set MBXHUMMUS03589 detects USP38 nucleic acid sequences. Expression of USP38 transcripts was increased in obese compared to lean patients in the gene profiling experiment. B/C indicates sample is from Basal or Clamp; "Mean Expr" indicates mean expression; "SEM" indicates standard error of mean; "n" indicates number of patient samples; "Fold Change" indicates fold change of obese in comparison to lean patients. [244] USP38 was also evaluated using real-time PCR. The results further show that USP38 is significantly over-expressed in muscle from obese individuals when compared to muscle from lean individuals. "Fold Change" indicates the fold expression calculated as the ratio of the mean obese expression/ mean lean expression. Numbers in parentheses indicates the number of patient samples analyzed by real-time PCR. [245] USP38 contains the following protein domains (designated with reference to SEQ ID NO: 24): Ubiquitin carboxyl-terminal hydrolase (PF00443) at amino acids 442 to 946. USP38 belong to the large protein family of cysteine proteases that specifically cleave ubiquitin from ubiquitin-conjugated protein substrates. SEQIDNO: 1

gi|31982874|ref]NM_138809.2|Homosapienshypotheticalprotei nBC001573(LOC134147), mRNA 1 gactttgcag atttctcttc ccccaggcct ccctcctcca cctctccgcc ccctccgggc 61 ttggctctcc caggaggcta cgactggagc cactggtccc gcaggatccc cgcgtcctcg 121 gtcgccgcgt ccacgtccct ctcgcgtccc cgcccggcgc cacgccgcct ccctctgggt 181 tcggcctccg cgcggtgcag cgcagttctc aggccgcggg acaagcccga cttaaatctc 241 tgcaatggct aacgaagctt atccttgtcc gtgtgacatt ggccacagac ttgagtatgg 301 agggctaggc cgtgaagttc aagtcgagca catcaaggct tatgtcacca aatcccccgt 361 tgatgcaggc aaagctgtga ttgtcattca agatatattt ggctggcagt tgcccaatac 421 cagatatata gctgacatga tctcaggaaa tggatacaca accattgttc cagacttctt 481 tgtagggcaa gagccttggg acccctctgg cgactggtct atcttccctg agtggctgaa 541 aacaagaaat gcccagaaga tcgatagaga gatcagtgct atcttgaagt atctgaaaca 601 acagtgtcat gcccagaaaa ttggcatcgt gggattctgc tggggtggaa ctgctgtcca 661 tcatttgatg atgaaatact cagaattcag ggcaggggtg tccgtctatg gcattgtcaa 721 ggattctgaa gacatttaca atttaaagaa ccccactttg ttcatttttg ctgaaaatga 781 tgttgtgatt ccactcaagg acgtatcttt gctgactcag aagttgaaag aacactgcaa 841 agttgaatat caaattaaaa cattttctgg gcagactcat gggttcgtgc atcggaagag 901 agaagattgc tcacctgcag acaagcccta cattgacgag gccagaagga atttaattga 961 gtggctgaac aagtacatgt agcaagaatc aagggcaagc cttcctagaa tagctttcat 1021 cccaaaattt gcttggaaat agttagatca tttgatttaa ttttcacttt tataaaataa 1081 gtgtaggaat cctaaaattg attatttcat ttgaaacaca aattcagtag gacgtaatgc 1141 atgaaataat ttaatttttg acatgtacat cgaatcataa tttaaaaaca aggtctgacc 1201 aggtgtagtg cctcatgcct gtaattccag cactttggga ggccaaagtg ggtggatcac 1261 ctgaggtcag gagtttgaga ccagcctggc caacatggtg agaccccatc tctacaaaaa 1321 atacaaaaat tagcctggtg tggtggtgca cacctgtagt cccagctact tgggaggctg 1381 aggcacagga atcaatagaa ccccaggagg tggagactgc agtgagccaa gattgtgcca 1441 ctactgtact ctagcctggg cagcagagtg agaccctgtc tcaaaaataa ataagtaaat 1501 aaataaataa ataaaataaa aaccaggtca gtacctggag aatttgaatg atagagaatg 1561 atagagtaat accctaatta ttagttaaaa cctacaggcc gggtgyggtg gctcacgcct 1621 gtaatcccag cactttggga ggccgaggtg ggtggatcac ttgaggtcag gagttcaaga 1681 ccagcctggc caacatggtg aaaccccatc tctactaaaa atataaaaat cagccgggca 1741 tggtggcatg tgcctgtaat cccagctact caggagtctg aggaggagaa tcacttgaac 1801 ctggaggcag aggttgcagt gagtcgaggt tgcgctactg cactccagcc tggacaacag 1861 agggagactc tgtctcaaaa aaaaaaaacc tacagctgtt caaggaccag ctgacaggtc 1921 aagtgtggcc ttttctggtc tttgaacaca tcatagaaag tgacaaatgc tgcaaagcca 1981 tgaagaacat gaactataaa cgggtagact aactgcccag cttagacact tatctatgcc 2041 acaaaacagc tgaatttgtc acatttatat attgcaatat gggaagtatt gagatcaaaa 2101 caggattcca ttgacctaat tatattaagc taataaacta attttttgaa tttttg

SEQIDNO:2

AminoacidsequenceofhumanBCOOl573encodedbytheDNAsequencesh owninSEQID NO:1. MANEAYPCPCDIGHRLEYGGLGREVQVEHIKΆYVTKSPVDAGKAVIVIQDIFGWQLPNT R YIADMISGNGYTTIVPDFFVGQEPWDPSGDWSIFPEWLKTRNAQKIDREISAILKYLKQQ CHAQKIGIVGFCWGGTAVHHLMMKYSEFRAGVSVYGIVKDSEDIYNLKNPTLFIFAENDV VIPLKDVSLLTQKLKEHCKVEYQIKTFSGQTHGFVHRKREDCSPADKPYIDEARRNLIEW LNKYM SEQIDNO:3

gi|31795583|ref|NM_l81588.2)MusmusculusRIKENcDNA2310016A09ge ne (2310016A09Rik),mRNA 1 cccacgcgtc cgcccacgcg tccggctgac ttgctcttca ccattagaca agccctgacg 61 tcaagacccc aatggctaac gaagctaacc cttgcccatg tgacattggt cacaaactag 121 agtatggtgg catgggccac gaagttcagg ttgagcacat caaggcatat gtcacccggt J81 cccccgtgga tgcaggcaaa gccgtgattg tcgtccagga tatatttggc tggcagctgc 241 ccaacaccag gtatatggct gacatgattg ccagaaatgg atacacaact attgtcccag 301 acttctttgt gggtcaagag ccatgggacc cagctggtga ctggtccacc ttccccgcgt 361 ggttgaaatc aagaaatgcc agaaaagtca atcgggaggt tgatgctgtc ttgaggtatc 421 tgagacaaca gtgtcatgct cagaagattg gcattgtggg cttctgctgg gggggtgttg 481 tggtgcacca agtgatgacg gcatacccgg acatcagggc gggggtgtct gtctatggta 541 tcatcagaga ttctgaagat gtttataatt tgaagaaccc aacgttgttt atttttgcag 601 aaaatgacac tgtgattcca cttgagcagg tttctacact gacccagaag cttaaagaac 661 actgcatagt taattaccaa gttaaaacgt tttctgggca aactcatggc tttgtgcatc 721 ggaagagaga agactgctcc cctgctgaca agccctacat cgaggaggca aggaggaacc 781 taattgaatg gctgaacaag tatgtctaac agcacccaag cacaaggcag ctgttgccca 841 aatttgcctg aataattaac catctcttac tatccatgat agcatgactg ttaggggaac 901 tgggggcaat tcagggacca gctagattct cagatatggc cttacattgg taattcaagt 961 gttaaaatgt acaaaaccag gaagagcagt tgtcactatg tatattcaac ttcccctaat 1021 gagcagtgtc gacattcaac ctaataaagt gatttttaaa acttgaaaaa aaaaaaaaaa 1081 aaaaaaaaaa aaa SEQIDNO:4

Amino acid sequence of mouse BCOOl 573 encoded by the DNA sequence shown in SEQ ID NO: 3. MANEANPCPCDIGHKLEYGGMGHEVQVEHIKAYVTRSPVDAGKAVIVVQDIFGWQLPNTR YMADMIARNGYTTIVPDFFVGQEPWDPAGDWSTFPAWLKSRNARKVNREVDAVLRYLRQQ CHAQKIGIVGFCWGGVVVHQVMTAYPDIRAGVSVYGIIRDSEDVYNLKNPTLFIFAENDT VIPLEQVSTLTQKLKEHCIVNYQVKTFSGQTHGFVHRKREDCSPADKPYIEEARRNLIEW LNKYV SEQ ID NO: 5

gi|34854914]reflXM_226908.2| Rattus norvegicus similar to Ab2-225 (LOC310201), mRNA i gaggtctttg ctttatcctg aagctcgcct tctagaaaat gtatcatggg gaaaagcaaa 61 aatcctggag gcagacccag aaaacacggg ctgccaaggg tcaagcatgt attcttgatt 121 cttagcctga acccccacag gcctgtccgt gggtagaaac tgcttgccct gtccagagta 181 cacttggctc ctgcttctta gactgggctc agcggttgag accatgtgaa agagctgggc 241 aggactccag ccttccacat gtggcgttct tgggcctatg ttggctctac cccattctgt 301 gggagaaaaa ccttggctct cctggatgta aaaattcctt ttgtaattgg agctcgagtc 361 ggaaaagcct gttgccagca ccttcgaagg gactagattc ctttgggtct tcccagcaga 421 gtccagcctg acgtcaagac cccaatggct aacgaagcta acccttgccc atgtgacatt 481 ggtcacaggc tagactatgg tggcatgggc caggaagttc aggttgagca catcaaggca 541 tatgtcaccc ggtcccctgt ggatgcaggc aaagctgtga ttgttgtcca ggatatattt 601 ggctggcagc tgtccaacac caggtatatg gctgacatga ttgctggaaa tggatacaca 661 actattgtcc cagacttctt tgtgggtcaa gagccatggg acccggctgg tgattggtcc 721 accttccctg agtggttgaa atcaagaaat gccagaaaaa tcaaccgaga ggttgatgct 781 gtcttgaggt atctgaaaca acagtgtcat gcccagaaga ttggcattgt gggcttctgc 841 tgggggggta ttgtggtgca ccacgtgatg acgacatatc cagaagtcag agcgggggtg 901 tctgtctatg gtatcatcag agattctgaa gatgtttata atttgaagaa cccaacgttg 961 tttatctttg cagaaaatga tgctgtgatt ccacttgagc aggtttctat actgatccag 1021 aagcttaaag aacactgcat agttaattac caagttaaga cattttctgg gcaaactcat 1081 ggctttgtgc atcggaagag agaagactgc tcccctgcag acaaacccta cattgaggaa 1141 gcgaggagga atctcatcga atggctgaac aagtatattt aacagcactc aagcacaaat 1201 tttgaataat taaattgacc cgaataatta accatctcct aaaattctcc tgaaacatac 1261 agtaggaact aaatgatcaa tatctgtaag acttaaatga tagcataact aattattagg 1321 ggaacctggg gccattcagg gaccagctag attctcagat atggccttac cttggtaatt 1381 tgttaacatg tacaaaccca ggaagatctt gagttgtctc tatgtgtatt caatctcccc 1441 tagtgaagtt caattctgca cttcagtgta agcagtgttg acattcagtt ctcaccactc 1501 taactattca acctaataaa gtgattttta aaacttg SEQIDNO:6 Amino acid sequence of rat BCOOl 573 encoded by the DNA sequence shown in SEQ ID NO: 5. MANEANPCPCDIGHRLDYGGMGQEVQVEHIKAYVTRSPVDAGKAVIWQDIFGWQLSNTR YMADMIAGNGYTTIVPDFFVGQEPWDPAGDWSTFPEWLKSRNARKINREVDAVLRYLKQQ CHAQKIGIVGFCWGGIVVHHVMTTYPEVRAGVSVYGIIRDSEDVYNLKNPTLFIFAENDA VIPLEQVSILIQKLKEHCIVNYQVKTFSGQTHGFVHRKREDCSPADKPYIEEARRNLIEW LNKYI SEQIDNO:7

gi|4502844|ref]NM_003613.1 j Homo sapiens cartilage intermediate layer protein, nucleotide pyrophosphohydrolase (CILP), mRNA i tgaggagtcc tgctcaagac acggtcactg gatctgagaa acttcccagg ggaccgcatt 61 ccagagtcag tgactctgtg aagcacccac atctacctct tgccacgttc ccacgggctt 121 gggggaaaga tggtggggac caaggcctgg gtgttctcct tcctggtcct ggaagtcaca 181 tctgtgttgg ggagacagac gatgctcacc cagtcagtaa gaagagtcca gcctgggaag 241 aagaacccca gcatctttgc caagcctgcc gacaccctgg agagccctgg tgagtggaca 301 acattgttca acatcgacta cccaggcggg aagggcgact atgagcggct ggacgccatt 361 cgcttctact atggggaccg tgtatgtgcc cgtcccctgc ggctagaggc tcggaccact 421 gactggacac ctgcgggcag cactggccag gtggtccatg gtagtccccg tgagggtttc 481 tggtgcctca acagggagca gcggcctggc cagaactgct ctaattacac cgtacgcttc 541 ctctgcccac caggatccct gcgccgagac acagagcgca tctggagccc atggtctccc 601 tggagcaagt gctcagctgc ctgtggtcag actggggtcc agactcgcac acgcatttgc 661 ttggcagaga tggtgtcgct gtgcagtgag gccagcgaag agggtcagca ctgcatgggc 721 caggactgta cagcctgtga cctgacctgc ccaatgggcc aggtgaatgc tgactgtgat 781 gcctgcatgt gccaggactt catgcttcat ggggctgtct cccttcccgg aggtgcccca 841 gcctcagggg ctgctatcta cctcctgacc aagacgccga agctgctgac ccagacagac 901 agtgatggga gattccgaat ccctggcttg tgccctgatg gcaaaagcat cctgaagatc 961 acaaaggtca agtttgcccc cattgtactc acaatgccca agactagcct gaaggcagcc 1021 accatcaagg cagagtttgt gagggcagag actccataca tggtgatgaa ccctgagaca 1081 aaagcacgga gagctgggca gagcgtgtct ctgtgctgta aggccacagg gaagcccagg 1141 ccagacaagt atttttggta tcataatgac acattgctgg atccttccct ctacaagcat 1201 gagagcaagc tggtgctgag gaaactgcag cagcaccagg ctggggagta cttttgcaag 1261 gcccagagtg atgctggggc tgtgaagtcc aaggttgccc agctgattgt cacagcatct 1321 gatgagactc cttgcaaccc agttcctgag agctatctta tccggctgcc ccatgattgc 1381 tttcagaatg ccaccaactc cttctactat gacgtgggac gctgccctgt taagacttgt 1441 gcagggcagc aggataatgg gatcaggtgc cgtgatgctg tgcagaactg ctgtggcatc 1501 tccaagacag aggaaaggga gatccagtgc agtggctaca cgctacccac caaggtggcc 1561 aaggagtgca gctgccagcg gtgtacggaa actcggagca tcgtgcgggg ccgtgtcagt 1621 gctgctgaca atggggagcc catgcgcttt ggccatgtgt acatggggaa cagccgtgta 1681 agcatgactg gctacaaggg cactttcacc ctccatgtcc cccaggacac tgagaggctg 1741 gtgctcacat ttgtggacag gctgcagaag tttgtcaaca ccaccaaagt gctacctttc 1801 aacaagaagg ggagtgccgt gttccatgaa atcaagatgc ttcgtcggaa agagcccatc 1861 actttggaag ccatggagac caacatcatc cccctggggg aagtggttgg tgaagacccc 1921 atggctgaac tggagattcc atccaggagt ttctacaggc agaatgggga gccctacata 1981 ggaaaagtga aggccagtgt gaccttcctg gatccccgga atatttccac agccacagct 2041 gcccagactg acctgaactt catcaatgac gaaggagaca ctttccccct tcggacgtat 2101 ggcatgttct ctgtggactt cagagatgag gtcacctcag agccacttaa tgctggcaaa 2161 gtgaaggtcc accttgactc gacccaggtc aagatgccag agcacatatc cacagtgaaa 2221 ctctggtcac tcaatccaga cacagggctg tgggaggagg aaggtgattt caaatttgaa 2281 aatcaaagga ggaacaaaag agaagacaga accttcctgg tgggcaacct ggagattcgt 2341 gagaggaggc tctttaacct ggatgttcct gaaagcaggc ggtgctttgt taaggtgagg 2401 gcctaccgga gtgagaggtt cttgcctagt gagcagatcc agggggttgt gatctccgtg 2461 attaacctgg agcctagaac tggcttcttg tccaacccta gggcctgggg ccgctttgac 2521 agtgtcatca caggccccaa cggggcctgt gtgcctgcct tctgtgatga ccagtcccct 2581 gatgcctact ctgcctatgt cttggcaagc ctggctgggg aggaactgca agcagtggag 2641 tcttctccta aattcaaccc aaatgcaatt ggcgtccctc agccctatct caacaagctc 2701 aactaccgtc ggacggacca tgaggatcca cgggttaaaa agacagcttt ccagattagc 2761 atggccaagc caaggcccaa ctcagctgag gagagcaatg ggcccatcta tgcctttgag 2821 aacctccggg catgtgaaga ggcaccaccc agtgcagccc acttccggtt ctaccagatt 2881 gagggggatc gatatgacta caacacagtc cccttcaacg aagatgaccc tatgagctgg 2941 actgaagact atctggcatg gtggccaaag ccgatggaat tcagggcctg ctatatcaag 3001 gtgaagattg tggggccact ggaagtgaat gtgcgatccc gcaacatggg gggcactcat 3061 cggcggacag tggggaagct gtatggaatc cgagatgtga ggagcactcg ggacagggac 3121 cagcccaatg tctcagctgc ctgtctggag ttcaagtgca gtgggatgct ctatgatcag 3181 gaccgtgtgg accgcaccct ggtgaaggtc atcccccagg gcagctgccg tcgagccagt 3241 gtgaacccca tgctgcatga gtacctggtc aaccacttgc cacttgcagt caacaacgac 3301 accagtgagt acaccatgct ggcacccttg gacccactgg gccacaacta tggcatctac 3361 actgtcactg accaggaccc tcgcacggcc aaggagatcg cgctcggccg gtgctttgat 3421 ggcacatccg atggctcctc cagaatcatg aagagcaatg tgggagtagc cctcaccttc 3481 aactgtgtag agaggcaagt aggccgccag agtgccttcc agtacctcca aagcacccca 3541 gcccagtccc ctgctgcagg cactgtccaa ggaagagtgc cctcgaggag gcagcagcga 3601 gcgagcaggg gtggccagcg ccagagtgga gtggtggcct ctctgagatt tcctagagtt 3661 gctcaacagc ccctgatcaa ctaagttttg tggtacttca ccctcttctg ccctcatttc 3721 atgtgacagc cattgtgaga ctgatgcaca aactgtcact tggttaattt aagcacttct 3781 gttttcgtga atttgcttgt ttgtttcttc atgcctttac ttactttgtc ccatgctact 3841 gattggcacg tggcccccac aatggcacaa taaagcccct ttgtgaaact gttctttaaa 3901 tgaaacacaa gaaattggcc actggtaaaa ctctgcagct tcaactgtac ttcatttaat 3961 gccattaatg caaatatact tcctcttctt tttgcatggt tttgcccacc tctgcaatag 4021 tgataatctg atgctgaaga tcaaataacc aatataaagc atatttcttg gccttgctcc 4081 acaggacata ggcaagcctt gatcatagtt catacatata aatggtggtg aaataaagaa 4141 ataaaacaca atacttttac tggaaaaaaa aaaaa SEQIDNO: 8

AminoacidsequenceofhumanCILPencodedbytheDNAsequenceshowni nSEQIDNO: 7. MVGTKAWVFSFLVLEVTSVLGRQTMLTQSVRRVQPGKKNPSIFAKPADTLESPGEWTTLF NIDYPGGKGDYERLDAIRFYYGDRVCARPLRLEARTTDWTPAGSTGQVVHGSPREGFWCL NREQRPGQNCSNYTVRFLCPPGSLRRDTERIWSPWSPWSKCSAACGQTGVQTRTRICLAE MVSLCSEASEEGQHCMGQDCTACDLTCPMGQVNADCDACMCQDFMLHGAVSLPGGΆPΆ SG AAIYLLTKTPKLLTQTDSDGRFRIPGLCPDGKSILKITKVKFAPIVLTMPKTSLKAATIK AEFVRAETPYMVMNPETKARRAGQSVSLCCKATGKPRPDKYFWYHNDTLLDPSLYKHESK LVLRKLQQHQAGEYFCKAQSDAGAVKSKVAQLIVTASDETPCNPVPESYLIRLPHDCFQN ATNSFYYDVGRCPVKTCAGQQDNGIRCRDAVQNCCGISKTEEREIQCSGYTLPTKVAKEC SCQRCTETRSIVRGRVSAADNGEPMRFGHVYMGNSRVSMTGYKGTFTLHVPQDTERLVLT FVDRLQKFVNTTKVLPFNKKGSAVFHEIKMLRRKEPITLEAMETNIIPLGEVVGEDPMAE LEIPSRSFYRQNGEPYIGKVKASVTFLDPRNISTATAAQTDLNFINDEGDTFPLRTYGMF SVDFRDEVTSEPLNAGKVKVHLDSTQVKMPEHISTVKLWSLNPDTGLWEEEGDFKFENQR RNKREDRTFLVGNLEIRERRLFNLDVPESRRCFVKVRAYRSERFLPSEQIQGVVISVINL EPRTGFLSNPRAWGRFDSVITGPNGACVPAFCDDQSPDAYSAYVLASLAGξELQAVESS P KFNPNAIGVPQPYLNKLNYRRTDHEDPRVKKTAFQISMAKPRPNSAEESNGPIYAFENLR ACEEAPPSAAHFRFYQIEGDRYDYNTVPFNEDDPMSWTEDYLAWWPKPMEFRACYIKVKI VGPLEVNVRSRNMGGTHRRTVGKLYGIRDVRSTRDRDQPNVSAACLEFKCSGMLYDQDRV DRTLVKVIPQGSCRRASVNPMLHEYLVNHLPLAVNNDTSEYTMLAPLDPLGHNYGIYTVT DQDPRTAKEIALGRCFDGTSDGSSRIMKSNVGVALTFNCVERQVGRQSAFQYLQSTPAQS PAAGTVQGRVPSRRQQRASRGGQRQSGVVASLRFPRVAQQPLIN SEQIDNO:9

AminoacidsequenceofhumanCILP,asolubleactivesecretedformderiv edfromSEQID NO:8. RQTMLTQSVRRVQPGKKNPSIFAKPADTLESPGEWTTLFNIDYPGGKGDYERLDAIRFYY GDRVCARPLRLEARTTDWTPAGSTGQVVHGSPREGFWCLNREQRPGQNCSNYTVRFLCPP GSLRRDTERIWSPWSPWSKCSAACGQTGVQTRTRICLAEMVSLCSEASEEGQHCMGQDCT ACDLTCPMGQVNADCDACMCQDFMLHGAVSLPGGAPASGAAIYLLTKTPKLLTQTDSDGR FRIPGLCPDGKSILKITKVKFAPIVLTMPKTSLKAATIKAEFVRAETPYMVMNPETKARR AGQSVSLCCKATGKPRPDKYFWYHNDTLLDPSLYKHESKLVLRKLQQHQAGEYFCKΆQS D AGAVKSKVAQLIVTASDETPCNPVPESYLIRLPHDCFQNATNSFYYDVGRCPVKTCAGQQ DNGIRCRDAVQNCCGISKTEEREIQCSGYTLPTKVAKECSCQRCTETRSIVRGRVSAADN GEPMRFGHVYMGNSRVSMTGYKGTFTLHVPQDTERLVLTFVDRLQKFVNTTKVLPFNKKG SAVFHEIKMLRRKEPITLEAMETNIIPLGEVVGEDPMAELEIPSRSFYRQNGEPΎIGKV K ASVTFLDPRNISTATAAQTDLNFINDEGDTFPLRTYGMFSVDFRDEVTSEPLNAGKVKVH LDSTQVKMPEHISTVKLWSLNP SEQIDNO: 10

AminoacidsequenceofhumanCILP,asolubleactivesecretedformde rivedfromSEQID NO:8. DTGLWEEEGDFKFENQRRNKREDRTFLVGNLEIRERRLFNLDVPESRRCFVKVRAYRSER FLPSEQIQGVVISVINLEPRTGFLSNPRAWGRFDSVITGPNGACVPAFCDDQSPDAYSAY VLASLAGEELQAVESSPKFNPNAIGVPQPYLNKLNYRRTDHEDPRVKKTAFQISMAKPRP NSAEESNGPIYAFENLRΆCEEAPPSAΆHFRFYQIEGDRYDYNTVPFNEDDPMSWTEDY LA WWPKPMEFRACYIKVKIVGPLEVNVRSRNMGGTHRRTVGKLYGIRDVRSTRDRDQPNVSA ACLEFKCSGMLYDQDRVDRTLVKVIPQGSCRRASVNPMLHEYLVNHLPLAVNNDTSEYTM LAPLDPLGHNYGIYTVTDQDPRTAKEIALGRCFDGTSDGSSRIMKSNVGVALTFNCVERQ VGRQSAFQYLQSTPAQSPAAGTVQGRVPSRRQQRASRGGQ SEQIDNO: 11

gi|27734195|ref|NM_173385.1| Mus musculus cartilage intermediate layer protein, nucleotide pyrophosphohydrolase (CiIp), mRNA i ggaagaagtc ttggaccccc agctaagagt cttgctcagg acacgaaaga agacatcttc 61 tgcacccgac ttagtgatta aagatcattg gctctatgaa atatctctgg ggaactacag 121 gttcaacttt gtgaagcagc gtcacttatc tgtgacactt ctacaaggga gagatggcag 181 caatcaagac ttgggtattc tctttcctgg tccttgaggt caccaccgtg ctggggagac 241 agacgatgct tgcccagtca gtgagaagag tccagcctgt gaagaggacc cccaagaccc 301 ttgccaagcc tgccgactcc caggagagtc ccggagagtg gacaacatgg tttaacatcg 361 accacccagg tgggcagggt gactatgagc ggctggatgc cattcgcttc tactatgggg 421 agcgtgtgtg tgcccgtccc ctgcggctag aggcccggac cactgactgg atgcctgcgg 481 gcagcactgg ccaggtggtc cacggcagtc cccgtgaggg cttctggtgc ctcaacaggg 541 agcagagacc aggccagaac tgttccaatt ataccgtgcg cttcctctgc ccaccagggt 601 ccttgcgagg agatgcagag cacatctgga gttcctggtc tccctggagc aagtgctcag 661 ctgcctgcgg tcacactggg gtgcagaccc gtacccgcac ctgcttggca cagacagtgt 721 cactgtgcag tgaggcgacc gaggagggcc agctctgcat gagccaggcc tgcacagctt 781 gtgacctgac ctgccccatg ggccaggtaa atgctgactg tgatgcctgc atgtgccagg 841 acttcatgct tcacggggcc atctccctcc ctggaggtgg cccagctcca ggagccgctg 901 tctacctgct ggccaaggca ccaaagatgc tgacccgaac agacagcagc gggaggttcc 961 gagttcctgg cttgtgtcct gatggcaaaa ccatcctgaa aatcaccaag accaagtttg 1021 ccccaattat gatcacgatg CGcaagacta gcctgaagtc agctaccatc aatgcggagt 1081 ttgtgagggc agagacccca tacattgtaa tgaaccctga gatgaaagca cgtcgggctg 1141 ggcagagtgt gtctctgtgc tgcaaagcca cggggaaacc cagtccggac aagtacttct 1201 ggtaccacaa caacacactg ctggatccct ccctctataa gcacgagagc aagctggtgc 1261 tgaggaacct gcagcaggac caagcagggg agtacttctg taaggctcag agtgatgctg 1321 gggctgtgaa gtccaaggtc acccagctca ctgtcattgc acatgatgag actccttgca 1381 acccaacccc agagagctac cttatccggc tgccccatga ctgctttcag aacgccagca 1441 attccttcta ctacgacgta ggccgctgcc cgatcaagac ctgtgcaggg cagcaggaca 1501 atgggatccg gtgccgagat gctgtggaga actgctgcgg catttccaga acagaagaga 1561 gggagatcca gtgcagtggg tacacactgc ccaccaaggt ggctgtggag tgcagctgcc 1621 aacgatgcgC agagacacgg agcatcgtcc ggggccgagt cactgctgct gacaacgggg 1681 agcccatgcg ctttggccat gtgtacatgg ggaataaccg cgtgagcatg actggctaca 1741 aaggcacctt cacactccac atcccccagg acacggagag gctggtgctc acatttgtgg 1801 acaggctgca gaagtttgtc aacacaacca aagtgctgcc cttcaacaag aagggaagcg 1861 cagtgttcca cgagatcaag atgcttcggc agaaagagcc catcacgttg gaagccatgg 1921 agaccaacat tatccccctg ggagaggtga ttggtgaaga ccctgtggct gagctggaga 1981 tcccatccaa aagtttctat aggcagaacg gggagccctt cacaggaaag gtaaaggcca 2041 gcgtgacctt cctggatcct cggaacatct cgacggccac agctgcccaa agtgacctca 2101 acttcatcaa tgatgaagga gacacttttc cccttagaac atatggtatg ttttcagtgg 2161 actttagaga tgaggccact tccgagtcac ttaatgctgg caaggtgaag gtccaccttg 2221 actcgaccca ggtcaaaatg ccagagcatg tgccagccat gaaactctgg tcgctcaacc 2281 cagacacagg actgtgggaa gaggaagggg atttcaagtt tgaaagccaa aggaggaaca 2341 agagggaaga gagaaccttc ctagtgggca atatggagat ccgggagaga aggctcttta 2401 acctggacgt ccctgaaagc cgaaggtgct tcattaaggt gaggacttac cggagtgaga 2461 ggttcctgcc cagtgagcaa attcagggag ttgtggtctc agtgatcaat ctggagccca 2521 gaactggctt ctcatctaac cccagggcct ggggccgatt tgacagtgtc atcacagggc 2581 ccaatggggc ctgcctgcct gccttctgtg atgatcagtc ccccgatgct tactctgtct 2641 atgtcttggc aagcctttct ggagaagaac tagaggcagt tgagtcttct cctaaattca 2701 acccaaatgc aatcggtgtc cctcagcctt acctcaacaa acttaaatac cgtcggacag 2761 accatgagga tccacgggtt aaaaagacgg ctttccaaat cagtatggcc aaaccaaggc 2821 ccaactcagc tgaggagagc aatgggccta tctatgcctt tgagaacctt cgggcgtgtg 2881 aggaggcacc ccccagtgca gctcacttcc ggttctatca gatcgagggg gatcgctatg 2941 actataacac agttcctttc aatgaagatg accctatgag ctggactgaa gactacctgg 3001 catggtggcc caagccaatg gagttcaggg cctgctatat caaggttaag attgtggggc 3061 cactggaggt gaatgtacga tcccgtaaca tgggaggcac ccaccggcag actgtgggga 3121 agctctatgg gatccgggat gtgaagagta ctcgggacag agaccaacct aatgtctcat 3181 ctgcctgcct ggagttcaag tgcagtggaa tgctgtatga ccaagaccgt gtagaccgca 3241 cattagtaaa ggttatcccc cagggcagct gccatcgagc tagtgttaac tctatgttgc 3301 acgagtacct ggtcaaccac cttcccctgg cagtcaacaa tgacaccagc gagtatacta 3361 tgctggcacc cctggaccct ctgggccaca attacggtat ctatactgtt actgatcagg 3421 accctcgtac agctaaggag attgcgctcg gccggtgctt tgatggcact tccgatggct 3481 cctccagaat catgaagagc aatgtgggag ttgccctgac ctttaactgt gcagaaaggc 3541 aggtaggccg ccagagtgcc ttccagtacc tccaaagcac accggccagg tccccagcta 3601 caggcactgt ccaaggaaga gtaccagcca tgaggcaaca acgggcaagc aggggtggcc 3661 tacgccggcg tggaagcatg ggcccctctg agattttctg gtgttgctca acaacctctg 3721 agcaactaag tcttgtggtt cttagtcctt tctgtccatc tcatgtgaca gctgtcgtga 3781 gactgatgca cagactgtca cttgttaatt taaacatatc tggtttggtg cagtttgttt 3841 ctttgtgcct ttacttactg tctctgcctc atgatgctga ttgccacagg gcccccacga 3901 tgacaaaaca aagattctcc tcttaaaaag aaacaaaagg aattggtaaa cgtctgcagc 3961 tttggctgaa aacacacatc ttattgtatg gttttgccag cctttacact aatgataatc 4021 taatactgag gctcaagtga acaacaataa aacatttttc ttggccatgt tccacaagat 4081 gtaagcaaag tctctgtcac agtccatata tatataaatg gtggtgaata aggaaataaa 4141 caatgttttt atgtc SEQIDNO: 12

AminoacidsequenceofmouseCILPencodedbytheDNAsequenceshowninSE QIDNO: 11. MAAIKTWVFSFLVLEVTTVLGRQTMLAQSVRRVQPVKRTPKTLAKPADSQESPGEWTTWF NIDHPGGQGDYERLDAIRFYYGERVCARPLRLEARTTDWMPAGSTGQVVHGSPREGFWCL NREQRPGQNCSNYTVRFLCPPGSLRGDAEHIWSSWSPWSKCSAACGHTGVQTRTRTCLΆ Q TVSLCSEATEEGQLCMSQACTACDLTCPMGQVNADCDACMCQDFMLHGAISLPGGGPAPG AAVYLLAKAPKMLTRTDSSGRFRVPGLCPDGKTILKITKTKFAPIMITMPKTSLKSATIN AEFVRAETPYIVMNPEMKARRAGQSVSLCCKATGKPSPDKYFWYHNNTLLDPSLYKHESK LVLRNLQQDQAGEYFCKAQSDAGAVKSKVTQLTVIAHDETPCNPTPESYLIRLPHDCFQN ASNSFYYDVGRCPIKTCAGQQDNGIRCRDAVENCCGISRTEEREIQCSGYTLPTKVAVEC SCQRCAETRSIVRGRVTAADNGEPMRFGHVYMGNNRVSMTGYKGTFTLHIPQDTERLVLT FVDRLQKFVNTTKVLPFNKKGSAVFHEIKMLRQKEPITLEAMETNIIPLGEVIGEDPVAE LEIPSKSFYRQNGEPFTGKVKASVTFLDPRNISTATAAQSDLNFINDEGDTFPLRTYGMF SVDFRDEATSESLNAGKVKVHLDSTQVKMPEHVPAMKLWSLNPDTGLWEEEGDFKFESQR RNKREERTFLVGNMEIRERRLFNLDVPESRRCFIKVRTYRSERFLPSEQIQGVVVSVINL EPRTGFSSNPRAWGRFDSVITGPNGACLPAFCDDQSPDAYSVYVLASLSGEELEAVESSP KFNPNAIGVPQPYLNKLKYRRTDHEDPRVKKTAFQISMAKPRPNSAEESNGPIYAFENLR ACEEAPPSAAHFRFYQIEGDRYDYNTVPFNEDDPMSWTEDYLAWWPKPMEFRACYIKVKI VGPLEVNVRSRNMGGTHRQTVGKLYGIRDVKSTRDRDQPNVSSACLEFKCSGMLYDQDRV DRTLVKVIPQGSCHRASVNSMLHEYLVNHLPLAVNNDTSEYTMLAPLDPLGHNYGIYTVT DQDPRTAKEIALGRCFDGTSDGSSRIMKSNVGVALTFNCAERQVGRQSAFQYLQSTPARS PATGTVQGRVPAMRQQRASRGGLRRRGSMGPSEIFWCCSTTSEQLSLVVLSPFCPSHVTA VVRLMHRLSLVNLNISGLVQFVSLCLYLLSLPHDADCHRAPTMTKQRFSS SEQIDNO: 13 gi|34864686|ref|XM_236348.2|Rattusnorvegicussimilartocartila geintermediatelayer protein,nucleotidepyrophosphohydrolase(LOC315761),mRNA i atggcagcaa ccaagacctg ggtactctct ttcctggtcc ttgaagtcac caccgtgctg 61 gggagacaga tgatgcttac ccagtcagca agaagagtcc agcctgtgaa gaggacccct 121 aagatcctca ccaagcctgt taactcccag gagagtcctg gagagtggac gacttggttt 181 aacatcgacc acccaggtgg gcagggtgac tatgagcgac tggatgccat tcgattctac 241 tacggggagc gcgtgtgcgc ccgtcccctc cggctagagg ctcggaccac tgactggatg 301 cctgcgggca tcactggcca ggtggtccat ggcagtcccc gtgagggctt ctggtgcctc 361 aacaaggaac agagaccagg ccagaactgt tccaattaca cagtgcgctt cctctgccca 421 ccagggtcct tgcggagaga tacagagcat atctggagtt cctggtctcc ctggagcgcg 481 tgctcagccg cctgtggtca caccggggtc cagacccgta cccgcacctg cttggcacag 541 acagtgtcac tatgtaatga ggccactgag gagggacagc tctgcatgag ccaggcctgc 601 acagcttgtg acctgacctg ccccatgggc caggtaaatg ctgactgtga tgcctgcatg 661 tgtcaggact tcatgcttca cggggccatc tccctccctg gaggtggccc agctccaggg 721 gccactgtct acctgctggc taaggcacca aagatgctga cccaaacaga cagcagcgga 781 agtttccgag ttcctggctt gtgtcctgat ggcaaaaccg tcctgaaagt taccaagagc 841 aagtttgccc caattatgat cacgatgccc aagactagcc tgaagtcagc caccatcaac 901 gcagagtttg tgagggcaga gaccccatac attgtaatga accctgagac gaaagcacgt 961 cgggctgggc agagtgtgtc tctgtgctgc aaggccacag ggaaacctag tccagacaag 1021 tacttctgga acctgcagca ggaccaagca ggggagtact tctgtaaggc gcagagtgat 1081 gctggggctg tgaagtccaa ggtcacccaa ctcactgtca ttgcacacga tgaggctcct 1141 tgcaacccaa ccccagagag ctaccttatc cggctgcccc atgattgctt ccagaatgcc 1201 accaactcct tctactatga tgtaggccgc tgcGccatca agacctgtgc agggcagcaa 1261 gacaatggga tccggtgcca agatgccgtg gagaactgct gtggtatttc cagaacagag 1321 gagagggaga tccagtgcag tgggtacaca ctgcccacca aggtggccat ggagtgcagc 1381 tgtcaacgat gtgcagagac acggagcatc gttcggggcc gggtcactgc tgctgacgat 1441 ggggagccca tgcgctttgg ccacgtgtac atggggaata accgtgtgag catgactggc 1501 tacaaaggca cattcaccct ccacgtcccc caggacacgg agaggctggt gctcacgttt 1561 gtggacaggc tgcagaaatt tgtcaacact accaaagtgc tgcccttcaa caagaaaggg 1621 agcgcagtgt tccatgagat caagatgctt cggcagaaag agcccatcac gttggaagcc 1681 acggaaacca acattatccc cctgggagag gtggctggtg aggatcctgt ggctgaactg 1741 gagattccac ccaaaagttt ctataagcag aatggggagc ccttcacagg aaaagtaaag 1801 gccagtgtga ccttcctgga tcctcggaat gtctctacgg ccacagccac ccaaagtgac 1861 ctgaacttca ttaatgatga aggagacacc tttcccctta gaacatacgg gatgttttct 1921 gtggacttca gagatgaggc cacttcggag tcacttaatg ctggcaaggt gaaggtccat 1981 cttgactcga cccaggtcaa aatgccagag catgcgcggg ccatgaaact ctggtcgctc 2041 aacccaaaca cggggctgtg ggaagaggaa ggcgatttca aatttgaaag ccagaggagg 2101 aacaagaggg aagagagaac cttcctagtg ggaaatatgg agatccgtga gagaaggctc 2161 tttaacctgg atgtcccaga aagcagaagg tgcttcatca aggtgaggac ttatcggagt 2221 gagaggttcc tgcccagtga gcaaatccag ggggttgtag tctcagtgat caccctggag 2281 cccagaactg gcttttcatc taaccccagg gcctggggcc gatttgacag tgtcatcaca 2341 gggcccaatg gagcctgcct gcctgccctt ttggtacgac aagtccctta atgctatctc 2401 tgttaatgtt ttgccagccc tttctgaaaa gaattagagc cagtggagtc ttctcctaaa 2461 ttcacccaaa atgcaatcgg tgtccctaag ccttccttca acaaactaaa atacggtcgg 2521 acagaccatg aggatccacg ggttaaaaag acgggtttcc aaatcagtat ggccaaacca 2581 aggcccaact cagctgagga gagcaatggg cccatctatg cctttgagaa ccttcgggca 2641 tgcgaggagg caccacccag tgcagctcac ttccggttct atcagattga gggggatcgc 2701 tatgactata acacagttcc tttcaatgaa gatgacccta tgagctggac tgaagactac 2761 ctggcatggt ggcccaagcc aatggagttc agagcctgct acatcaaggt aaagattgtg 2821 gggccacttg aggtgaatgt acgatcccgt aacatgggcg gcacccaccg gcagactgtg 2881 gggaagctct atgggatccg ggatgtgaaa agtactcggg acagagacca acctaatgtc 2941 tcatctgcct gtctggagtt caagtgcagt ggaatgctgt atgaccaaga ccgtgtagac 3001 cgcacattag tgaaggtcat cccccagggc agctgccatc gagccagtgt taactctatg 3061 ctgcacgagt acctggtcaa ccaccttccc ctggcagtca acaatgacac cagcgagtat 3121 accatgctgg cacccctgga ccctctgggc cacaattacg gtatctatac tgtcactgac 3181 caggaccctg gtacagctaa ggagattgcg cttggccggt gctttgacgg cacttctgat 3241 ggctcctcca gaatcatgaa gagcaacgtg ggagtcgccc tgacctttaa ctgtgcagaa 3301 aggcaggtag gccgccagag cgccttccag tacctccaaa gcaccccggc ccggtcccca 3361 gctacaggca ctgtccaagg aagagtaccc gccatgaggc aacaacgggc aagcaggggt 3421 ggcctacgcc ggcgtggaag catggccccc tctgagattt tctggtgtcg ctcaacaacc 3481 tctgagcaac taagtcttat gattctttgt agtcctctct gtccacctca tgagacagcc 3541 atcgtgagac tgatgcacac attgtcactt gttaatttaa acacatctgg tttagtgcag 3601 ttttcttctt tgtgccttta cttcctgtct ctgtcttgtg atactgattg gcacaaggcc 3661 cccaagatga caaaacagag atctcctctt agaaagaaac aaaaggaatt ggtaaatgcc 3721 cgaggctttg tctggaaaca cttcctattg gatggttttg ccaaccttcg caccaatgat 3781 aatctaatac cgaggtgcaa gtga SEQIDNO:14

AminoacidsequenceofratCILP encodedbytheDNAsequenceshowninSEQIDNO: 13. MAATKTWVLSFLVLEVTTVLGRQMMLTQSARRVQPVKRTPKILTKPVNSQESPGEWTTWF NIDHPGGQGDYERLDAIRFYYGERVCARPLRLEARTTDWMPAGITGQWHGSPREGFWCL NKEQRPGQNCSNYTVRFLCPPGSLRRDTEHIWSSWSPWSACSAACGHTGVQTRTRTCLAQ TVSLCNEATEEGQLCMSQACTACDLTCPMGQVNADCDACMCQDFMLHGAISLPGGGPAPG ATVYLLAKAPKMLTQTDSSGSFRVPGLCPDGKTVLKVTKSKFAPIMITMPKTSLKSATIN AEFVRAETPYIVMNPETKARRAGQSVSLCCKATGKPSPDKYFWNLQQDQAGEYFCKAQSD AGAVKSKVTQLTVIAHDEAPCNPTPESYLIRLPHDCFQNATNSFYYDVGRCPIKTCAGQQ DNGIRCQDAVENCCGISRTEEREIQCSGYTLPTKVAMECSCQRCAETRSIVRGRVTAADD GEPMRFGHVYMGNNRVSMTGYKGTFTLHVPQDTERLVLTFVDRLQKFVNTTKVLPFNKKG SAVFHEIKMLRQKEPITLEATETNIIPLGEVAGEDPVAELEIPPKSFYKQNGEPFTGKVK ASVTFLDPRNVSTATATQSDLNFINDEGDTFPLRTYGMFSVDFRDEATSESLNAGKVKVH LDSTQVKMPEHARAMKLWSLNPNTGLWEEEGDFKFESQRRNKREERTFLVGNMEIRERRL FNLDVPESRRCFIKVRTYRSERFLPSEQIQGVVVSVITLEPRTGFSSNPRAWGRFDSVIT GPNGACLPALLVRQVPXCYLCXCFASPFXKELEPVESSPKFTQNAIGVPKPSFNKLKYGR TDHEDPRVKKTGFQISMAKPRPNSAEESNGPIYAFENLRACEEAPPSAAHFRFYQIEGDR YDYNTVPFNEDDPMSWTEDYLAWWPKPMEFRACYIKVKIVGPLEVNVRSRNMGGTHRQTV GKLYGIRDVKSTRDRDQPNVSSACLEFKCSGMLYDQDRVDRTLVKVIPQGSCHRASVNSM LHEYLVNHLPLAVNNDTSEYTMLAPLDPLGHNYGIYTVTDQDPGTAKEIALGRCFDGTSD GSSRIMKSNVGVALTFNCAERQVGRQSAFQYLQSTPARSPATGTVQGRVPAMRQQRASRG GLRRRGSMAPSEIFWCRSTTSEQLSLMILCSPLCPPHETAIVRLMHTLSLVNLNTSGLVQ FSSLCLYFLSLSCDTDWHKAPKMTKQRSPLRKKQKELVNARGFVWKHFLLDGFANLRTND NLIPRCK SEQIDNO: 15

mbx|FLJ45434HomosapiensMetabolexclone;similartogi:42657506 1 gatcaagagc catattaatt taaacagggg ctgaaagtgg catttgagag agaagatact 61 tcctctcgta gtggacactt gtcttttcag aagatttata cgactgagag tcaaaacttt 121 taccagagac tttcatagtc agtttgaatt ctgcaaaatt tctctttatc ttctggaatg 181 tatgctctag acttggagtg gctcaagctc ttcgatgtga tcctatcaat gtcaagagaa 241 agaagcactg aagagtcata attgtgtcct gatttataaa tcggagacag agggagacga 301 aaaccacact ccagaaagta gcttaatcgg actcactact aagatgttaa aagtgaagag 361 gctggaagaa ttcaacacgt gttataacag caaccagctg gagaaaatgg ccttttttca 421 gtgcagggaa gaggtggaga aagtgaagtg ttttctggaa aaaaattctg gggaccagga 481 ttcaagatct ggacataatg aggcgaagga ggtgtggtca aacgccgacc tgacggaaag 541 gatgcccgtc aaaagcaaaa ggacatcagc cctcgcagtt gacatcccgg ctcctccggc 601 cccatttgat catcgtattg tgacagccaa gcaaggagcg gtcaacagct tctatactgt 661 gagcaagaca gaaatcctag gaggagggcg tttcggccag gttcacaagt gtgaggagac 721 ggccacaggt ctgaagctgg cagccaaaat catcaagacc agaggcatga aggacaagga 781 ggaggtgaag aacgagatca gcgtcatgaa ccagctggac cacgcgaacc tcatccagct 841 gtacgatgcc ttcgagtcta agaacgacat tgtcctggtc atggagtatg tggatggtgg 901 ggagctgttt gaccgcatca tcgatgagag ctacaatttg acggagcttg ataccatcct 961 gttcatgaag cagatatgtg aggggataag gcacatgcat cagatgtaca ttctccactt 1021 ggacctgaag cctgagaata tcctgtgtgt gaatcgggat gctaagcaaa taaaaattat 1081 tgattttgga ttggccagaa gatacaaacc cagagagaag ctgaaggtga actttggaac 1141 cccagaattt ctcgcccctg aagttgtgaa ctatgatttt gtttcatttc ccactgacat 1201 gtggagtgtg ggggtcatcg cctatatgct acttagcggt ttgtcgcctt tcctgggtga 1261 caatgatgct gagacgctga acaacatcct ggcctgcagg tgggacttag aggatgaaga 1321 atttcaggac atctcggagg aggccaagga gttcatctct aagcttctga ttaaggagaa 1381 gagttggcga ataagtgcaa gcgaagctct caagcacccc tggttgtcag accacaagct 1441 ccactccaga ctcaatgccc agaagaagaa gaatcgtggc tctgatgccc aggactttgt 1501 gaccaaatag tctacaggag gcagccattt ggaaggaaaa ctgctgtggt tgctgctgct 1561 tcgagaaaat tttttgaaaa atcagcagtt ctgatgcctt gacccctgtg atgacctggt 1621 agtcttagca gggggagccc tcgaccctga atgtgaactt gaactggagt gcctctgctg 1681 cgctcagagg aacacccagc gctgcggtct ggtctcaggg cgcaaacaca tccctgcacc 1741 cggtggtggt gatgttggga agatgtttcc ctgccatctt gagatttttt acttttttaa 1801 aaaaattttt tgatcttagt cactggggca gggggtgcat tatgtctgcc atttctcaaa 1861 tgactacagc tgtgacactt aagattgatt ggagatttta aaactggagg atgcatttag 1921 agggagaaag ctacttccag ccattagact atgcctttgg aaggggcaat tagatggcca 1981 gaaagaattc tgactcccat gcacccttgg agacttagtt tcacagactc tgtcattttg 2041 atttggtctg tggtcacgtg actacaatgg ataaataatt tgtttgctgg aacagaagcc 2101 gtctttgaat tctccaacaa gaggcatcag ccactgtgtc tttcatttat tctgaattgt 2161 ccccagcgtg cgttttcttc catgtagctg attctgtttt tcacatctat actccctgca 2221 cccttctgac tgacagttca tccccacaaa acacattggt tttgagaggt gatgcaaaaa 2281 gtatctgttt atggttggac tttgaaattc tgacctttga aataatgtca acagtcacaa 2341 caggtttcct caagccccat ttgtcaacta aaagagacgg atctgcacct cctggagcaa 2401 gatagcactg agctttcaac attgaagtga cttgggaaat aacattcaag cttctgggta 2461 aacacataca aacagtggca aagaccaagt acagtctctc gcctgtgctg tctcatggga 2521 ctcaatggtg gtgaagaaat ctcaatcgga caccagaggt gggggcaact ctgcaagctc 2581 acctgctgga cgtgagaatg ttggaacgtt ccagttggaa ggttcctatg cctagcctgt 2641 gtgccatttc atggcagata tgtaagaaag gaaatggccc tactgtagac agcttatggg 2701 gcccagaaag gcactgggct ttaaagagcg aaatgagtgt gtcatttcca catgaaggag 2761 aggcaaggaa aagccccaca aaattcctgt tacccgctga actccaagct caccaaccag 2821 attcatgagc tttctcacat tttccatttc caaataactg ttgtgttgaa tcagctgtgt 2881 taatgccttt attctttgtt tttttttttt agacggagtc tcgctctgtc acgaggctgg 2941 agtgcagtgg ccccatctcg gctcactgca acctccgcct cccaggttca agcgattctc 3001 ctgcagcctc ccgagtagct gggattacag gcgcacgcca ccacgcacgg ctgatttttg 3061 catttttagt agagatgggg tttctccatg ttggccagaa tggtctcaat tactgttgtc 3121 gcaatagcaa tgaaaggcat aagaattact tggtggcctc atgagggatg gcagtttcac 3181 tttgcatcag ctttccccca ttatctctca gatccactta aggaatcctt acagcatgcc 3241 gtgaatgtgt gcaacatcct tattctcatc tttaaggtaa ggaggaagta gagatagaag 3301 gaaatacgtg atttggtgaa ggccacatgc gaattttgtg gtcgtgttag ttataaaact 3361 gactcctttc cagcctttgc tσttacatta tgatgcgttt ctttttctcc tttggccttg 3421 catttatccg aggttggttt cttatgttcc tgtgatgtga ctatgcttaa gtgtttaatt 3481 tagtttgctt aaaacacaca caccacacac acacacacac acaaaaccct gtgactggag 3541 tgttatatta ctatgctatc tttagctgat gatattcatt gagcctggaa aaatacaagc 3601 cagtgcatgc attaatactt agttagaccg cagagagctg gcattcactc aaatgctaaa 3661 caggacacag gggtctgatt tgaagcccct cgtggcaggg cgttttctgt cctgtggtgt 3721 gcattctcta ccaaatcaag caggagatct tttgcacgac gtgcaatttg catccacctt 3781 cccttctgac tcaggttggc agataaactg tccccagacc ctgccacaca tggaagagac 3841 agctcaggcc aaatacaggc atgatttctt tgccaagtta atgcactgat gaccctgcag 3901 tgagtgggtg tgatttttgt tttgttttga ggtttagaaa ggagactgta ctttgaaaat 3961 ggatcttgtt tgattctgta gttaaatttc cacatttgga atgggaaggc atctccatgc 4021 cagcagggtg ccttttatat gagtgctgag ggtagtttgc attgcagagt tcatgattac 4081 agtctagagt tacttgaaat gggagattat cagtctagtg ttcatcccgc ccctgctcca 4141 tgtccttgtg tggctgctcc agcgagagct ggtaactagg tagactgctg ctctaatggg 4201 gaaagggggc tttgcaaagc cgaggaatca agccagatgc gagcatggac tcagaaaagc 4261 tgagtctggc tagaccaggg ctatggagat caatttttcc aacaggtcat taggatttgg 4321 gaggctgtat ttctcctcca ccctcttaat ggatggtatg gctttgctca ctcactaggc 4381 agttcccact ttctttagca aatggaggaa attaagcacc gatgtagcac ttcccactgc 4441 ctggagagaa ataaacctcg caagttatta aggagaaggc atgcctcatt ggtaatttgg 4501 aagacaccaa cacttaccca gaatctgttg ctatttaaaa atcagggaga ggagggcaca 4561 tgtccccatt tgctacatta caaactttgg acgagactct catctgaact aacttgggga 4621 aacaatttct aggttgaagc tggtaacact catgtctgtt gagtgccata agtttgtttt 4681 cagttatttc ctctaacttg atattaattt gctttctttc ctcaaaaatg taagtaggca 4741 tgttgccatt gttgtggctt gctttgatgt atgtttggaa agatgtgtga atacttaatt 4801 tttatccatt ttatggaaat tagcttagca attttttttt ctcttttaag aatcaaaata 4861 tggatttgat actaggaaca gaatctgctc tttatttcat tccttttttg tctcttctgc 4921 ttcctctgcc cctcccccag ccacgcagca cgggtgccag tgtgcatgcg tgtgctgggg 4981 gccaaatgac agaaccccgg ttgaaagtcc gcttgtaaaa gcacaagtga gaggσagcat 5041 caggcccctg gagggctgtg gtcctcggac tcggttcttt cctgtcccct ggccgtcctg 5101 tcatccctgt gtgctgcagc atcaccacgg cctacttccc aatatccaca aagcttcgcc 5161 tttgcttttg cactctcttt tacatttttt tacttctttc ctttttctta agttcggatc 5221 caagtttgtc ttcttatcca tcaccatgct ccctgaaagc ctttttgaga ttaagaaatt 5281 ttcttaccag gaggcaactt tccgtctgtc ctaaagggaa gtgggtgctg gatttattag 5341 atattgggtt cctggattaa gatgacttcc tcacatagct tggagtctgc agtttctcag 5401 ttagaagtgt tctatatttc tttttgctcc attttagtgc gctgtgcttt gtataagtgg 5461 tctttctatg ggcagtttca ccttccatga aggcttataa ctcaaccact actacccatt 5521 cagaagagca gaaagcatct gctgtgtgag cgttcatatg ttacctaatg tgtattttgc 5581 tgtcgtctca gctgatagat gtcagtgagt ctttctgact aggtttattg gtgttgataa 5641 gaaattaaaa aaaggattca taacgtggtg attgtgtcta tcatgtatat tttaaatttc 5701 tgaacgtact ttcatttagc tgtttttctt tgctttctaa agatgtataa caatcatgta 5761 tatgtattat attgaagaat aaacatggac cttatttatc ttttgaaaaa aaaaaaaaaa 5821 aaa SEQIDNO: 16

AminoacidsequenceofhumanFLJ45434encodedbytheDNAsequencesh owninSEQID NO: 15. MLKVKRLEEFNTCYNSNQLEKMAFFQCREEVEKVKCFLEKNSGDQDSRSGHNEAKEVWSN ADLTERMPVKSKRTSALAVDIPAPPAPFDHRIVTAKQGAVNSFYTVSKTEILGGGRFGQV HKCEETATGLKLAAKIIKTRGMKDKEEVKNEISVMNQLDHANLIQLYDAFESKNDIVLVM EYVDGGELFDRIIDESYNLTELDTILFMKQICEGIRHMHQMYILHLDLKPENILCVNRDA KQIKIIDFGLARRYKPREKLKVNFGTPEFLAPEVVNYDFVSFPTDMWSVGVIAYMLLSGL SPFLGDNDAETLNNILACRWDLEDEEFQDISEEAKEFISKLLIKEKSWRISASEALKHPW LSDHKLHSRLNAQKKKNRGSDAQDFVTK SEQIDNO: 17

gi|33359693jreflNM_l 82678.11 Homo sapiens ubiquitin-conjugating enzyme E2E 3 (UBC4/5 homolog, yeast) (UBE2E3), transcript variant 2, mRNA 1 gcaggggcca gcccgccggg tccccgccgc gctctttgtc ctggccgcgc gcgggcggac 61 gcgtgcaccc ccagggtgag gggaggtgtg gagagcgcgc acgggagcgc gcgggaacag 121 ctcgcgcccg ccgggcgtcg gcggcgcgcg cccctccgcc agcggcgcgc gagcgggccg 181 ggcgggcggc cgagttttcc aagagataac ttcaccaaga tgtccagtga taggcaaagg 241 tccgatgatg agagccccag caccagcagt ggcagttcag atgcggacca gcgagaccca 301 gccgctccag agcctgaaga acaagaggaa agaaaacctt ctgccaccca gcagaagaaa 361 aacaccaaac tctctagcaa aaccactgct aagttatcca ctagtgctaa aagaattcag 421 aaggagctag ctgaaataac ccttgatcct cctcctaatt gcagtgctgg gcctaaagga 481 gataacattt atgaatggag atcaactata cttggtccac cgggttctgt atatgaaggt 541 ggtgtgtttt ttctggatat cacattttca tcagattatc catttaagcc accaaaggtt 601 actttccgca ccagaatcta tcactgcaac atcaacagtc agggagtcat ctgtctggac 661 atccttaaag acaactggag tcccgctttg actatttcaa aggttttgct gtctatttgt 721 tcccttttga cagactgcaa ccctgcggat cctctggttg gaagcatagc cactcagtat 781 ttgaccaaca gagcagaaca cgacaggata gccagacagt ggaccaagag atacgcaaca 841 taattcacat aatttgtatg cagtgtgaag gagcagaagg catcttctca ctgtgctgca 901 aatctttata gcctttacaa tacggacttc tgtgtatatg ttatactgat tctactctgc 961 ttttatcctt tggagcctgg gagactcccc aaaaaggtaa atgctatcaa gagtagaact 1021 ttgtagctgt agattagtta tgtttaaaac gcctacttgc aagtcttgct tctttgggat 1081 atcaaaatgt attttgtgat gtactaagga tactggtcct gaagtctacc aaatattata 1141 gtgcatttta gcctaattca ttatctgtat gaagttataa aagtagctgt agatggctag 1201 gaattatgtc atttgtatta aacccagatc tatttctgag tatgtggttc atgctgttgt 1261 gaaaaatgtt ttacctttta cctttgtcag tttgtaatga gaggatttcc ttttaccctt 1321 tgtagctcag agagcacctg atgtatcatc tcaaacacaa taaacatgct cctgaaggaa 1381 aaaaaaaaaa aaaa SEQ ID NO: IS

Amino acid sequence of human UBE2E3 encoded by the DNA sequence shown in SEQ ID NO: 17. MSSDRQRSDDESPSTSSGSSDADQRDPAAPEPEEQEERKPSATQQKKNTKLSSKTTAKLS TSAKRIQKELAEITLDPPPNCSAGPKGDNIYEWRSTILGPPGSVYEGGVFFLDITFSSDY PFKPPKVTFRTRIYHCNINSQGVICLDILKDNWSPALTISKVLLSICSLLTDCNPADPLV GSIATQYLTNRAEHDRIARQWTKRYAT SEQ ID NO: 19

gi|31981935|reflNM_009454.2| Mus musculus ubiquitin-conjugating enzyme E2E 3, UBC4/5 homolog (yeast) (Ube2e3), mRNA 1 cttgctgact gcgcgaccgg gaggagccga gccgagccga gccgagcgca gcggcgggag 61 gctgagcgcg cggggggctc ccgcgtccct gcgccttcgc cttcggccgg cagagcgccc 121 accccaccgt ggctcccgga gcgcggggcg gcgccaccct ccggccgcag cgcggcactg 181 cacggccgcc tgcgactttc gatgttccac actcccgggc cggagcctcg gcggcggctt 241 cttttcccgc ccccccggtg ccccccccag gctgcctcca tttcctcaag gaaggttcct 301 cggcgctctc cctcccccac agcgtagcgg cgcacgagcg ggccgggcgg gcggccgaga 361 agttttccca gagataagtt caccaagatg tccagtgaca ggcaaaggtc ggatgatgag 421 agccctagca ccagcagtgg cagttcagat gcagatcagc gagaccctgc cgctccagag 481 cccgaggaac aggaagaaag aaaaccttct gccacccagc agaagaaaaa caccaaactc 541 tctagcaaaa cgactgctaa gttatccact agtgctaaaa gaattcagaa ggagctagca 601 gagataaccc ttgatcctcc tcctaactgc agtgctgggc ctaaaggaga taacatttat 661 gaatggaggt caactatact tggtccacca ggttctgtat atgaaggtgg tgtttttttt 721 ctggatatca cattttcttc agattatcca tttaagccac caaaggttac tttccgtacc 781 agaatctatc actgcaacat caacagtcag ggagtcatct gcctggatat tctgaaagac 841 aactggagtc ctgctttgac tatttcaaag gttttgctct ctatttgttc ccttttgaca 901 gattgcaacc ctgcggatcc tctggtcgga agcatagcca ctcagtattt gaccaacaga 961 gcagaacatg acaggatagc cagacagtgg accaagagat acgcaacata attcacatca 1021 tttgtatgca gtgtgaagga gcagaaggcc tcttcccact gtgctgcgga tctttatagc 1081 ctttacaata cggacttctg tgtatatgtt atactgattc tactcttgtc tttatccttt 1141 ggagactggg agactccccc aaaaggtaaa agctaccaag agtagaactt tgtagctgta 1201 gattagttat gtttaaaatg cctacttgca agtcttgctt ctttgggata tcaaaatgta 1261 ttttgtgatg tactaaggat actggtcctg aagtctacca aatattatag tgcattttag 1321 cctaattcat tatctgtatg aagttataaa agtagctgta gatgactagg aattatgtca 1381 tttgtattaa gcccagatct atttctgagt atgtggttca tgctgttgtg aaatgtttta 1441 cctttgtcag tttgtaatga gaggatttcc ttctaccctt tgtagctcag agagcactga 1501 tgtatcatct caaacacaat aaacatgctc ctgaaggcaa aaaaaaaaaa aaaaaaaaaa 1561 aa SEQIDNO:20

Amino acid sequence of mouse UBE2E3 encoded by the DNA sequence shown in SEQ ID NO: 19. MSSDRQRSDDESPSTSSGSSDADQRDPAAPEPEEQEERKPSATQQKKNTKLSSKTTAKLS TSAKRIQKELAEITLDPPPNCSAGPKGDNIYEWRSTILGPPGSVYEGGVFFLDITFSSDY PFKPPKVTFRTRIYHCNINSQGVICLDILKDNWSPALTISKVLLSICSLLTDCNPADPLV GSIATQYLTNRAEHDRIARQWTKRYAT SEQIDNO:21

ENSRNOT00000006429cDNAsequence,EnsEMBLtranscript[Rattusnorve gicus] 1 atgtccagtg ataggcaaag gtcggatgat gagagcccta gcaccagcag tggtagttca 61 gatgcagacc agcgagaccc tgccgctcca gagcctgagg aacaggaaga aagaaaacct 121 tctgccaccc agcagaagaa aaacaccaaa ctctctagca aaacgactgc taagttatcc 181 actagtgcta aaagaattca gaaggagcta gctgagatca cccttgatcc tcctcctaac 241 tgcagtgctg ggcctaaagg agataacatt tatgaatgga ggtcaactat acttggtcca 301 ccaggttctg tgtatgaagg tggtgttttt tttctggata tcacattttc ttcagattat 361 ccatttaagc caccaaaggt taccttccgc accagaatct accactgcaa catcaacagt 421 cagggagtca tctgcctgga tattctgaaa gacaactgga gtcccgcttt gactatttca 481 aaggttttgc tctctatttg ttcccttttg acagattgca accctgcgga ccctctggtt 541 ggaagcatag ccactcagta tttgaccaac agagcagaac acgacaggat agccagacag 601 tggaccaaga gatacgcaac ataa SEQIDNO:22 AminoacidsequenceofratUBE2E3 encodedbytheDNAsequenceshowninSEQIDNO: 21. MSSDRQRSDDESPSTSSGSSDADQRDPAΆPEPEEQEERKPSATQQKKNTKLSSKTTAKL S TSAKRIQKELAEITLDPPPNCSAGPKGDNIYEWRSTILGPPGSVYEGGVFFLDITFSSDY PFKPPKVTFRTRIYHCNINSQGVICLDILKDNWSPALTISKVLLSICSLLTDCNPADPLV GSIATQYLTNRAEHDRIARQWTKRYAT SEQIDNO:23

gi|31559774|ref|NM_032557.4| Homo sapiens ubiquitin specific protease 38 (USP38), mRNA i cttcggtctc ttctccccgg ctgatcccag cactctccgt gacagcgcct cctgactcag 61 cccaggaccg gcttcttctc acgacctgct ggagactgga cgcccacacc tgacccggaa 121 ctcggaggcg tgcttcctcc acccgccggc tagcagcccc gggccctgag ctcccgccga 181 cgccgctggg gggcccgaca ggcccctcgg cgctgatgct gagtgggatc gagggcccgg 241 ggcggcggcg gagtacgggc ctctggcgcc ttaggccagc cgcaggtgtc ggttcttagg 301 ctctccaggc tcgctagctc ccgccccggc ttggatgggt ctccctgcgc cataaatgtg 361 gctgctgagg cggcggtggc cgtggcccgt cgcgctgctg ctgcggcgct ccaagttcat 421 ctccgccccg gggctctcct gccccacctc ggggctgccg ccacccgctc cttatcccct 481 ggccctggcc ttgcagcgtg gcgacaatgg acaagatcct ggagggcctt gtgagttcct 541 cgcatcccct gcccctcaag cgggtgattg tgcggaaggt ggtggaatcg gcggagcact 601 ggctagacga ggcgcagtgc gaggccatgt ttgacctgac gacccggctc atcctggagg 661 gccaggaccc tttccagcgg caggtggggc accaggtgct ggaggcctac gcacgatacc 721 accggccaga gttcgagtcc ttcttcaaca agaccttcgt gttgggcctc cttcatcagg 781 gctaccactc tctggacagg aaggatgtag ccatcctgga ctacattcac aacggcctga 841 agctgattat gagctgtccg tcggtgctgg atctctttag cctcctgcag gtagaggtgt 901 tacggatggt gtgtgagagg ccggagccgc agctctgtgc ccgactgagc gaccttctga 961 ccgactttgt gcaatgcatc cccaagggga aattgtccat cacgttctgt caacagctgg 1021 ttcgaacgat aggccatttc cagtgcgtgt ccacccagga aagagagctg cgggaatatg 1081 tctcccaggt gacaaaagtg agtaacttgc tgcagaacat ctggaaggcc gagcctgcca 1141 cactactgcc ttccctgcaa gaagtttttg caagcatctc ttccacagat gcatcatttg 1201 aaccttctgt agcattggca agccttgtgc agcatattcc tcttcagatg attacagttc 1261 tcatcaggag ccttactacg gatccaaatg taaaagatgc aagtatgacc caagcccttt 1321 gcagaatgat tgactggcta tcctggccat tggctcagca tgtggataca tgggtaattg 1381 cactcctgaa aggactggca gctgtccaga agtttactat tttgatagat gttactttgc 1441 tgaaaataga actggttttt aatcgacttt ggtttcctct tgtgagacct ggtgctcttg 1501 cagttctttc tcacatgctg cttagctttc agcattctcc agaggcgttc catttgattg 1561 ttcctcatgt ggttaatttg gttcattctt tcaaaaatga tggtctgcct tcaagtacag 1621 ccttcttagt acaattaaca gaattgatac actgtatgat gtatcattat tctggatttc 1681 cagatctcta tgaacctatt ctggaggcaa taaaggattt tcctaagccc agtgaagaga 1741 agattaagtt aattctcaat caaagtgcct ggacttctca atccaattct ttggcgtctt 1801 gcttgtctag actttctgga aaatctgaaa ctgggaaaac tggtcttatt aacctaggaa 1861 atacatgtta tatgaacagt gttatacaag ccttgtttat ggccacagat ttcaggagac 1921 aagtattatc tttaaatcta aatgggtgca attcattaat gaaaaaatta cagcatcttt 1981 ttgcctttct ggcccataca cagagggaag catacgcacc tcggatattc tttgaggctt 2041 ccagacctcc atggtttact cccagatcac agcaagactg ttctgaatac ctcagatttc 2101 tccttgacag gctccatgaa gaagaaaaga tcttgaaagt tcaggcctca cacaagcctt 2161 ctgaaattct ggaatgcagt gaaacttctt tacaggaagt agctagtaaa gcagcagtac 2221 taacagagac ccctcgtaca agtgacggtg agaagacttt aatagaaaaa atgtttggag 2281 gaaaactacg aactcacata cgttgtttga actgcaggag tacctcacaa aaagtggaag 2341 cctttacaga tctttcgctt gccttttgtc cttcctcttc tttggaaaac atgtctgtcc 2401 aagatccagc atcatcaccc agtatacaag atggtggtct aatgcaagcc tctgtacccg 2461 gtccttcaga agaaccagta gtttataatc caacaacagc tgccttcatc tgtgactcac 2521 ttgtgaatga aaaaaccata ggcagtcctc ctaatgagtt ttactgttct gaaaacactt 2581 ctgtccctaa cgaatctaac aagattcttg ttaataaaga tgtacctcag aaaccaggag 2641 gtgaaaccac accttcagta actgacttac taaattattt tttggctcca gagattctta 2701 ctggtgataa ccaatattat tgtgaaaact gtgcctctct gcaaaatgct gagaaaacta 2761 tgcaaatcac ggaggaacct gaatacctta ttcttactct cctgagattt tcatatgatc 2821 agaagtatca tgtgagaagg aaaattttag acaatgtatc actgccactg gttttggagt 2881 tgccagttaa aagaattact tctttctctt cattgtcaga aagttggtct gtagatgttg 2941 acttcactga tcttagtgag aaccttgcta aaaaattaaa gccttcaggg actgatgaag 3001 cttcctgcac aaaattggtg ccctatctat taagttccgt tgtggttcac tctggtatat 3061 cctctgaaag tgggcattac tattcttatg ccagaaatat cacaagtaca gactcttcat 3121 atcagatgta ccaccagtct gaggctctgg cattagcatc ctcccagagt catttactag 3181 ggagagatag tcccagtgca gtttttgaac aggatttgga aaataaggaa atgtcaaaag 3241 aatggttttt atttaatgac agtagagtga catttacttc atttcagtca gtccagaaaa 3301 ttacgagcag gtttccaaag gacacagctt atgtgctttt gtataaaaaa cagcatagta 3361 ctaatggttt aagtggtaat aacccaacca gtggactctg gataaatgga gacccacctc 3421 tacagaaaga acttatggat gctataacaa aagacaataa actatattta caggaacaag 3481 agttgaatgc tcgagcccgg gccctccaag ctgcatctgc ttcatgttca tttcggccca 3541 atggatttga tgacaacgac ccaccaggaa gctgtggacc aactggtgga gggggtggag 3601 gaggatttaa tacagttggc agactcgtat tttgatcctg agagagtcca aaatgcactg 3661 gtcacgaaac gtctaatact atgactgtta aaatgtcaga ctataacaaa tatctatctt 3721 ttatttttca ttagaccctt atacttcaag agaacacact cagtgcttgt ttttattttc 3781 ttgacacatt tattaacaaa atgcatcatg gaaaaaaaat ctacctctta aaattccatt 3841 tgcttttatg gttagacatg cttgaccaaa aatgttcaga agaaaatatg tacctggtcc 3901 ctaattaagc tgcgttaaat ttggtagaag catttaaatg gtctatcttc agttttactg 3961 aacaaaaaat gtaatttatt tagcattctt tataaaagaa ttgatgctag aggtaaaaaa 4021 aaatacttgt ttttaaaaaa tcctttacgt cttgtgtaat taccccatta ttaaattcaa 4081 gtccttgaaa atcaactaga gattataaag tctctaaaga aggcaataac aaaatttatc 4141 aagatatagt acttttcagt ttttgtttag tgtcttcagc atcactgtgt ctgtatttca 4201 agtacaaatg tttttaaaaa ggattcttta tacatatgtg ctgaattgat tttaagaaag 4261 ttgcatgatc ctgtaggagc aacattttta cctaaaaaat gctaacttta tagtatttct 4321 aattgttcaa ggattttaaa attctatttc agggagtata tcttctgtgg ttttgaagga 4381 ggtgagttct gtatgtgcct tgcagtactg taattcaaaa ataggaatct ttggctgcaa 4441 aattttaatg aaatgttagg aagtaatttt cgtgctaaca ttaaaattat aactttttga 4501 aaggtaatag attttccaga agtaaaatct gatggttcta aatcaatcaa tgtgatagtt 4561 catttttaac tcttagaaga attcagagga aattaaccca gctaagtaaa aaatctgtct 4621 tgattttgtt acttattcct cagaatatta aacattgatc acataaaaaa aaaaaaaaaa 4681 aaaa SEQIDNO:24

AminoacidsequenceofhumanUSP38encodedbytheDNAsequenceshown inSEQID NO:23. MDKILEGLVSSSHPLPLKRVIVRKVVESAEHWLDEAQCEAMFDLTTRLILEGQDPFQRQV GHQVLEAYARYHRPEFESFFNKTFVLGLLHQGYHSLDRKDVAILDYIHNGLKLIMSCPSV LDLFSLLQVEVLRMVCERPEPQLCARLSDLLTDFVQCIPKGKLSITFCQQLVRTIGHFQC VSTQERELREYVSQVTKVSNLLQNIWKAEPATLLPSLQEVFASISSTDASFEPSVALASL VQHIPLQMITVLIRSLTTDPNVKDASMTQALCRMIDWLSWPLAQHVDTWVIALLKGLAAV QKFTILIDVTLLKIELVFNRLWFPLVRPGALAVLSHMLLSFQHSPEAFHLIVPHVVNLVH SFKNDGLPSSTAFLVQLTELIHCMMYHYSGFPDLYEPILEAIKDFPKPSEEKIKLILNQS AWTSQSNSLASCLSRLSGKSETGKTGLINLGNTCYMNSVIQALFMATDFRRQVLSLNLNG CNSLMKKLQHLFAFLAHTQREAYAPRIFFEASRPPWFTPRSQQDCSEYLRFLLDRLHEEE KILKVQASHKPSEILECSETSLQEVASKAAVLTETPRTSDGEKTLIEKMFGGKLRTHIRC LNCRSTSQKVEAFTDLSLAFCPSSSLENMSVQDPASSPSIQDGGLMQASVPGPSEEPVVY NPTTAAFICDSLVNEKTIGSPPNEFYCSENTSVPNESNKILVNKDVPQKPGGETTPSVTD LLNYFLAPEILTGDNQYYCENCASLQNAEKTMQITEEPEYLILTLLRFSYDQKYHVRRKI LDNVSLPLVLELPVKRITSFSSLSESWSVDVDFTDLSENLAKKLKPSGTDEASCTKLVPY LLSSVVVHSGISSESGHYYSYARNITSTDSSYQMYHQSEALΆLASSQSHLLGRDSPSAV F EQDLENKEMSKEWFLFNDSRVTFTSFQSVQKITSRFPKDTAYVLLYKKQHSTNGLSGNNP TSGLWINGDPPLQKELMDAITKDNKLYLQEQELNARARALQAASASCSFRPNGFDDNDPP GSCGPTGGGGGGGFNTVGRLVF SEQ ID NO: 25

gi|34328300|reflNM_027554.2| Mus musculus ubiquitin specific protease 38 (Usρ38), mRNA 1 gggtgttccg cgcctccggc tcctctcctt ggcagatcga ggtactcgca ttgacagtgc 61 ctcctgactc aacccagggc gacttttaac tgcccacccg cgggagccgg ggctccgcac 121 tgacccggga cacgaaggcg ctcctcacct tccagctcag cctggagggt ccctgcgccc 181 ggaggctggg agaggtcgcg ggcccagcga ggcagcctta tgcggacctc cggcccctga 241 ggcggaccgc ggggacccgc agcctttcgg cgtcccagaa tcgctcgctc ccgtcccggc 301 tcggatgggc cttcccactc gataaatgtg gctgctgagg cggcggtggc ggtggccggt 361 cctgctgctg gttggctcca agttcctccc cgccggggcg ctttgctgcc ccccggggct 421 gccgctgcca gctctttatc cccagggcct ggccttgcag agtggcgacg atggacaaga 481 ttctggaagg cctggtgagc tcctcgcacc cgctgcccct caagcggatg attgtgcgga 541 aggtggtgga gttcgcggag cattggctcg acgaggcgca gtgcgaggcc atgtttgacc 601 tgacgacccg cctcatcctg gagggccagg accctttcca gcgacaagtg ggccaccagg 661 tgctggaggc ctacgcgcgg taccaccggc cggagttcga gtccttcttc aacaagactt 721 tcgtgttggg cctccttcag cagggctacc actcagtgga caggaaggac gtcgccatcc 781 tggactacat tcacaacggc ctgaagctga ttatgagctg cccgtcggtg ctggatctct 841 tcagcctgct ccaggtggag gtcctgcgaa tggtttgcga gaggccggag ccagtgctct 901 gtgcccggct gagcgacctc ctgaccgatt tcgtgcagtg cgtccccaaa gggaaactgt 961 cagttacctt ctgtcaacag ctggttcgaa cgataggtca tttccagtgc gtgtccactc 1021 aggagaagga actacgggag tatgtctccc aggtaacgaa agtgagtacc ttactacaga 1081 atatttggaa ggctgagccc tccacgctgc taccttctct gcaagaagtt tttgcaagca 1141 tctcttccac agatgcatcc tttgagccct cggttgcctt ggcaagcctg gtgcagcata 1201 ttcctctgca gatgatcaca gtcctcatca ggagcctaac caccgaccca aatgtaaaag 1261 atgcgagtat gacccaagct ctctgcagaa tgattgactg gctgtcctgg ccattggctc 1321 agcatgtaga tacgtgggta attgctctcc tgaaaggact ggcagctgtt cagaagttta 1381 ctattttgat agatgttact ttgctgaaaa tagaactggt ttttaaccga ctttggtttc 1441 ctctcgtgag acctggtgct cttgcagtcc tctctcatat gctgcttagt tttcagcatt 1501 ctcctgaggc attccatgtg attgtccctc atatagttaa cttggttcac tccttcagaa 1561 gtgatggcct accttccagc acagccttcc tagtacagtt aacagagttg gttcactgta 1621 tgatgtacca ttactccgga ttcccagatc tctatgagcc tattctggag gcagtaaagg 1681 actttcctaa gcccagtgaa gaaaagatta agctgattct caatcaaagc gcctggactt 1741 ctcaatccaa tgctttggca tcttgcttgt ctagactttc tggaaaatct gaaactggaa 1801 aaactggtct tattaacctg ggaaacacgt gttacatgaa cagtgtttta caagcattgt 1861 ttatggctac agagttcaga agacaagtat tatctttaaa tctaaatggg tgtaattcgt 1921 taatgaaaaa attacaacat ctttttgctt ttttggctca tacacaaagg gaagcttacg 1981 cacctcggat attttttgag gcttccagac ctccgtggtt tactcccagg tcacaacagg 2041 actgctctga gtacctcagg tttctgctgg acagactcca tgaagaagaa aagatcttgc 2101 gagttcagtc ctcacataag ccttcggaag gtctggactg tgctgaaact tgtttacaag 2161 aggtgaccag taaggtagca gtgcccacag agtctcctgg tacaggtgac agtgagaaga 2221 ctctaataga gaaaatgttt ggagggaagc tacggactca catctgctgc ctaaactgtg 2281 ggagtacctc acacaaagtt gaagccttca cagatctttc gctagccttt tgtcctagcc 2341 cttctgtgga agacctgtct ttccaagata cagcttcgtt acccagtgct caagatgatg 2401 ggctaatgca aaccagtgtc gctgacccag aagaagagcc agtagtttat aatccagcaa 2461 cagctgcctt tgtctgcgac tcagttgtga atcaaagagt attaggcagt cctcctgttg 2521 agtttcactg tgctgaaagc tcttctgtcc ctgaagaatc tgcaaagatt ctcatcagta 2581 aagatgtacc tcagaaccca ggaggtgaaa gcacaacttc agtgactgac ttactaaact 2641 actttctggc tccagaggtt cttactgggg agaaccaata ctactgtgaa agctgtgcct 2701 ctctacagaa cgcagagaaa actatgcaaa tcacggagga gcctgagtat cttatcctta 2761 cgctcctgag attctcctac gatcagaagt accatgtgag aagaaaaatt ttagacaatg 2821 tgtcactgcc tctggttttg gagttgccag ttaaaagaac tgcttccttc tcttccttgt 2881 cccagagttg gtctgtagat gttgacttca ctgatattaa tgagaaccta cctaaaaaat 2941 taaaaccttc tgggactgaa gaagctttct gccccaaatt agtgccctac ctattaagtt 3001 ctgttgttgt gcactctggt gtgtcctctg agagtggcca ttactactct tatgccagaa 3061 acatcacagg tacagagtct tcataccaga tgtgccccca gtctgaaagt ctggcattag 3121 caccctccca gagttgttta ctaggggtag aaagcGccaa tacagtcatt gaacaggact 3181 tggaaaataa ggaaatgtca caagagtggt ttttatttaa tgacagcagg gtgacattta 3241 cttcatttca gtcagttcag aaaatcacaa gtaggtttcc aaaagacaca gcttatgtgc 3301 ttttgtacaa aaaacaaagc agagctaatg gcatagacag tgacaatcca gccagtggtg 3361 tctgggccaa tggagatcca cccctacaga aagagctgat ggatgccata acaaaggaca 3421 acaagctgta tttgcaggaa caagaattga atgctcgagc ccgggccctg caagctgcat 3481 ctgcttcctg ttcgttccga cccaatggat ttgatgacaa tgacccacca ggaagctgtg 3541 ggccaactgg tgggggcggt ggaggaggct tcaatacagt tggcagactc gtattttgac 3601 cctgagagaa ctgaaaatgt atctggtcac aaaacatcca atactgtggc tgttaaaatg 3661 tcaggctgta acctatagca tatctatctt ttatttttct tttcattaga cccgaatact 3721 ttaagaaaat atactcagtg gttgttttta ttttcttgac aaaaattcat catggaaaaa 3781 aaaaatctac ctcttaaact catttgcagt tacagttaga catgtttggc cacaagtatt 3841 tagaggaaaa ggcatacctg ctccctaatt aagctgcatt aaatttggta gaaggattta 3901 aatggtgtat tttcagttgt actgaacaaa ataattcact tatcattctt tgtacaaaaa 3961 ttgatgctag atatatctta agaaaaatga aattcttatt tctggtataa ttaccccaat 4021 attaaattca agtctttgaa aatagtagag atggtgagtc tctggaggcg atagcacctt 4081 ccaccccagc tgccagtctc tgggctgctg tctccgctgt cctcggcgtt gcactccaga 4141 ttcaggaaca agtgtcattt acacacgtgg caagtgaatt cacacaaggt gcatgacgcc 4201 agagggcagg ggttttaact gaactttgta ggatttccag ttgtccaagg actttaaaat 4261 cctgtttcag ggagtgtatc tcctatgtgt tttaaaggag gtgagtctct gaatgtgcct 4321 tgcagtactg taattcaaaa ataggttctc tttggctgcg gcggaaagtt ttctaaaaca 4381 aagcgttagg aaggaatttt gtgctaacat taaaattata attttttaag gtattagata 4441 tttcagaagt aagatctggt ggttctaaac tagtaagtgt aatggtaaat ttctaactct 4501 tagaaaaatt cggaggaaat gaagctagtt taagtaaaaa atctttaatt gatttgttac 4561 ttaatcttca cattaaacat tgataacatt tttagagttg tatatttgga tttttttcct 4621 ttgagacaca tgccttacac ataaaatata atctgcttca tgaaatatta aaatgccaca 4681 cctcaaaaaa aaaaaaaaaa aaaaaaaaaa a SEQ ID NO: 26

Amino acid sequence of mouse USP38 encoded by the DNA sequence shown in SEQ ID NO: 25. MDKILEGLVSSSHPLPLKRMIVRKVVEFAEHWLDEAQCEAMFDLTTRLILEGQDPFQRQV GHQVLEAYARYHRPEFESFFNKTFVLGLLQQGYHSVDRKDVAILDYIHNGLKLIMSCPSV LDLFSLLQVEVLRMVCERPEPVLCARLSDLLTDFVQCVPKGKLSVTFCQQLVRTIGHFQC VSTQEKELREYVSQVTKVSTLLQNIWKAEPSTLLPSLQEVFASISSTDASFEPSVALASL VQHIPLQMITVLIRSLTTDPNVKDASMTQALCRMIDWLSWPLAQHVDTWVIALLKGLAAV QKFTILIDVTLLKIELVFNRLWFPLVRPGALAVLSHMLLSFQHSPEAFHVIVPHIVNLVH SFRSDGLPSSTAFLVQLTELVHCMMYHYSGFPDLYEPILEAVKDFPKPSEEKIKLILNQS AWTSQSNALASCLSRLSGKSETGKTGLINLGNTCYMNSVLQALFMATEFRRQVLSLNLNG CNSLMKKLQHLFAFLAHTQREAYAPRIFFEASRPPWFTPRSQQDCSEYLRFLLDRLHEEE KILRVQSSHKPSEGLDCAETCLQEVTSKVAVPTESPGTGDSEKTLIEKMFGGKLRTHICC LNCGSTSHKVEAFTDLSLAFCPSPSVEDLSFQDTASLPSAQDDGLMQTSVADPEEEPVVY NPATAAFVCDSVVNQRVLGSPPVEFHCAESSSVPEESAKILISKDVPQNPGGESTTSVTD LLNYFLAPEVLTGENQYYCESCASLQNAEKTMQITEEPEYLILTLLRFSYDQKYHVRRKI LDNVSLPLVLELPVKRTASFSSLSQSWSVDVDFTDINENLPKKLKPSGTEEAFCPKLVPY LLSSVVVHSGVSSESGHYYSYARNITGTESSYQMCPQSESLALAPSQSCLLGVESPNTVI EQDLENKEMSQEWFLFNDSRVTFTSFQSVQKITSRFPKDTAYVLLYKKQSRANGIDSDNP ASGVWANGDPPLQKELMDAITKDNKLYLQEQELNARARALQAASASCSFRPNGFDDNDPP GSCGPTGGGGGGGFNTVGRLVF SEQ ID NO: 27

ENSRNOT00000035084 Coding sequence, EnsEMBL transcript [Rattus norvegicus] 1 atggacaaga ttctggaggg tctggtgagt tcctcgcacc cgctgcccct caagcggatg 61 attgtgcgga aggtggtgga atccgcggag cattggctcg acgaggcgca gtgcgaggcc 121 atgtttgacc tgacgacccg cctcatcctg gagggccagg accccttcca gcgaσaagtg 181 ggccaccagg tgctggaggc ctacgcgcgg taccaccggc cggagttcga gtccttcttc 241 aacaagactt tcgtgttggg cctccttcag cagggctacc actcactgga caggaaggac 301 gtagccatcc tggactacat tcacaacggc ctgaagctga ttatgagctg cccgtcggtg 361 ctggacctct tcagcctgtt acaggtggag gtcttacgaa tggtttgcga gaggccggag 421 ccactactct gtgccaggct gagcgacctc ctgacggatt tcgtgcagtg cgtccccaaa 481 gggaaactgt ccattacctt ctgtcaacag ctggttcgga cgataggtca cttccagtgt 541 gtgtccaccc aggagaagga gctccgggag tatgtctccc aggtaacaaa agtgagtaac 601 ttactacaga atatttggaa ggctgagccc tccacgctgc taccttctct gcaagaagtt 661 tttgcaagca tctcttccac agatgcgtcc tttgagccct cggtggcctt ggcaagcctg 721 gtgcagcata ttcctctgca gatgatcacg gtcctcatca ggagcctgac caccgaccca 781 aacgtgaagg atgcaagcat gacccgagct ctctgcagaa tgattgactg gctgtcctgg 841 ccattggctc agcatgtgga tacatgggta attgctctcc tgaagggact ggcagctgtt 901 cagaagttta ctattttgat agatgttact ttgctgaaaa tagaactggt ttttaatcga 961 ctttggtttc ctcttgtgag acctggtgct cttgcagtcc tctctcatat gctgcttagt 1021 tttcagcatt ctcσagaggc attccatgtg attgtccctc atatagttaa cttggttcac 1081 tccttcagaa gtgatggcct gccttccagc acagccttct tagtgcagtt aacagagctg 1141 gttcactgta tgatgtacca ttattctggg tttccagatc tctatgagcc tattctggag 1201 gcggtgaagg attttcctaa gcccactgaa gagaagatta agttgattct caatcaaagt 1261 gcctggactt ctcaatccaa cgctttggca tcttgcttgt ctagactttc tggaaaatct 1321 gaaactggga aaactggtct tattaacctg ggaaacacgt gttacatgaa cagtgtttta 1381 caagcattgt' ttatggctac agagttcaga agacaagtat tatctttaaa tctaaatggg 1441 tgtaattcct taatgaaaaa attacaacat ctttttgctt ttttggctca tacacaaagg 1501 gaagcatacg cacctcggat attttttgag gcttccagac ctccgtggtt tactcccagg 1561 tcacaacagg attgctctga gtacctcagg tttctgctgg acagactcca tgaagaagaa 1621 aagatcttac gaattcagtc ctcacataag ccttctgaag gtctgggctg tactgaaact 1681 tgtttacaag aagtaaccaa taaggtagct gtgccctcag agtcccctag cacaggtggc 1741 agtgagaaga ctctaataga gaaaatgttt ggagggaaac tacggactca catctgctgc 1801 ctgaactgca ggagtacctc acacaaagtg gaagccttca cagatctttc gctagccttc 1861 tgtcctagcc cttctgtgga agactcgtct ttccaagacc cagcttcgct acccagtgca 1921 caagatgatg ggctaatgca aaccagtgtc actgacgcag aagaagagcc agtagtttgt 1981 aacccagcag cagctgcctt tgtttgtgac tcagttgtga atgaaagacg attgggcagt 2041 cctcctgttg agtttccctg tgccaaaaac tcttctgtcc ctggggaatc tgcaaagatt 2101 cttgtcagta aagatgtacc tcagaaccca ggaggtgaga gcacaacttc ggtgactgac 2161 ttactgaact actttttggc tcccgaggtt ctgactggtg agaaccaata ttactgtgag 2221 agctgtgcct ctctacagaa tgcagagaag actatgcaga tcacagagga gcctgagtac 2281 cttatcctta cgctcctgag attctcctat gatcagaagt accacgtgag aagaaagatt 2341 ttagacaatg tgtcactgcc gctggttttg gagttgcccg ttaaaaggac ttccctctct 2401 tccttgtcgg aaggttggtc tgcagatgct gacttcactg atgttaatga gaacctagct 2461 aaaaaattaa aaccttctgg gactgaagaa gctttctgcc ccaaattagt gccctaccta 2521 ttaagttctg ttgttgtgca ctctggtgtg tcctctgaaa gtggccatta ctattcttat 2581 gccagaaaca tcacaggtac agagtcttcc taccagatgc gcccccagtc tgaaagtctg 2641 tcattaacac cctcccagag tagtttatta gggggagaaa gccccaatac agtcattgaa 2701 caggacttgg aaaataagga aatgtcacaa gagtggtttt tatttaatga cagcagggtg 2761 acatttactt catttcagtc agtccagaaa atcacaagta ggtttccaaa agacacagcg 2821 tacgtgcttt tctacaaaaa gcaaagcaga actaatggtg tacgcagtga caatccagcc 2881 agtggcgtct gggtcaatgg agagccacca ctacagaaag agctcatgga tgccataaca 2941 aaggacaaca aactgtattt gcaggaacaa gaattgaatg ctcgagcccg ggccctacaa 3001 gctgcgtcag cgtcatgctc ctttcggccc aatggatttg acgacaatga cccaccagga 3061 agctgtgggc cgacgggtgg agggggcgga ggaggattta acgcagttgg cagactcgtc 3121 ttttga SEQIDNO:28

AminoacidsequenceofratUSP38encodedbytheDNAsequenceshownin SEQIDNO:27. MDKILEGLVSSSHPLPLKRMIVRKWESAEHWLDEAQCEAMFDLTTRLILEGQDPFQRQV GHQVLEAYARYHRPEFESFFNKTFVLGLLQQGYHSLDRKDVAILDYIHNGLKLIMSCPSV LDLFSLLQVEVLRMVCERPEPLLCARLSDLLTDFVQCVPKGKLSITFCQQLVRTIGHFQC VSTQEKELREYVSQVTKVSNLLQNIWKAEPSTLLPSLQEVFASISSTDASFEPSVALASL VQHIPLQMITVLIRSLTTDPNVKDASMTRALCRMIDWLSWPLAQHVDTWVIALLKGLAAV QKFTILIDVTLLKIELVFNRLWFPLVRPGALAVLSHMLLSFQHSPEAFHVIVPHIVNLVH SFRSDGLPSSTAFLVQLTELVHCMMYHYSGFPDLYEPILEAVKDFPKPTEEKIKLILNQS AWTSQSNALASCLSRLSGKSETGKTGLINLGNTCYMNSVLQALFMATEFRRQVLSLNLNG CNSLMKKLQHLFAFLAHTQREAYAPRIFFξASRPPWFTPRSQQDCSEYLRFLLDRLHEE E KILRIQSSHKPSEGLGCTETCLQEVTNKVΆVPSESPSTGGSEKTLIEKMFGGKLRTHIC C LNCRSTSHKVEAFTDLSLAFCPSPSVEDSSFQDPASLPSAQDDGLMQTSVTDAEEEPVVC NPAAAAFVCDSVVNERRLGSPPVEFPCAKNSSVPGESAKILVSKDVPQNPGGESTTSVTD LLNYFLAPEVLTGENQYYCESCASLQNAEKTMQITξξPEYLILTLLRFSYDQKYHVRR KI LDNVSLPLVLELPVKRTSLSSLSEGWSADADFTDVNENLAKKLKPSGTEEAFCPKLVPYL LSSVVVHSGVSSESGHYYSYARNITGTESSYQMRPQSESLSLTPSQSSLLGGESPNTVIE QDLENKEMSQEWFLFNDSRVTFTSFQSVQKITSRFPKDTAYVLFYKKQSRTNGVRSDNPA SGVWVNGEPPLQKELMDAITKDNKLYLQEQELNARARALQAASASCSFRPNGFDDNDPPG SCGPTGGGGGGGFNAVGRLVF